You are on page 1of 11

Ann. N.Y. Acad. Sci.

ISSN 0077-8923

A N N A L S O F T H E N E W Y O R K A C A D E M Y O F SC I E N C E S
Issue: Translational Neuroscience in Psychiatry

Translational psychiatry—light at the end of the tunnel


Kenneth A. Jones,1 Frank S. Menniti,2 and Digavalli V. Sivarao3
1
Cyanaptic LLC, Glen Rock, New Jersey. 2 Mnemosyne Pharmaceuticals, Inc., Providence, Rhode Island. 3 Bristol–Myers
Squibb Co., Wallingford, Connecticut

Address for correspondence: Kenneth A. Jones, Cyanaptic LLC, 25 Warren Place, Glen Rock, NJ 07452.
kenjones120@gmail.com

Neuroscience has made tremendous progress delineating the cellular and molecular processes important for under-
standing neuronal development and behavior, but this knowledge has been slow to translate to new treatments for
psychiatric illness. To accelerate this transfer of knowledge to the human condition requires the wide-scale adoption
of biomarkers that can bridge preclinical and clinical discoveries, and serve as surrogate measures of efficacy before
commencing expensive phase III studies. Several biomarker methodologies, including imaging, electroencephalog-
raphy (EEG), and blood transcriptomics/proteomics, are now showing promise. From an industry perspective, we
highlight the utility of quantitative EEG as one example of a translatable biomarker applicable to psychiatric drug
development and discuss recent insights into glutamate system dysfunction in schizophrenia and depression gained
through translational studies of the drug ketamine.

Keywords: translational; psychiatry; biomarker; electroencephalography; glutamate; ketamine

Introduction: translational psychiatry imbalance? While we may infer answers to these


questions from animal studies, the limited abil-
Despite the tremendous growth in the field of neu-
ity to gather data directly from humans suffering
roscience over the past 30 years, the number of
from central nervous system (CNS) dysfunction
truly novel treatments for psychiatric disorders has
has been a longstanding bottleneck to treatment
been disappointing. Many reasons come to mind
development.
to explain this apparent disconnect, including an
The good news for brain research is that an
early naiveté about the complexity of the nervous
increasingly sophisticated armament of tools is
system, lack of disease models, and the inadequacy
being applied by neuroscientists to the human brain
of currently used end points for gauging the suc-
to answer these questions, offering new insights into
cess of clinical trials. However, perhaps the most
the nature of brain dysfunction that gives rise to
significant impediment has been the relative inac-
psychiatric maladies. To discuss this rapidly advanc-
cessibility of the human brain to direct measure-
ing area was the impetus for a 1-day conferencea
ments of molecular and cellular function. There
“Translational Psychiatry: Light at the End of the
is no question that the complexity of the human
Tunnel,” the spirit of which can be captured in the
brain is staggering, where numbers of neurons and
following: nothing can be done to reduce the com-
permutations of synaptic connections are immense,
plexities of the brain so they should be embraced,
and yet somehow from this complexity, sentience,
and the heretofore inadequate accessibility of the
dexterity, planning, creativity, and emotion are
brain, lack of disease models, and quantitative
born. But what happens when something goes
wrong?—when the developmental path deviates at
some point or a cluster of genes fails to main- a
Held at the New York Academy of Sciences, New York,
tain the balance of synapses, and mood plummets, NY, on April 8, 2014. Papers prepared by speakers at the
thoughts race, or hallucinations arise? Which are conference are published in Ann. N.Y. Acad. Sci. 1344:
the circuits or neurotransmitters that manifest the 1–119 (2015).

doi: 10.1111/nyas.12725
Ann. N.Y. Acad. Sci. 1344 (2015) 1–11 
C 2015 New York Academy of Sciences. 1
Translational psychiatry Jones et al.

clinical end points are essentially technical issues aspect of the human illness. As a result, new drugs
that ultimately will be addressed. Optimism for discovered using these models looked very much
this comes, in part, from sophisticated imaging and like the ones used to validate the models in the first
electroencephalography (EEG) technologies that place; the current generation of antidepressant and
open a rich information window into human brain antipsychotic drugs has provided limited improve-
function and dysfunction. These insights, in turn, ments in efficacy relative to drugs described 30 years
are being translated back to the preclinical realm to ago.
guide yet more sophisticated analyses in preclinical Another major reason for the exit of a significant
species, the development of better animal disease portion of the pharma industry from psychiatric
models, and the investigation of new drug targets. drug R&D has been the lack of target engagement
The circle is being completed by forward translation and efficacy biomarkers. A careful analysis2 of data
of this preclinical research into new biomarkers for from dozens of clinical drug development studies at
target engagement and efficacy that may be used in Pfizer concluded that to maximize odds for success
the clinical evaluation of potential new drugs. This in phase III, it was imperative to ensure that the
paper provides the background and impetus for drug in question not only is present at the target
the conference, an industry perspective on recent site but also has a demonstrable pharmacodynamic
trends in clinical development in psychiatry, and a activity commensurate with engagement.2 Not hav-
brief overview of some current approaches to find ing a clear pharmacodynamic marker often leads to
suitable biomarkers for psychiatric diseases. We a highly unsatisfactory situation of being unable to
focus our discussion on the emerging recognition determine whether the original hypothesis had been
of the role of glutamate neurotransmission dys- tested. While it may seem obvious that unequivo-
function in psychiatric illness and the use of EEG cal evidence for functional engagement is an essen-
methodology as a tool for novel discovery. tial component of a drug’s clinical development,
demonstrating such engagement has been a signif-
An industry perspective and the growing
icant challenge for most psychiatric drug discovery
need for biomarkers
to date.3–5
Despite the compelling need for new and improved The current situation has drawn the attention
medications to address the large unmet needs of of the National Institute of Mental Health (NIMH)
psychiatry patients, currently there is a near stand- and other thought leaders, who have reflected on
still in industry discovery.1 This is particularly true past failures to suggest new approaches to accelerate
at large pharmaceutical companies, traditionally the scientific breakthroughs.1,6 Over the past several
most prolific stakeholders in shepherding new drugs years, attention has shifted away from behav-
to clinical practice. Large pharmaceutical compa- ioral animal models based on predictive validity
nies occupy a particularly important niche in the derived from currently used agents toward genetic
lengthy and costly process of the late stages of drug approaches to understanding brain function and
development: the planning and execution of phase II dysfunction at the molecular and circuit levels.7,8
and phase III efficacy trials. However, the frequently The affordability of genomic sequencing has driven
ambiguous outcomes of phase II proof-of-concept the discovery of multiple susceptibility genes for
studies, in the face of increased costs of running large schizophrenia and autism,9–11 as but two examples.
clinical trials, have led to a recent exodus of pharma Although the polygenic nature of these and other
from their traditional niche. The perception, for psychiatric diseases presents a new challenge for
better or for worse, is that the underlying science those building genetically based disease models,
remains immature relative to the investment risks collectively, the gene defects point to key foci of
for success in clinical development. A major stum- neural function, such as synaptic transmission and
bling block often cited is the poor predictive value of circuit development.7,9,10 These promising discov-
animal models of psychiatric disease. A case in point eries portend a more molecular and circuit-based
is animal models of depression. Models such as the understanding of the processes that are compro-
forced swim test were widely adopted because they mised in disease, and importantly, improved animal
showed responses to existing antidepressant drugs, models and translatable endophenotypes that will
not because they replicated a key neurophysiological have prognostic value in the clinic.8,12

2 Ann. N.Y. Acad. Sci. 1344 (2015) 1–11 


C 2015 New York Academy of Sciences.
Jones et al. Translational psychiatry

Genetics alone, however, will not solve the cur- biomarkers may one day offer the practicality of
rent drug discovery bottleneck. There is also new use required for inclusion in large clinical trials.
urgency for developing truly translatable measures
Use of quantitative EEG in drug discovery
of brain activity relevant to psychiatry that span
both the laboratory and the clinic. In other CNS dis- EEG-based techniques are used to illuminate cir-
eases such as Alzheimer’s disease, the first biomark- cuit dysfunction underlying psychiatric illness and
ers will soon be available as surrogate end points for quantify circuit-relevant pharmacodynamic activ-
proof-of-concept clinical trials.13 The importance of ity of novel drugs. Glutamatergic principal neu-
these developments cannot be overstated especially rons form the core circuitry of the forebrain,
for long-duration, and therefore expensive, human and modulation of glutamatergic synapses is the
studies. Translatable biomarkers for psychiatric dis- brain’s informational currency. Activity in forebrain
orders may not be far behind. To spur the develop- glutamatergic circuits, as modulated by the gamma-
ment of tools that will provide these measures, the aminobutyric acid (GABA)ergic interneuron net-
NIMH recently released new guidelines for requir- work, gives rise to the translational imaging and
ing the incorporation of biomarkers in every clinical electrophysiological signals currently under investi-
trial sponsored by the institute.14 Although perhaps gation by many investigators.b Emerging from these
burdensome, this new requirement promises to pro- studies is insight into the circuit-level dysfunction
vide critical data linking surrogate end points to that gives rise to schizophrenia and depression.
primary measures of clinical efficacy. It is impor- Several EEG-based functional biomarkers, such as
tant to distinguish prognostic biomarkers that can mismatch negativity and 40-Hz auditory steady-
be used to predict efficacy in humans from diagnos- state response, lend themselves to interrogate
tic biomarkers that can be used to correctly identify specific circuit-level dysfunction.23
disease subtypes. It is primarily the former that is We highlight here the characteristics of quanti-
discussed here, particularly biomarkers with both tative EEG (qEEG) as a relatively inexpensive and
prognostic and translational value, even though accessible functional engagement measure that can
diagnostic biomarkers will be needed in psychiatry be used to bridge preclinical and early clinical stud-
to correctly identify patient segments that may bene- ies for glutamatergic and other targets. We then
fit from specific treatments. Examples of prognostic focus on the recent explosion of cross-species stud-
biomarker development include more sophisticated ies using glutamatergic drugs, notably ketamine,
and widely available magnetic resonance imaging that are beginning to yield an understanding of the
(MRI) and functional MRI technologies used in the molecular mechanisms underlying circuit dysfunc-
study of depression that are zeroing in on areas of tion in depression and schizophrenia. The conver-
the brain, such as the amygdala and medial pre- gence of these two themes may well represent the
frontal cortex, that emit different signals in nor- beginning of a renaissance in CNS drug discovery.
mal and depressed individuals.15,16 Likewise, in the On the basis of our experience from the applica-
field of schizophrenia, EEG methods are revealing tion of qEEG to multiple CNS discovery programs
electrical signatures of cognitive deficits whose early as well as selected examples from the published
appearance in children predict the development of literature, we will discuss several key factors that
the illness later in life.17,18 When used in concert impact translatability. Many articles and reviews
with event-related potentials, EEG exhibits the tem- have been published on qEEG as a potential phar-
poral resolution necessary to detect deficits in sen- macodynamic measure for early discovery.19,24–26
sory processing or attention.19 Finally, blood-based Some have focused on analytical methods for
biomarkers, particularly for depression, are reveal- qEEG that need to be appreciated for effective
ing underappreciated biochemical links between the application.27,28 We encourage the reader to consult
periphery and the brain. Interestingly, some of these these monographs for an in-depth review of qEEG
biomarkers normalize with treatment while others as a technique, as well as the analytical approaches
do not, raising the possibility that certain blood used. Here, we summarize key attributes that we
biomarkers could be helpful for identifying patient
subpopulations responsive to treatment.20–22 If
b
sufficiently developed and validated, peripheral Including some of the speakers at the conference.

Ann. N.Y. Acad. Sci. 1344 (2015) 1–11 


C 2015 New York Academy of Sciences. 3
Translational psychiatry Jones et al.

believe are essential for using qEEG as a transla- drug-related activity in the brain and to drive early
tional pharmacodynamic measure and illustrate clinical discovery.2,3,5
how these have facilitated its use as a critical In the absence of any existing clinical data that
biomarker for N-methyl-D-aspartate (NMDA) support the use of qEEG as a pharmacodynamic
channel blockers, especially in relation to treatment biomarker for a given target, which often is the
options for treatment-resistant depression. case for novel targets, a starting point would be
The EEG signal arises principally from summated to demonstrate a robust effect in experimental ani-
postsynaptic activity of large populations of neurons mals in a dose-ranging study. Using rodents as a
within the cortical mantle. While neurons through- starting point to investigate the potential for qEEG
out the brain contribute to fluctuations in the field as a pharmacodynamic biomarker is a viable strat-
potential, the relatively large and numerous cor- egy, provided the cross-species target physiology is
tical pyramidal neurons with their extensive den- conserved. If qEEG effects in rodents are robust and
dritic processes and laminar arrangement are the are determined to be specific to the target, and in
principal sources of fluctuating field potential reg- the absence of clinical data, moving on to a higher
istered by scalp electrodes. Quantitative EEG as a species, such as a nonhuman primate, may be con-
technique refers to dividing the oscillatory EEG sig- sidered as a bridge to clinical studies.
nal into broad frequency bands and analyzing the Preclinically, EEG is frequently recorded from
signal power within these bands.29 While we do epidural electrodes that bypass the skull and sample
not completely understand the individual signifi- directly from the brain where the signal registration
cance of these bands and how they may interact is stronger and spectrally more complete. In order to
with each other in support of physiological func- have a greater probability for success across species,
tion, we do know that in many regions of the cor- it is important to consider not only statistical sig-
tex, higher-frequency oscillations are nested within nificance but also how robust the drug-mediated
a slower-frequency field potential change,30,31 and effects are. Thus, an effect size analysis using an indi-
that inhibitory and excitatory neurons in the cor- cator such as Cohen’s d (mean difference between
tex and thalamic nuclei interact to produce these control and treatment in pooled standard deviation
fluctuations.32,33 Rhythmic oscillations of a fre- units) is warranted. As a rule of thumb, a Cohen’s
quency band are often associated with particular d of 0.8 or greater could trigger further investi-
physiological states. For example, delta frequency gation for translatability. A recent study used two
(0.5–4 Hz) oscillations occur in deep sleep as a NMDA channel blockers, ketamine and lanicem-
result of thalamic burst firing. Sleep spindles in the ine, to show robust and dose-dependent increases
alpha frequency range (9–13 Hz) are generated in gamma oscillations in rodents that were subse-
by the thalamic reticular nucleus whereas the occip- quently translated into healthy human volunteers.38
ital alpha rhythm, characteristic of relaxed wake- In addition to these two examples, there is a sig-
fulness, is generated in deep layers of the visual nificant body of literature that suggests that this
cortex. Higher frequency beta (13–30 Hz) and translational approach is indeed viable for certain
gamma (30–100 Hz) oscillations, associated with classes of drugs.24,39,40
mental alertness, appear to originate locally in the Despite showing consistent and strong effects, it
cortex,34,35 although these too can be markedly is frequently not possible to explain the exact basis
influenced by thalamic input.36,37 Since several for qEEG changes because they ensue following
classes of drugs affect specific oscillatory bands drug exposure to very large and diverse populations
in an exposure- and time-dependent fashion, pre- of neurons in the brain. Moreover, multiple drug
sumably owing to their effects on the underlying classes can in some cases produce the same qEEG
circuits, a tactical opportunity arises to use these response. For example, both benzodiazepine ago-
changes as a pharmacodynamic biomarker as well nists and NMDA antagonists can increase cortical
as to model effects over time to make predictions high-frequency oscillations, albeit through distinct
regarding dose regimen and interval. It is important molecular targets.41,42 However, it is possible to
to recognize that the qEEG changes may or may determine specificity by demonstrating a causal
not be related to efficacy of the drug, yet they can relationship between the target and the response.
be used as an engagement marker to demonstrate For example, benzodiazepine agonist effects on

4 Ann. N.Y. Acad. Sci. 1344 (2015) 1–11 


C 2015 New York Academy of Sciences.
Jones et al. Translational psychiatry

qEEG are blocked by selective antagonism with An important attribute for a pharmacodynamic
flumazenil, a benzodiazepine-selective antagonist, biomarker is for the signal to scale in a dose- or
in rodents41 and in humans.43 On the other hand, exposure-dependent manner. Apart from NMDA
gamma oscillations elicited by NMDA channel channel blockers, several other pharmacologi-
blockers are thought to result from the disinhibi- cal classes of drugs evoke dose- or exposure-
tion of pyramidal cells owing to the NMDA block proportional EEG responses, including opiates,
of parvalbumin+ inhibitory interneurons.44,45 barbiturates, benzodiazepines, and inhalational and
In fact, MK-801–induced gamma oscillations injectable anesthetics, and lend their effects to phar-
have been shown to be attenuated in genetically macokinetic–pharmacodynamic correlation.24,25 A
engineered mice that express a deficient NMDA corollary follows that the lack of a clear linearity of
channel complex in parvalbumin+ interneurons.46 dose response in qEEG measures would suggest that
Additional means to ensure causality would be this tool would be of limited utility for a phase-I
to demonstrate, where available, differentiation biomarker. For example, nicotinic alpha7 recep-
of response using enantiomers (e.g., R- versus tor agonists frequently show an inverted U-shaped
S-baclofen47 ) or by contrasting two compounds response across multiple paradigms,51,52 perhaps
with similar in vitro activity against the target but owing to rapid desensitization of the nicotinic chan-
different CNS penetration. The characterization of nels at higher exposure. We have observed that EEG
specificity at the preclinical stage is important not changes seen at lower doses frequently disappear at
only to ensure that the EEG response is linked to higher doses (unpublished data), making the use
the target in question but also because, frequently, of qEEG for this particular target unsuitable as a
this is the only stage where it is practical to do biomarker.
so. In summary, a significant challenge in psychiatric
Although any drug that works on the nervous drug discovery is to have translational biomarkers
system could in theory affect the qEEG response, that can inform the researchers about the unequivo-
in practice such changes are large and consistent cal presence of the drug being tested in the brain and
enough only for certain classes of drugs. Since its time course, using affordable, noninvasive, and
synchronization within cortical columns is largely nondeleterious approaches. Translatability implies
responsible for the scalp-recorded EEG signal, it is that the qualitative changes noted in preclinical
important for any candidate drug to target, either species (e.g., rodents and/or primates) are essen-
directly or indirectly, cortical neurons. Immunohis- tially reproducible in humans. Quantitative EEG
tochemical labeling with an antibody raised against offers a simple and relatively inexpensive modal-
the obligatory subunit NR1 of the NMDA channel ity for translation across preclinical species and
demonstrates a robust presence of the receptor humans. While this measure cannot always be linked
within the cortex, both on pyramidal cell layers to the overall efficacy of a drug, it can offer a pharma-
and on inhibitory neurons across multiple species, codynamic sign of the drug’s presence and activity
including rodents and humans.48–50 Commensurate in the brain. Such functional markers, as opposed to
with the widespread expression of its molecular physical engagement markers (e.g., receptor occu-
target, nonselective channel blockers, such as pancy or positron emission tomography imaging),
ketamine, produce a strong cortical disinhibition may be especially relevant for high-efficacy agonists
as evidenced by a selective reduction of fast-spiking that produce pharmacological responses at very low
putative GABAergic interneurons and a widespread occupancy, an observation explained by the con-
increase in pyramidal cell firing in rodents.44 NMDA cept of high receptor reserve.53,54 Quantitative EEG
channel blockers produce a robust, dose-dependent, effects in early discovery can guide dose selection
and long-lasting increase in high-frequency gamma and dosing interval for subsequent phases of clini-
oscillations (e.g., 30–55 Hz) in mice and rats, as well cal discovery. However, qEEG is not suitable for all
as in humans as discussed above.38,42,45,46 Perhaps centrally acting targets. Its translational utility
for this reason alone, many of the clinical studies in would be target-specific and determined by such
psychiatry that employ EEG as a response measure factors as the cortical footprint of the target, effect
are evaluating drugs that target the NMDA channel. size, and exposure–response linearity.

Ann. N.Y. Acad. Sci. 1344 (2015) 1–11 


C 2015 New York Academy of Sciences. 5
Translational psychiatry Jones et al.

New treatments targeting glutamate psychosis in schizophrenia.60 Subsequently, it was


neurotransmission determined that these effects of PCP result from
NMDA receptor inhibition. Ketamine, a PCP ana-
A second translational approachc is the use of the
log, was then adapted as the clinical translational
drug ketamine to probe the role of glutamate in
tool to systematically probe these psychotomimetic
psychiatric illness. Ketamine is an antagonist of the
effects of NMDA receptor blockade.61,62 In fact,
NMDA receptor, one of the four classes of receptors
clinical studies by Javitt et al. established that
(AMPA, kainate, NMDA, and metabotropic) that
ketamine produces not only acute psychotic symp-
mediate glutamate neurotransmission. NMDA
toms, but also broader cognitive and negative
receptors have a critical function in regulating the
symptoms experienced by patients suffering from
excitation/inhibition balance that governs infor-
schizophrenia.62–64 Collectively, these clinical data
mation flow through glutamatergic/GABAergic
have given rise to the hypothesis that NMDA recep-
networks.55,56 These receptors also play the prin-
tor hypofunction is a key molecular defect underly-
cipal role in regulating glutamate synaptic strength
ing the expression of all of the complex symptoms
through the activation of pathways that insert or
of schizophrenia.64,65 Recent genetic evidence sup-
remove AMPA receptors from the synapse and that
ports this hypothesis.66 Indeed, the NMDA hypo-
trigger protein translational and transcriptional
function hypothesis is now driving the search for
pathways that support long-term modification
new treatments for schizophrenia.63,67
of synapses.57–59 There is a rich pharmacology
The more recent story of ketamine in depression
of NMDA receptor antagonists and these agents
arises from the drug’s use as a clinical translational
have been part of the experimental toolbox that
probe, in this case of cognitive dysfunction in
has helped gain our current understanding of
depression.68 Berman et al. gained approval in the
glutamate system physiology.56 Numerous NMDA
late 1990s to use ketamine to investigate cognition
receptor antagonists have been advanced into
in depression.68 In their study, a short intravenous
clinical trials for indications including anesthesia,
infusion of ketamine resulted in the expected acute
neuroprotection in stroke, neuropathic pain,
PCP-like psychotomimetic effects; these waned
Parkinson disease, and Alzheimer’s disease. While
rapidly after the infusion was stopped, consistent
these efforts failed to yield commercial successes
with the drug’s short half-life and indicative that
(with the exception of memantine for Alzheimer’s
these were the direct result of on-target NMDA
disease), the clinical observations proved a treasure
receptor inhibition. However, as patients waited to
trove of insight into how glutamate system dys-
be discharged, a number reported lessening of their
function may be involved in both schizophrenia
feelings of depression. Berman et al. documented
and depression. Ketamine is an approved drug,
this phenomenon to reveal a clinically significant
albeit only for limited use as a pediatric anesthetic.
antidepressant response to ketamine that developed
However, because of its availability for clinical
in the hours after the drug had been cleared from
studies, ketamine has emerged as a powerful cross-
the body.68 This finding, published in 2000, was
species translational tool in the systematic investi-
replicated in 2006 by Zarate et al.69 Since then,
gation of the previous clinical observations. This
interest in the rapid-onset antidepressant effects of
line of investigation has been, and continues to be,
ketamine gained momentum and well over a dozen
provocative of a renewed interest in the pursuit of
replications have now been published.70–72
drugs to treat psychiatric disease, as summarized
The robust clinical findings with ketamine, both
below.
as a model of schizophrenia and as an antidepres-
In the late 1950s, Luby and coworkers found that
sant, implicate glutamate system dysfunction at
in healthy volunteer studies, phencyclidine (PCP)
the core of two of the most debilitating and costly
produced a syndrome strikingly similar to acute
psychiatric illnesses. These clinical findings have
spurred back-translational studies to understand
c
Discussed by several speakers at the conference. See Ann. the molecular underpinnings that account for
N.Y. Acad. Sci. 1344: 1–119 (2015), including the papers by these findings.73–78 What are the drug-on effects
Javitt, Featherstone et al., Sivarao, Abdallah et al., Leuchter that induce schizophrenia-like symptoms, and
et al., and Lener & Iosifescu. what is the nature of the drug-off effect that

6 Ann. N.Y. Acad. Sci. 1344 (2015) 1–11 


C 2015 New York Academy of Sciences.
Jones et al. Translational psychiatry

alleviates depression? Are these effects interrelated stage for back-translational studies in both humans
and, if so, what might that imply regarding the and preclinical species that have yielded an impor-
relatedness of these psychiatric conditions? (See tant mechanistic framework for understanding the
Refs. 79–84 for a discussion of these questions.) nature of this dysfunction. Significantly, ketamine
A prevalent hypothesis stems from the putative as a clinical probe continues to deliver new insights,
selectivity of ketamine for NMDA receptors on with reports of beneficial therapeutic effects in
neurons that are most active. Essentially, NMDA patients suffering from bipolar disorder,89 obses-
receptor channel block by ketamine is activity sive compulsive disorder,90 and posttraumatic stress
dependent,85 and since NMDA receptors are most disorder.91 However, although the translational
active on active neurons, such neurons are most studies with ketamine shed light, they are not the end
sensitive to ketamine. Attention has focused on of the tunnel. For example, preclinical data suggest
selective ketamine inhibition of cortical GABAergic additional complexity to the mechanistic frame-
fast-spiking, parvalbumin+ interneurons, which work laid out above. There are multiple NMDA
are believed to account for two functional effects receptor subtypes, of which NR2A and NR2B
of the drug.44,86 Inhibition of NMDA receptors subtypes predominate in the forebrain.58,92 Com-
on the parvalbumin interneurons disrupts their pounds highly selective for inhibition of the NR2B
ability to regulate cortical synchrony,86 reflected subtype have been developed, including Ro 25–
in a shift of cortical gamma band frequency and 698193 and CP-10160694 that have been used exten-
power and decreased responsiveness to external sively in preclinical studies to investigate the role of
stimuli.87 The disruption of gamma synchrony the NR2B subtype. Significantly, in humans, CP-
is hypothesized to underlie the acute, drug- 101606 causes dose-dependent psychotomimetic
on cognitive-disrupting and psychotomimetic effects that, at high doses, appear to be similar to
effects of the drug. Selective inhibition of NMDA those produced by ketamine.95–98 Also similar to
receptors on parvalbumin interneurons also ketamine, a short exposure to CP-101606 produces a
reduces inhibition by these neurons of gluta- robust antidepressant response that occurs after the
matergic principal neurons, resulting in a net drug has been cleared from the body.98 These clin-
cortical hyperglutamatergic state.44,86 This drug-on ical data suggest that selective NR2B inhibition has
induction of gamma activity in the context of a therapeutic and adverse effects similar to ketamine
hyperglutamatergic state is hypothesized to engage in humans. Thus, back-translational studies of the
synaptic plasticity mechanisms that result in a long- mechanistic similarities and differences between
lasting synaptic potentiation, which may underlie ketamine and NR2B antagonists open a new path to
the drug-off antidepressant response. Clinical sup- further understanding of the role of glutamate sys-
port for this hypothesis is found in the observation tem dysfunction in psychiatric illness. To date, pre-
of an increase in somatosensory-evoked potentials, clinical data indicate that both drug classes induce
measured in response to tactile stimulation as an similar drug-off synaptic potentiation.73,74,76,77
increase in magnetoencephalographic gamma-band However, Sivarao et al. and others83,87,99 have
power that correlates with a drug-off reduction in found that the NR2B antagonists do not dis-
depressive symptoms in patients that responded to rupt gamma synchrony in rodents. This lack of
ketamine, but not in those that failed to respond.88 effect on gamma synchrony may be accounted for
Back-translational preclinical studies have provided by the fact that there appears to be little NR2B
biochemical73–77 evidence of such a long-lasting expression by fast-spiking GABAergic interneu-
synaptic potentiation after short ketamine expo- rons, which predominately express NR2A.100 These
sure. Thus, the hyperglutamatergic state hypothesis data raise questions regarding the role of gamma
of ketamine action provides a mechanistic link disruption in relation to both psychotomimetic
between the drug-on and drug-off effects. and antidepressant effects of NMDA receptor inhi-
Ketamine has proven to be a powerful trans- bition. Future preclinical studies should include
lational tool to probe the glutamate system in investigations of whether NR2B inhibition induces
psychiatric illness. The clinical behavioral data a hyperglutamatergic state similar to that of
strongly implicate glutamate system dysfunction in ketamine. Follow-on human translational stud-
schizophrenia and depression. These data set the ies might include investigations of the effects of

Ann. N.Y. Acad. Sci. 1344 (2015) 1–11 


C 2015 New York Academy of Sciences. 7
Translational psychiatry Jones et al.

NR2B antagonists on EEG signatures, particularly Conflicts of interest


gamma synchrony, and whether an antidepressant
F.S.M. is currently the Chief Scientific Officer of
response to such a drug is accompanied by a drug-
Mnemosyne Pharmaceuticals, Inc., which is devel-
off synaptic potentiation. Fortunately, the transla-
oping glutamate receptor modulators to treat psy-
tional approaches highlighted at the symposium are
chiatric disorders.
well suited to triangulating on mechanistic effects of
these two classes of NMDA antagonists and thereby
provide further insight into the role of glutamate
system dysfunction in psychiatric illness, that is, References
bringing us steps closer to the end of the tunnel. 1. Hyman, S.E. 2012. Revolution stalled. Sci. Transl. Med. 4:
155cm11.
Summary and perspective 2. Morgan, P. et al. 2012. Can the flow of medicines be
improved? Fundamental pharmacokinetic and pharmaco-
Advances in the understanding of psychiatric ill- logical principles toward improving Phase II survival. Drug
nesses and the ability to improve current pharma- Discov. Today 17: 419–424.
cotherapy will benefit from new tools that bridge 3. Berk, M. 2012. Pathways to new drug discovery in neu-
preclinical and clinical studies. The most powerful ropsychiatry. BMC Med. 10: 151.
4. Brady, L.S. & T.R. Insel. 2012. Translating discoveries into
of these tools will provide mechanistically concor-
medicine: psychiatric drug development in 2011. Neuropsy-
dant insights in both patients and laboratory test chopharmacology 37: 281–283.
species. We have provided some specific examples 5. Singh, I. & N. Rose. 2009. Biomarkers in psychiatry. Nature
of some promising translatable approaches, with a 460: 202–207.
focus on the use of EEG; other methods, such as 6. Insel, T.R. & N. Gogtay. 2014. National Institute of Mental
Health clinical trials: new opportunities, new expectations.
functional and structural MRI and blood biomark-
JAMA Psychiatry 71: 745–746.
ers, are being applied as well. EEG satisfies many 7. Grayton, H.M. et al. 2012. Copy number variations in neu-
criteria of a good translatable biomarker, namely rodevelopmental disorders. Prog. Neurobiol. 99: 81–91.
large effect size, ability to inform about disease- 8. Markou, A. et al. 2009. Removing obstacles in neuroscience
relevant brain circuits, homologous measurements drug discovery: the future path for animal models. Neu-
ropsychopharmacology 34: 74–89.
in both patients and laboratory animals, and dose-
9. Corvin, A. & D.W. Morris. 2014. Genome-wide association
dependent effects of the drug. Historical and recent studies: findings at the major histocompatibility complex
studies point to glutamatergic mechanisms as cen- locus in psychosis. Biol. Psychiatry 75: 276–283.
tral to the understanding of mental disorders, and 10. Gatt, J.M. et al. 2015. Specific and common genes impli-
although the role of glutamatergic signaling in dis- cated across major mental disorders: a review of meta-
analysis studies. J. Psychiatr. Res. 60: 1–13.
ease causation is poorly understood, glutamate and
11. Harrison, P.J. 2015. Recent genetic findings in schizophre-
its receptors dominate synaptic signaling, especially nia and their therapeutic relevance. J. Psychopharmacol. 29:
in cortical areas affected in psychiatric disorders. 85–96.
Perturbation of cortical function by ketamine and 12. Donaldson, Z.R. & R. Hen. 2015. From psychiatric dis-
other NMDA antagonists is readily detected by EEG orders to animal models: a bidirectional and dimensional
approach. Biol. Psychiatry 77: 15–21.
methods that are now being used to define surro-
13. Flood, D.G., G.J. Marek & M. Williams. 2011. Developing
gate markers for both the adverse and therapeutic predictive CSF biomarkers—a challenge critical to success
effects of these novel therapies.37 It may well be that a in Alzheimer’s disease and neuropsychiatric translational
marriage of the renewed interest in glutamate phar- medicine. Biochem. Pharmacol. 81: 1422–1434.
macology with robust translatable biomarkers will 14. Brady, L. 2014. Biomarker development and validation:
establishing standards of evidence for their context of use
provide the impetus for an improved mechanistic
in clinical trials. NAMHC Concept Clearance. National
understanding of psychiatric disorders. Institute of Mental Health. Accessed February 17, 2015.
http://www.nimh.nih.gov/funding/grant-writing-and-app
Acknowledgments lication-process/concept-clearances/2014/biomarker-deve
lopment-and-validation-establishing-standards-of-eviden
We wish to acknowledge the Biochemical Pharma-
ce-for-their-context-of-use-in-clinical-trials.shtml
cology Discussion Group at the New York Academy 15. Borsook, D. & L. Becerra. 2010. Using NMR approaches
of Sciences for their partnership in organizing the to drive the search for new CNS therapeutics. Curr. Opin.
conference. Investig. Drugs 11: 771–778.

8 Ann. N.Y. Acad. Sci. 1344 (2015) 1–11 


C 2015 New York Academy of Sciences.
Jones et al. Translational psychiatry

16. Borsook, D., L. Becerra & R. Hargreaves. 2006. A role for high frequency (gamma-band) oscillating potentials in rat
fMRI in optimizing CNS drug development. Nat. Rev. Drug auditory cortex. Brain Res. 721: 155–166.
Discov. 5: 411–424. 35. Gray, C.M. & D.A. McCormick. 1996. Chattering cells:
17. Kaur, M. et al. 2013. Longitudinal associations between superficial pyramidal neurons contributing to the genera-
mismatch negativity and disability in early schizophrenia- tion of synchronous oscillations in the visual cortex. Science
and affective-spectrum disorders. Prog. Neuropsychophar- 274: 109–113.
macol. Biol. Psychiatry 46: 161–169. 36. Barth, D.S. & K.D. MacDonald. 1996. Thalamic modula-
18. Perez, V.B. et al. 2014. Automatic auditory processing tion of high-frequency oscillating potentials in auditory
deficits in schizophrenia and clinical high-risk patients: cortex. Nature 383: 78–81.
forecasting psychosis risk with mismatch negativity. Biol. 37. Steriade, M. et al. 1996. Synchronization of fast (30-40 Hz)
Psychiatry 75: 459–469. spontaneous oscillations in intrathalamic and thalamocor-
19. Babiloni, C. et al. 2009. Fundamentals of electroencefalog- tical networks. J. Neurosci. 16: 2788–2808.
raphy, magnetoencefalography, and functional magnetic 38. Sanacora, G. et al. 2014. Lanicemine: a low-trapping
resonance imaging. Int. Rev. Neurobiol. 86: 67–80. NMDA channel blocker produces sustained antidepressant
20. Pajer, K. et al. 2012. Discovery of blood transcriptomic efficacy with minimal psychotomimetic adverse effects.
markers for depression in animal models and pilot valida- Mol. Psychiatry 19: 978–985.
tion in subjects with early-onset major depression. Transl. 39. Buhrer, M. et al. 1990. Electroencephalographic effects
Psychiatry 2: e101. of benzodiazepines. II. Pharmacodynamic modeling of
21. Redei, E.E. et al. 2014. Blood transcriptomic biomarkers in the electroencephalographic effects of midazolam and
adult primary care patients with major depressive disorder diazepam. Clin. Pharmacol. Ther. 48: 555–567.
undergoing cognitive behavioral therapy. Transl. Psychiatry 40. Danhof, M. et al. 2008. Mechanism-based pharmaco-
4: e442. kinetic-pharmacodynamic (PK-PD) modeling in transla-
22. Redei, E.E. & N.S. Mehta. 2015. Blood transcriptomic tional drug research. Trends Pharmacol. Sci. 29: 186–191.
markers for major depression: from animal models to clin- 41. Mandema, J.W., E. Tukker & M. Danhof. 1992. In vivo
ical settings. Ann. N.Y. Acad. Sci. 1344: 37–49. characterization of the pharmacodynamic interaction of
23. Luck, S.J. et al. 2011. A roadmap for the development a benzodiazepine agonist and antagonist: midazolam and
and validation of event-related potential biomarkers in flumazenil. J. Pharmacol. Exp. Ther. 260: 36–44.
schizophrenia research. Biol. Psychiatry 70: 28–34. 42. Sebban, C. et al. 2002. Effects of phencyclidine (PCP) and
24. Bewernitz, M. & H. Derendorf. 2012. Electroencepha- MK 801 on the EEGq in the prefrontal cortex of conscious
logram-based pharmacodynamic measures: a review. Int. rats; antagonism by clozapine, and antagonists of AMPA-,
J. Clin. Pharmacol. Ther. 50: 162–184. alpha(1)- and 5-HT(2A)-receptors. Br. J. Pharmacol. 135:
25. Leiser, S.C. et al. 2011. Aligning strategies for using EEG as 65–78.
a surrogate biomarker: a review of preclinical and clinical 43. Fiset, P. et al. 1995. Pharmacodynamic modeling of the
research. Biochem. Pharmacol. 81: 1408–1421. electroencephalographic effects of flumazenil in healthy
26. Mandema, J.W. & M. Danhof. 1992. Electroencephalogram volunteers sedated with midazolam. Clin. Pharmacol. Ther.
effect measures and relationships between pharmacokinet- 58: 567–582.
ics and pharmacodynamics of centrally acting drugs. Clin. 44. Homayoun, H. & B. Moghaddam. 2007. NMDA receptor
Pharmacokinet. 23: 191–215. hypofunction produces opposite effects on prefrontal cor-
27. Bronzino, J.D. 2000. “Principles of electroencephalog- tex interneurons and pyramidal neurons. J. Neurosci. 27:
raphy.” In The Biomedical Engineering Handbook. J.D. 11496–11500.
Bronzino, Ed.: 201–212. Boca Raton: CRC Publishers LLC. 45. Pinault, D. 2008. N-methyl d-aspartate receptor antago-
28. Thakor, N.V. & S. Tong. 2004. Advances in quantitative elec- nists ketamine and MK-801 induce wake-related aberrant
troencephalogram analysis methods. Annu. Rev. Biomed. gamma oscillations in the rat neocortex. Biol. Psychiatry
Eng. 6: 453–495. 63: 730–735.
29. Buzsaki, G. & A. Draguhn. 2004. Neuronal oscillations in 46. Carlen, M. et al. 2012. A critical role for NMDA receptors
cortical networks. Science 304: 1926–1929. in parvalbumin interneurons for gamma rhythm induction
30. Chrobak, J.J. & G. Buzsaki. 1998. Gamma oscillations in and behavior. Mol. Psychiatry 17: 537–548.
the entorhinal cortex of the freely behaving rat. J. Neurosci. 47. Mandema, J.W. et al. 1992. Modeling of the effect site
18: 388–398. equilibration kinetics and pharmacodynamics of racemic
31. Lakatos, P. et al. 2005. An oscillatory hierarchy control- baclofen and its enantiomers using quantitative EEG effect
ling neuronal excitability and stimulus processing in the measures. J. Pharmacol. Exp. Ther. 261: 88–95.
auditory cortex. J. Neurophysiol. 94: 1904–1911. 48. Cao, Z. et al. 2000. Postnatal development of NR1, NR2A
32. Llinas, R.R. 1988. The intrinsic electrophysiological prop- and NR2B immunoreactivity in the visual cortex of the rat.
erties of mammalian neurons: insights into central nervous Brain Res. 859: 26–37.
system function. Science 242: 1654–1664. 49. Conti, F. et al. 1999. Neuronal and glial localization of NR1
33. Llinas, R.R. & M. Steriade. 2006. Bursting of thalamic neu- and NR2A/B subunits of the NMDA receptor in the human
rons and states of vigilance. J. Neurophysiol. 95: 3297–3308. cerebral cortex. Cereb. Cortex 9: 110–120.
34. Brett, B., G. Krishnan & D.S. Barth. 1996. The effects of 50. Mower, G.D. & L. Chen. 2003. Laminar distribution of
subcortical lesions on evoked potentials and spontaneous NMDA receptor subunit (NR1, NR2A, NR2B) expression

Ann. N.Y. Acad. Sci. 1344 (2015) 1–11 


C 2015 New York Academy of Sciences. 9
Translational psychiatry Jones et al.

during the critical period in cat visual cortex. Brain Res. 68. Berman, R.M. et al. 2000. Antidepressant effects of keta-
Mol. Brain Res. 119: 19–27. mine in depressed patients. Biol. Psychiatry 47: 351–354.
51. Huang, M. et al. 2014. The novel alpha7 nicotinic acetyl- 69. Zarate, C.A. et al. 2006. A randomized trial of an
choline receptor agonist EVP-6124 enhances dopamine, N-methyl-D-aspartate antagonist in treatment-resistant
acetylcholine, and glutamate efflux in rat cortex and major depression. Arch. Gen. Psychiatry 63: 856–864.
nucleus accumbens. Psychopharmacology (Berl). 231: 4541– 70. Aan Het Rot, M. et al. 2012. Ketamine for depression: where
4551. do we go from here? Biol. Psychiatry 72: 537–547.
52. Rezvani, A.H. et al. 2009. Effect of R3487/MEM3454, a 71. Murrough, J.W. & D.S. Charney. 2010. Cracking the moody
novel nicotinic alpha7 receptor partial agonist and 5-HT3 brain: lifting the mood with ketamine. Nat. Med. 16: 1384–
antagonist on sustained attention in rats. Prog. Neuropsy- 1385.
chopharmacol. Biol. Psychiatry. 33: 269–275. 72. Niciu, M.J. et al. 2014. Glutamate receptor antagonists as
53. Grimwood, S. & P.R. Hartig. 2009. Target site occupancy: fast-acting therapeutic alternatives for the treatment of
emerging generalizations from clinical and preclinical stud- depression: ketamine and other compounds. Annu. Rev.
ies. Pharmacol. Ther. 122: 281–301. Pharmacol. Toxicol. 54: 119–139.
54. Hume, S.P. et al. 2007. Low sensitivity of the positron emis- 73. Duman, R.S. et al. 2012. Signaling pathways underlying the
sion tomography ligand [11C]diprenorphine to agonist rapid antidepressant actions of ketamine. Neuropharma-
opiates. J. Pharmacol. Exp. Ther. 322: 661–667. cology 62: 35–41.
55. Niswender, C.M. & P.J. Conn. 2010. Metabotropic glu- 74. Dwyer, J.M. & R.S. Duman. 2013. Activation of mammalian
tamate receptors: physiology, pharmacology, and disease. target of rapamycin and synaptogenesis: role in the actions
Annu. Rev. Pharmacol. Toxicol. 50: 295–322. of rapid-acting antidepressants. Biol. Psychiatry 73: 1189–
56. Traynelis, S.F. et al. 2010. Glutamate receptor ion channels: 1198.
structure, regulation, and function. Pharmacol. Rev. 62: 75. Kavalali, E.T. & L.M. Monteggia. 2012. Synaptic mecha-
405–496. nisms underlying rapid antidepressant action of ketamine.
57. Citri, A. & R.C. Malenka. 2008. Synaptic plasticity: multi- Am. J. Psychiatry 169: 1150–1156.
ple forms, functions, and mechanisms. Neuropsychophar- 76. Li, N. et al. 2010. mTOR-dependent synapse formation
macology 33: 18–41. underlies the rapid antidepressant effects of NMDA antag-
58. Cull-Candy, S.G., S. Brickley & M. Farrant. 2001. NMDA onists. Science 329: 959–964.
receptor subunits: diversity, development and disease. Curr. 77. Li, N. et al. 2011. Glutamate N-methyl-D-aspartate recep-
Opin. Neurobiol. 11: 327–335. tor antagonists rapidly reverse behavioral and synaptic
59. Malenka, R.C. & R.A. Nicoll. 1993. NMDA-receptor- deficits caused by chronic stress exposure. Biol. Psychia-
dependent synaptic plasticity: multiple forms and mecha- try 69: 754–761.
nisms. Trends Neurosci. 16: 521–527. 78. Maeng, S. et al. 2008. Cellular mechanisms underlying
60. Domino, E.F. & E.D. Luby. 2012. Phencyclidine/ the antidepressant effects of ketamine: role of ␣-amino-
schizophrenia: one view toward the past, the other to the 3-hydroxy-5-methylisoxazole-4-propionic acid receptors.
future. Schizophr. Bull. 38: 914–919. Biol. Psychiatry 63: 349–352.
61. Javitt, D.C. et al. 2012. Has an angel shown the 79. Lener, M.S. & D.V. Iosifescu. 2015. In pursuit of neuroimag-
way? Etiological and therapeutic implications of the ing biomarkers to guide treatment selection in major
PCP/NMDA model of schizophrenia. Schizophr. Bull. 38: depressive disorder: a review of the literature. Ann. N.Y.
958–966. Acad. Sci. 1344: 50–65.
62. Krystal, J.H. et al. 2003. NMDA receptor antagonist effects, 80. Javitt, D.C. 2015. Neurophysiological models for new
cortical glutamatergic function, and schizophrenia: toward treatment development in schizophrenia: early sensory
a paradigm shift in medication development. Psychophar- approaches. Ann. N.Y. Acad. Sci. 1344: 92–104.
macology 169: 215–233. 81. Abdallah, C.G., L.A. Averill & J.H. Krystal. 2015. Ketamine
63. Javitt, D.C. et al. 2011. Translating glutamate: from patho- as a promising prototype for a new generation of rapid-
physiology to treatment. Sci. Transl. Med. 3: 102mr2. acting antidepressants. Ann. N.Y. Acad. Sci. 1344: 66–77.
64. Olney, J.W., J.W. Newcomer & N.B. Farber. 1999. NMDA 82. Featherstone, R.E. et al. 2015. EEG biomarkers of target
receptor hypofunction model of schizophrenia. J. Psychiatr. engagement, therapeutic effect, and disease process. Ann.
Res. 33: 523–533. N.Y. Acad. Sci. 1344: 12–26.
65. Javitt, D.C. & S.R. Zukin. 1991. Recent advances in the 83. Sivarao, D.V. 2015. The 40-Hz auditory steady-state
phencyclidine model of schizophrenia. Am. J. Psychiatry. response: a selective biomarker for cortical NMDA func-
148: 1301–1308. tion. Ann. N.Y. Acad. Sci. 1344: 27–36.
66. Hall, J. et al. 2015. Genetic risk for schizophrenia: con- 84. Leuchter, A.F. et al. 2015. Rhythms and blues: modulation
vergence on synaptic pathways involved in plasticity. Biol. of oscillatory synchrony and the mechanism of action of
Psychiatry 77: 52–58. antidepressant treatments. Ann. N.Y. Acad. Sci. 1344: 78–
67. Moghaddam, B. & J.H. Krystal. 2012. Capturing the angel 91.
in “angel dust”: twenty years of translational neuroscience 85. MacDonald, J.F., Z. Miljkovic & P. Pennefather. 1987. Use-
studies of NMDA receptor antagonists in animals and dependent block of excitatory amino acid currents in cul-
humans. Schizophr. Bull. 38: 942–949. tured neurons by ketamine. J. Neurophysiol. 58: 251–266.

10 Ann. N.Y. Acad. Sci. 1344 (2015) 1–11 


C 2015 New York Academy of Sciences.
Jones et al. Translational psychiatry

86. Jackson, M.E., H. Homayoun & B. Moghaddam. 2004. containing the NR2B subunit. Characterization in vitro. J.
NMDA receptor hypofunction produces concomitant Pharmacol. Exp. Ther. 283: 1285–1292.
firing rate potentiation and burst activity reduction in the 94. Menniti, F.S. et al. 1998. CP-101606: an NR2B-selective
prefrontal cortex. Proc. Natl. Acad. Sci. USA. 101: 8467– NMDA receptor antagonist. CNS Drug Rev. 4: 307–322.
8472. 95. Bullock, M.R. et al. 1999. An open-label study of CP-101606
87. Kocsis, B. 2012. Differential role of NR2A and NR2B in subjects with a severe traumatic head injury or sponta-
subunits in N-methyl-D-aspartate receptor antagonist- neous intracerebral hemorrhage. Ann. N.Y. Acad. Sci. 890:
induced aberrant cortical gamma oscillations. Biol. Psy- 51–58.
chiatry 71: 987–995. 96. Merchant, R.E. et al. 1999. A double-blind, placebo-
88. Cornwell, B.R. et al. 2012. Synaptic potentiation is critical controlled study of the safety, tolerability and pharmacoki-
for rapid antidepressant response to ketamine in treatment- netics of CP-101606 in patients with a mild or moderate
resistant major depression. Biol. Psychiatry 72: 555–561. traumatic brain injury. Ann. N.Y. Acad. Sci. 890: 42–50.
89. Zarate, C.A., Jr. et al. 2012. Replication of ketamine’s antide- 97. Nutt, J.G. et al. 2008. Effects of a NR2B selective NMDA glu-
pressant efficacy in bipolar depression: a randomized con- tamate antagonist, CP-101606 on dyskinesia and Parkin-
trolled add-on trial. Biol. Psychiatry 71: 939–946. sonism. Mov. Disord. 23: 1860–1866.
90. Rodriguez, C.I. et al. 2013. Randomized controlled 98. Preskorn, S.H. et al. 2008. An innovative design to estab-
crossover trial of ketamine in obsessive-compulsive dis- lish proof of concept of the antidepressant effects of the
order: proof-of-concept. Neuropsychopharmacology 38: NR2B subunit selective N-methyl-D-aspartate antagonist,
2475–2483. CP-101606 in patients with treatment-refractory major
91. Feder, A. et al. 2014. Efficacy of intravenous ketamine for depressive disorder. J. Clin. Psychopharmacol. 28: 631–
treatment of chronic posttraumatic stress disorder: a ran- 637.
domized clinical trial. JAMA Psychiatry 71: 681–688. 99. Sivarao, D.V. et al. 2014. NR2B antagonist CP-101606
92. Paoletti, P., C. Bellone & Q. Zhou. 2013. NMDA receptor abolishes pitch-mediated deviance detection in awake rats.
subunit diversity: impact on receptor properties, synaptic Front. Psychiatry 5: 96.
plasticity and disease. Nat. Rev. Neurosci. 14: 383–400. 100. Matta, J.A. et al. 2013. Developmental origin dictates
93. Fischer, G. et al. 1997. Ro 25–6981, a highly potent interneuron AMPA and NMDA receptor subunit compo-
and selective blocker of N-methyl-D-aspartate receptors sition and plasticity. Nat. Neurosci. 16: 1032–1041.

Ann. N.Y. Acad. Sci. 1344 (2015) 1–11 


C 2015 New York Academy of Sciences. 11

You might also like