Associate editor: L. Schechter
Ketamine and the next generation of antidepressants with a rapid onset of action

https://doi.org/10.1016/j.pharmthera.2009.02.010Get rights and content

Abstract

Existing treatments for major depressive disorder (MDD) usually take weeks to months to achieve their antidepressant effects, and a significant number of patients do not have adequate improvement even after months of treatment. In addition, increased risk of suicide attempts is a major public health concern during the first month of standard antidepressant therapy. Thus, improved therapeutics that can exert their antidepressant effects within hours or a few days of their administration are urgently needed, as is a better understanding of the presumed mechanisms associated with these rapid antidepressant effects. In this context, the N-methyl-d-aspartate (NMDA) antagonist ketamine has consistently shown antidepressant effects within a few hours of its administration. This makes it a valuable research tool to identify biomarkers of response in order to develop the next generation of fast-acting antidepressants. In this review, we describe clinical, electrophysiological, biochemical, and imaging correlates as relevant targets in the study of the antidepressant response associated with ketamine, and their implications for the development of novel, fast-acting antidepressants. We also review evidence that alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) to NMDA throughput may represent a convergent mechanism for the rapid antidepressant actions of ketamine. Overall, understanding the molecular basis of this work will likely lead to the ultimate development of improved therapeutics for MDD.

Introduction

Major depressive disorder (MDD) is a serious, recurrent, heterogeneous, and disabling psychiatric illness that affects millions of individuals worldwide, and has a major negative impact on public health and productivity (Baune et al., 2007, Kessler et al., 2006). More than half a century ago, it was observed that the antidepressant imipramine was clinically effective in treating MDD. Since then, a great many other antidepressants have been developed; these are largely similar to imipramine in their mechanism of action, but are more selective. For instance, selective serotonin reuptake inhibitors (SSRIs) are safer and better tolerated than tricyclic antidepressants, but they have not been equivocally shown to offer advantages in terms of efficacy or speed of onset of action. Most surprisingly, recent studies clearly show that antidepressants are of only limited effectiveness for many patients. For example, in the largest open-label study (STAR⁎D) evaluating standard therapies for MDD, less than one-third of patients receiving standard antidepressants experienced remission after up to four months of treatment (Thase et al., 2005, Trivedi et al., 2006). Furthermore, remission in half of the patients often required six months of treatment and two antidepressant trials (Judd et al., 2002, Trivedi et al., 2006).

In addition to the issue of efficacy, the delayed onset of therapeutic effects associated with standard antidepressants is still a major challenge in the treatment of MDD. This is a critical public health problem, because high rates of mortality and morbidity are present during this latency period, and these are associated with a worse long-term course (Machado-Vieira et al., 2008). Furthermore, despite the relatively large number of antidepressants available (Rush et al., 2006, Trivedi et al., 2006), our limited understanding of both the pathophysiology of MDD and the mechanisms of action involved in the therapeutic effects of antidepressants have been significant impediments to developing improved treatments. The key mechanism of action in the effects of standard antidepressants is thought to be their ability to increase monoamines in the synapses. Given that the clinical efficacy of these traditional agents is typically observed only after weeks of treatment, recent theories have suggested that monoamines are only primary effectors, modulating downstream signaling pathways responsible for their therapeutic efficacy (Manji and Lenox, 2000, Payne et al., 2002).

In recent years, it has become increasingly clear that there is an urgent need to develop pharmacological treatments for MDD that exert rapid and sustained antidepressant effects within hours or even a few days. In this context, the finding that the N-methyl-d-aspartate (NMDA) antagonist ketamine induces a rapid antidepressant response within hours has led to exciting new research into cellular mechanisms that affect rapid antidepressant action. Relatedly, accumulating evidence suggests that alterations in the regulation of glutamatergic neurotransmission contribute to the pathophysiology of MDD, as well as to the mechanism of existing antidepressants. This supporting evidence comes from: 1) findings of glutamatergic abnormalities in patients with MDD; 2) effects of existing antidepressants and mood stabilizing medications on the glutamatergic system; 3) preclinical evidence suggesting that drugs targeting various components of glutamate neurotransmission possess antidepressant and anxiolytic properties; and 4) recent studies demonstrating the effectiveness of glutamate-modulating agents in the treatment of mood disorders.

Thus, it appears that agents targeting the glutamatergic system may be key to developing a new generation of improved treatments for this devastating illness. In this article, we describe recent advances in this area with a special focus on the role of ketamine as a proof of concept agent for the development of new, improved treatments for MDD (Sanacora et al., 2008).

Glutamate is a critical excitatory neurotransmitter in the human brain system and is known to play a major role in cellular plasticity and cellular resilience (Sanacora et al., 2008). Diverse clinical studies have supported a critical role for the glutamatergic system in the pathophysiology of MDD, and it is believed to be a key target in mood regulation (reviewed in Maeng and Zarate (2007) ,Sanacora et al. (2008) andZarate et al. (2002)).

Glutamate acts pre- and postsynaptically through the activation of diverse receptors characterized by structural characteristics. Ionotropic glutamate receptors—NMDA, alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), and kainate (KA)—are ion channels that open the channel pore and allow ionic influx to the cell when activated. This influx regulates the neuronal surface's polarization, thus activating intracellular signaling cascades. In contrast, the metabotropic receptors (mGluRs) are G-protein-coupled receptors that directly modulate the second messenger pathways. The mGluRs in group I, including mGluR1 and mGluR5, stimulate the hydrolyzation of phosphoinositide phospholipids in the cell's plasma membrane. The receptors in groups II (including mGluR2 and mGluR3) and III (including mGluRs 4, 6, 7, and 8), with some exceptions, prevent the formation of cyclic adenosine monophosphate (cAMP) by activating a G-protein that inhibits the enzyme adenylyl cyclase, which forms cAMP from adenosine triphosphate (ATP). This review focuses on the ionotropic NMDA and AMPA receptors, which are believed to be directly involved in the antidepressant actions of ketamine (Fig. 1).

The NMDA receptor channel includes the combination of NR1, NR2 (NR2A–NR2D), and NR3 (NR3A and NR3B) subunits. Glutamate's binding site is believed to be in the NR2 subunit, while the NR1 subunit is the binding site for the glutamate co-agonist glycine. Two sites have been identified inside the ion channel: the “s” site, and the phencyclidine (PCP) site; the latter is ketamine's binding site. The AMPA receptors mediate the rapid, desensitizing excitation at most of the synapses, and are responsible for the early synaptic response to glutamate. When activated, AMPA receptors open the pore, thus allowing the influx of sodium and the consequent neuronal membrane's depolarization. The AMPA receptors comprise four subunits (GluR1–GluR4). At mature synapses, AMPA receptors are frequently co-expressed with NMDA receptors, where in concert they contribute to the synaptic plasticity processes involved in learning, memory, and neuroprotection (Malinow & Malenka, 2002).

Section snippets

Evidence for the involvement of the glutamatergic system in the pathophysiology of MDD: findings from human studies

Changes in the glutamatergic system have been described in the central nervous system (CNS) (CSF and brain tissue) as well as the periphery in subjects with MDD (Sanacora et al., 2008). It is not clear whether these peripheral changes in glutamate levels are a central dysfunction or represent a secondary finding regulated by other variables, including use of medication and peripheral sources of glutamate (Altamura et al., 1993, Frye et al., 2007, Kim et al., 1982, Mauri et al., 1998, Mitani et

The role of NMDA and AMPA receptor modulators as MDD therapeutics

Emerging data suggest that glutamate plays a critical role in both the acute and long-term action of antidepressants. Both in vitro and human studies show that new glutamate modulating agents as well as traditional antidepressants (Pittaluga et al., 2007, Sernagor et al., 1989) directly or indirectly target the glutamatergic system; furthermore, recent investigations specifically indicate the antidepressant efficacy of glutamate modulating agents in MDD. The glutamatergic modulators being

Ketamine: overview

Ketamine (dl2-(o-chlorophenyl)-2-(methylamino) cyclohexanone hydrochloride) is a non-competitive NMDA antagonist (Harrison & Simmonds, 1985) and a derivative of PCP. The original name for ketamine was CI581, and its discovery is credited to Dr. Calvin Stevens (Wayne State University) who isolated the compound in 1961. Ketamine is water- and lipid-soluble, and is easily administered through diverse routes with ample distribution in the body. Ketamine metabolism is regulated by hepatic microsomal

Enhancing AMPA to NMDA throughput as a convergent mechanism for the rapid antidepressant actions of ketamine

In order to develop the next generation of therapeutics that work more rapidly than ketamine and are better tolerated, it becomes crucial to understand the molecular and cellular basis for ketamine's antidepressant effects. With this goal in mind, our laboratory conducted a series of preclinical tests with ketamine in animal models of MDD. Preclinical evidence indicates a relevant interplay between AMPA and NMDA receptors in the rapid antidepressant effects of ketamine (Maeng and Zarate, 2007,

Conclusion and perspectives

The fact that monoaminergic antidepressants take weeks to achieve their full effect leaves patients receiving these medications particularly vulnerable to impairment in global functioning and at high risk of self-harm. The long and risky period of latency in MDD, as well as the persistent residual symptoms, low rates of remission, and frequent relapses are challenges that need to be better addressed by the next generation of improved therapeutics.

Several glutamatergic compounds are being tested

Acknowledgments

Funding for this work was supported by the Intramural Research Program of the National Institute of Mental Health (NIMH) and a NARSAD Award (CAZ). Ioline Henter provided outstanding editorial assistance. A patent application for the use of ketamine in depression has been submitted listing Dr. Zarate among the inventors. Dr. Zarate has assigned his rights on the patent to the US government.

References (97)

  • KarasawaJ. et al.

    AMPA receptor stimulation mediates the antidepressant-like effect of a group II metabotropic glutamate receptor antagonist

    Brain Res

    (2005)
  • KristiansenL.V. et al.

    Abnormal striatal expression of transcripts encoding NMDA interacting PSD proteins in schizophrenia, bipolar disorder and major depression

    Schizophr Res

    (2005)
  • LevineJ. et al.

    Increased cerebrospinal fluid glutamine levels in depressed patients

    Biol Psychiatry

    (2000)
  • MaengS. 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)
  • ManjiH.K. et al.

    Signaling: cellular insights into the pathophysiology of bipolar disorder

    Biol Psychiatry

    (2000)
  • ManjiH.K. et al.

    Enhancing neuronal plasticity and cellular resilience to develop novel, improved therapeutics for difficult-to-treat depression

    Biol Psychiatry

    (2003)
  • MaybergH.S. et al.

    Deep brain stimulation for treatment-resistant depression

    Neuron

    (2005)
  • MeloniD. et al.

    Dizocilpine antagonizes the effect of chronic imipramine on learned helplessness in rats

    Pharmacol Biochem Behav

    (1993)
  • MickleyG.A. et al.

    Ketamine blocks a conditioned taste aversion (CTA) in neonatal rats

    Physiol Behav

    (1998)
  • MitaniH. et al.

    Correlation between plasma levels of glutamate, alanine and serine with severity of depression

    Prog Neuropsychopharmacol Biol Psychiatry

    (2006)
  • Nudmamud-ThanoiS. et al.

    The NR1 subunit of the glutamate/NMDA receptor in the superior temporal cortex in schizophrenia and affective disorders

    Neurosci Lett

    (2004)
  • PadovanC.M. et al.

    Antidepressant-like effects of NMDA-receptor antagonist injected into the dorsal hippocampus of rats

    Pharmacol Biochem Behav

    (2004)
  • PappM. et al.

    Antidepressant activity of non-competitive and competitive NMDA receptor antagonists in a chronic mild stress model of depression

    Eur J Pharmacol

    (1994)
  • PappM. et al.

    Antidepressant-like effects of 1-aminocyclopropanecarboxylic acid and D-cycloserine in an animal model of depression

    Eur J Pharmacol

    (1996)
  • PayneJ.L. et al.

    Timing is everything: does the robust upregulation of noradrenergically regulated plasticity genes underlie the rapid antidepressant effects of sleep deprivation?

    Biol Psychiatry

    (2002)
  • PhelpsL.E. et al.

    Family history of alcohol dependence and initial antidepressant response to an N-methyl-d-aspartate antagonist

    Biol Psychiatry

    (2009)
  • PittalugaA. et al.

    Antidepressant treatments and function of glutamate ionotropic receptors mediating amine release in hippocampus

    Neuropharmacology

    (2007)
  • PrzegalinskiE. et al.

    Antidepressant-like effects of a partial agonist at strychnine-insensitive glycine receptors and a competitive NMDA receptor antagonist

    Neuropharmacology

    (1997)
  • SernagorE. et al.

    Open channel block of NMDA receptor responses evoked by tricyclic antidepressants

    Neuron

    (1989)
  • SkolnickP.

    Antidepressants for the new millennium

    Eur J Pharmacol

    (1999)
  • TrullasR. et al.

    Functional antagonists at the NMDA receptor complex exhibit antidepressant actions

    Eur J Pharmacol

    (1990)
  • AltamuraC.A. et al.

    Plasma and platelet excitatory amino acids in psychiatric disorders

    Am J Psychiatry

    (1993)
  • BeneytoM. et al.

    Lamina-specific abnormalities of AMPA receptor trafficking and signaling molecule transcripts in the prefrontal cortex in schizophrenia

    Synapse

    (2006)
  • Boyce-RustavJ.M. et al.

    Ethanol-related behaviors in mice lacking the NMDA receptor NR2A subunit

    Psychopharmacology (Berl)

    (2006)
  • BrittG.C. et al.

    A brief overview of the clinical pharmacology of "club drugs"

    Subst Use Misuse

    (2005)
  • CampbellS. et al.

    The role of the hippocampus in the pathophysiology of major depression

    J Psychiatr Neurosci

    (2004)
  • ChoudaryP.V. et al.

    Altered cortical glutamatergic and GABAergic signal transmission with glial involvement in depression

    Proc Natl Acad Sci U S A

    (2005)
  • ChourbajiS. et al.

    AMPA receptor subunit 1 (GluR-A) knockout mice model the glutamate hypothesis of depression

    FASEB J

    (2008)
  • CorrellG.E. et al.

    Two case studies of patients with major depressive disorder given low-dose (subanesthetic) ketamine infusions

    Pain Med

    (2006)
  • CorteseB.M. et al.

    The role of glutamate in anxiety and related disorders

    CNS Spectr

    (2005)
  • de GraafR.A. et al.

    In vivo 1H-[13C]-NMR spectroscopy of cerebral metabolism

    NMR Biomed

    (2003)
  • DeakinJ.F. et al.

    Glutamate and the neural basis of the subjective effects of ketamine: a pharmaco-magnetic resonance imaging study

    Arch Gen Psychiatry

    (2008)
  • DuJ. et al.

    Modulation of synaptic plasticity by antimanic agents: the role of AMPA glutamate receptor subunit 1 synaptic expression

    J Neurosci

    (2004)
  • DuJ. et al.

    The anticonvulsants lamotrigine, riluzole, and valproate differentially regulate AMPA receptor membrane localization: relationship to clinical effects in mood disorders

    Neuropsychopharmacology

    (2007)
  • EntsuahA.R. et al.

    Response and remission rates in different subpopulations with major depressive disorder administered venlafaxine, selective serotonin reuptake inhibitors, or placebo

    J Clin Psychiatry

    (2001)
  • FaragunaU. et al.

    A causal role for brain-derived neurotrophic factor in the homeostatic regulation of sleep

    J Neurosci

    (2008)
  • FrancisP.T.

    Glutamatergic systems in Alzheimer's disease

    Int J Geriatr Psychiatry

    (2003)
  • GoforthH.W. et al.

    Rapid relief of severe major depressive disorder by use of preoperative ketamine and electroconvulsive therapy

    J Ect

    (2007)
  • Cited by (230)

    • Arylcyclohexamines

      2021, Novel Psychoactive Substances: Classification, Pharmacology and Toxicology
    View all citing articles on Scopus
    View full text