Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Psychiatry Research: Neuroimaging 191 (2011) 122–127

Contents lists available at ScienceDirect

Psychiatry Research: Neuroimaging


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / p s yc h r e s n s

The antidepressant effect of ketamine is not associated with changes in occipital


amino acid neurotransmitter content as measured by [1H]-MRS
Gerald W. Valentine a,b, Graeme F. Mason a,d, Rosane Gomez d,e, Madonna Fasula a,b, June Watzl d,
Brian Pittman a,b, John H. Krystal a,b,c, Gerard Sanacora a,b,c,⁎
a
Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
b
Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, United States
c
REAP, Psychiatry Service, VA Connecticut Healthcare System, West Haven, CT, United States
d
Magnetic Resonance Center, Department of Diagnostic Imaging, Yale University School of Medicine, New Haven, CT, United States
e
Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, RS, Brazil

a r t i c l e i n f o a b s t r a c t

Article history: The NMDA receptor antagonist ketamine can induce a rapid improvement in depressive symptoms that often
Received 22 April 2010 endures for days after a single intravenous dose. The pharmacodynamic basis for this effect is poorly
Received in revised form 22 September 2010 understood. Using a proton magnetic resonance spectroscopy ([1H]-MRS) method that previously detected a
Accepted 25 October 2010
normalization of amino acid neurotransmitter (AANt) content after chronic treatment with conventional
antidepressant treatments, we examined whether the acute action of ketamine is associated with alterations
Keywords:
in AANt content as well. Ten subjects with major depressive disorder (MDD) received saline, then ketamine in
Major depressive disorder
NMDA antagonist
a fixed order, one week apart, under single-blind conditions. Each infusion was associated with three [1H]
Magnetic resonance spectroscopy MRS scans (baseline, 3 h and 48 h post-infusion) that measured glutamate, GABA and glutamine within the
Amino acid neurotransmitter occipital cortex. Rating scales were administered before, during and after each infusion. The rapid (1 h) and
sustained (at least 7 days) antidepressant effect we observed after ketamine infusion was not associated with
either baseline measures of, or changes in, occipital AANt content. Dissociative symptoms were not correlated
with changes in depression scores. While our results indicate that changes in occipital AANt content are not a
correlate of ketamine's antidepressant action, this may only apply to the regional and temporal windows of
our MRS measurements.
© 2010 Elsevier Ireland Ltd. All rights reserved.

1. Introduction perception and cognition commonly observed during an infusion of a


subanesthetic dose. The identification of how this relatively brief
Innovation in the clinical therapeutics of major depressive disorder pharmacological blockade of the NMDAR induces a rapid and sustained
(MDD) may be derived from revised pathophysiological models that improvement in depressive symptoms would have important theoret-
incorporate the accumulating evidence for a role of dysregulated ical and practical implications.
glutamatergic neurotransmission. Indeed, both preclinical and clinical Preclinically, NMDAR antagonists induce a variety neurophysio-
studies have demonstrated that a variety of glutamatergic compounds logical effects implicated in antidepressant action including the
have antidepressant effects (Sanacora et al., 2008; Machado-Vieira et al., modulation of monoaminergic neurotransmission (Paul et al., 1992;
2009). In particular, the N-methyl-D-aspartate receptor (NMDAR) Dall'Olio et al., 1999), the enhancement of both hippocampal
antagonist ketamine has emerged as a leading prototype for antide- neurogenesis (Gould et al., 1997; Nacher et al., 2001, 2003; Keilhoff
pressant development because it can be both rapidly acting and et al., 2004) and long-term potentiation (Buck et al., 2006), and
efficacious in ‘treatment-resistant’ MDD subjects (i.e. those without an increased hippocampal brain-derived neurotrophic factor (BDNF)
adequate clinical response to monoaminergic drugs) (Berman et al., levels (Garcia et al., 2008). In addition, recent studies suggest that
2000; Zarate et al., 2006; Mathew et al., 2009). Furthermore, the increased alpha-amino-3-hydroxy-5-methyl-4-isoxazole-propionic
antidepressant effect induced by a single, intravenous administration of acid (AMPA)-receptor-mediated synaptic potentiation and subse-
ketamine often endures for days, well beyond the transient changes in quent activation of the mTOR signaling pathway initiate the
physiological responses that generate ketamine's antidepressant-
like actions in rodent models (Maeng et al., 2008; Li et al., 2010).
⁎ Corresponding author. Abraham Ribicoff Research Facilities, Connecticut Mental
Health Center, 34 Park St., New Haven, CT 06519, United States. Tel.: + 1 203 974 7535;
These studies are in line with a previous report showing that ketamine
fax: + 1 203 974 7662. acutely increases glutamatergic neurotransmission in the prefrontal
E-mail address: gerard.sanacora@yale.edu (G. Sanacora). cortex of rodents (Moghaddam et al., 1997), and collectively, these

0925-4927/$ – see front matter © 2010 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.pscychresns.2010.10.009
G.W. Valentine et al. / Psychiatry Research: Neuroimaging 191 (2011) 122–127 123

findings suggest that an acute increase in non-NMDA mediated scans were performed at the Yale Magnetic Resonance Research Center
glutamate neurotransmission is a critical step in ketamine's mecha- at Yale University School of Medicine.
nism of antidepressant action.
In humans, subanesthetic doses of ketamine appear to acutely 2.2. Infusion schedule and MRS scan time line
increase glutamatergic activity in the prefrontal cortex (Breier et al.,
1997; Rowland et al., 2005), however we are not aware of any studies Following at least a 2-week period free of psychotropic medica-
characterizing changes in glutamatergic activity or AANt levels during tions, subjects received a single 40-minute infusion of 0.9% saline,
the window of symptomatic improvement that opens after the acute followed by racemic ketamine hydrochloride (0.5 mg/kg, Ketalar,
effects of ketamine have dissipated. Using proton magnetic resonance Monarch Pharmaceuticals, Bristol, Tennessee) one week later, in a
spectroscopy (1H-MRS), we sought to determine whether measure- single-blind, non-counter balanced design. This design was chosen to
able alterations in amino acid neurotransmitter (AANt) content are avoid anticipated carryover effects of ketamine that would complicate
associated with ketamine's antidepressant activity. Our spectroscopy the interpretation of data from repeated 1H-MRS measures. Infusions
group has previously identified decreased levels of GABA and were performed by i.v. infusion pump with subjects lying supine in a
increased levels of glutamate in the occipital cortex of adult patients hospital bed located in a private room near the spectroscopy suite.
with unipolar depression as compared to levels in healthy controls Vital signs were measured throughout each infusion session. MRS
(Sanacora et al., 2004). In addition, we have found that chronic measurements were made at six time points (Fig. 1). The first three
treatment with either a chemical antidepressant (Sanacora et al., scans (MRS 1–3) were associated with the saline infusion, and the
2002), or electroconvulsive treatment (ECT) (Sanacora et al., 2003), is second three scans (MRS 4–6) with ketamine. In each group of three
associated with a normalization of GABA levels. Consequently, MRS sessions, the first scan occurred just prior to the infusion, the
changes in AANt levels may represent a neurochemical correlate of second 3 h after infusion start, and the third scan was completed
antidepressant efficacy that extends to NMDAR antagonists. 2 days thereafter.
The primary aims of this study were to replicate the sustained
antidepressant effect in subjects with MDD that has been previously 2.3. 1H-MRS scanning procedure
observed after a single ketamine infusion, and to assess for
correlations between this clinical response and measures of AANt Occipital cortex 1H-MRS acquisitions were performed using a
content in the occipital cortex, the brain region that we have slight modification of the J-edited method as previously outlined
previously shown is associated with abnormal levels in MDD subjects. (Sanacora et al., 2004). Briefly, metabolite levels were measured in the
We predicted that ketamine would reduce scores on depression occipital cortex within a 13.5 cc voxel (3.0 × 1.5 × 3.0 cm) box placed
psychometrics, and that these changes would correlate with an across the midline of the brain, centered 1.5–2.0 cm from the dura.
increase in GABA content and alterations in glutamate and glutamine Cortical GABA content was determined using J-editing, where
content. Contrary to this prediction, we found that the antidepressant subspectra were acquired with 8 K data points over a 410-ms
effect of ketamine present at 3 h and 2 days post-infusion was not acquisition, a 2.5-s repetition time, and a TE of 68 ms on a 4 T Bruker
associated with changes in AANt content. spectrometer at Yale Magnetic Resonance Research Center (MRRC),
averaging data in 20-s blocks for 20 min. Glutamate and glutamine
were measured simultaneously using the unedited subspectra of the
2. Methods J-editing acquisition. For 10 of the studies (five with placebo, five with
ketamine), water spectra were collected for quantification relative to
2.1. Study participants tissue water. ANOVA showed no significant effect on creatine/water
for group, time (i.e., pre-infusion, immediate post-infusion, or post-
Between November 2004 and October 2006, subjects from the infusion two days), or group-by-time. The creatine, therefore, appears
greater New Haven area were recruited through advertisements in local as a stable reference in this case.
newspapers, on the Internet, and by direct contact with physicians. The unedited subspectrum and the J-edited difference spectrum
Men and women (21 to 65 years old) meeting DSM-IV criteria for Major were fitted simultaneously using a basis set of metabolite spectra
Depressive Disorder, as by determined by the Structured Clinical measured with the J-editing acquisition sequence. The metabolites
Interview for DSM-IV (SCID) — Patient Edition (First et al. 2001), who fitted were aspartate, GABA, glutamate, glutamine, NAA, NAAG,
also had a screening Hamilton Depression Rating Scale (HDRS-25, creatine, phosphocreatine, myoinositol, choline, phosphorylcholine,
Mazure et al., 1986) score N 25 were eligible. Subjects with a lifetime glycerophosphorylcholine, and scylloinositol. The subspectrum
history of additional DSM-IV Axis I diagnoses, other than substance obtained without the editing pulse was fitted simultaneously with
dependence in sustained full remission or co-morbid anxiety disorders, the J-edited difference spectrum of GABA (Fig. 2). Because of limited
were excluded. Individuals with stable medical conditions that were resolution in vivo, the results for NAA and NAAG were combined and
without primary CNS effects were included if medication adjustments recorded as NAA, creatine and phosphocreatine were combined and
were not made in the month before study onset. All subjects were free recorded as creatine, and the three choline-containing compounds
from psychotropic medications for at least 2 weeks before, and during were combined and recorded as choline. As part of the implementa-
the 2-week experimental period. Low doses of antihistamine (diphen- tion of the editing method, co-edited macromolecular contamination
hydramine) therapy were allowed for persistent insomnia. Female was evaluated in the GABA spectrum, using two methods: metabolite
subjects were either infertile, or had a negative pregnancy test at nulling (Behar et al., 1994; Rothman et al., 1994; Shen et al., 2004) and
screening, and were using an established birth control method. After frequency switching symmetrically about the coupled macromolec-
receiving a description of the study, including the possibility of ular resonance (Henry et al., 2001), and both cases showed no
experiencing distressing changes in mood, cognition and perception macromolecular contamination of the resonance. Therefore, the basis
of the world and oneself, written informed consent was obtained set for fitting did not include a macromolecular signal. The level of
according to Yale Human Investigation Committee guidelines. One aspartate, though present in the spectra, was poorly determined at the
subject was recruited while an inpatient on the Clinical Neuroscience echo time of 68 ms and was not used.
Research Unit (CNRU) of Connecticut Mental Health Center and Uncertainties of individual measurements were determined using
remained there throughout the study, otherwise, subjects were a Monte-Carlo analysis, in which the least-squares spectral fits were
outpatients that were admitted to the CNRU for one night of treated with random Gaussian noise whose standard deviation was
observation and ratings after each infusion. All infusions and MRS equal to that of the raw data and refitted, using 20 repetitions to
124 G.W. Valentine et al. / Psychiatry Research: Neuroimaging 191 (2011) 122–127

Fig. 1. Study design. MRS (4) was acquired either 7 or 8 days after MRS (1).

estimate the standard deviations of the uncertainty for each after each infusion. Perceptual changes were measured using the
metabolite measure. For each metabolite, a threshold for rejection Clinician Administered Dissociative States Scale (CADSS; Bremner
was set at twice the average noise-based standard deviation of the et al., 1998) at baseline, 20, 60, and 80 min after each infusion. The
respective metabolite. GABA levels whose uncertainties were greater assessments for psychotomimetic effects were performed at baseline,
than 11%, glutamine levels whose uncertainties were greater than 10, 20, 60, 80, 110, and 210 min after each infusion using four items
20%, and glutamate levels whose uncertainties were greater than 16% (unusual thought content, hallucinatory behavior, suspiciousness, and
were not included in subsequent analysis. conceptual disorganization) of the Brief Psychiatric Rating Scale
(BPRS) that constitute a positive symptom subscale (Krystal et al.,
2.4. Psychometric assessments 1994).

The antidepressant effects of ketamine were measured with the 2.5. Statistical analysis
Hamilton Depression Rating Scale (HDRS-25; baseline, 60 min,
180 min, 24 h, 48 h, 72 h, 5 days and 7 days after each infusion) and The primary behavioral outcome variable, HDRS-25 score, was
the Beck Depression Inventory (BDI) (Beck et al., 1961) at the same analyzed by a linear mixed effects model using within subjects factors
time points as HDRS-25. The 25-item HDRS was chosen as the primary of treatment (placebo vs. ketamine) and time (study time points) and
outcome measure secondary to its previously demonstrated sensitiv- random subject effects. The best-fitting variance–covariance structure
ity in measuring the acute effects of ketamine on depressive was selected according to information criteria. BDI scores, HARS
symptoms (Berman et al., 2000). The HDRS was slightly modified to scores, and changes in AANt content were analyzed using similar
account for the compressed temporal context of testing and was models. CADSS scores exhibited floor effects and thus, were analyzed
administered by two un-blinded raters with high inter-rater reliabil- using the nonparametric method for longitudinal data by Brunner
ity. Changes in anxiety were measured using the Hamilton Anxiety using the same factors as described previously (Brunner et al., 2002).
Rating Scale (HARS; Hamilton, 1969) at baseline, 2, 3, 5 and 7 days Potential associations between changes in AANt content and changes
in HDRS scores were assessed by correlation analysis. In secondary
NAA / NAAG analyses, potential associations between changes in AANt content and
Creatine changes in HARS, CADSS and BPRS scores were also evaluated. All
Creatine
analyses were considered significant at the alpha = 0.05 threshold.
Myoinositol Gln
Cho Because HDRS scores were our primary outcome measure, the results
NAA Glu
were not corrected for multiple comparisons. Otherwise, statistical
tests were adjusted with a Bonferroni correction (adj P) based on the
A number of conceptually related tests within each hypothesis (two
Scylloinositol tests for psychometric data, three tests for our primary analysis of
glutamate, GABA and glutamine).
B
3. Results

3.1. Participant characteristics


C
Seventy-three subjects were evaluated after phone screening.
Glu / Gln GABA Eighteen subjects were considered eligible and subsequently con-
D sented. Five subjects were deemed screen failures after providing
consent (one from a medical issue; two secondary to substance abuse
and two due to inability to wash-off medication). Three subjects
4.0 3.5 3.0 2.5 2.0 1.5 1.0
withdrew (one based on failure to return before the testing period;
ppm
one from difficulty with head placement in the scanner, and one from
Fig. 2. Representative MR spectra from a single subject. The data are shown with gray elevated blood pressure during the placebo infusion). Ten subjects
lines, the spectral fits with black lines superimposed on the data, and the residuals are [M/F (4/6); mean age 41.7 years (S.D. 12)] completed the protocol.
shown immediately beneath in black. (A) Pre-treatment subspectrum obtained Four subjects required an antidepressant washout [mean duration
without the editing pulse, yielding glutamate, glutamine and metabolites other than 6 weeks (S.D. 8)], and the rest were un-medicated at the time of
GABA. (B) Post-treatment subspectrum. (C) Pre-treatment J-edited difference
spectrum, yielding GABA levels. (D) Post-treatment J-edited difference spectrum. The
recruitment. The mean duration off antidepressants prior to the study
subspectra were fitted only to just beyond the myoinositol resonances, and the entry was 1.4 years (S.D. 1.78). The mean duration of the current
difference spectra were fitted only around the GABA resonance. episode was 3.3 years (S.D. 4.3) and the mean duration of depressive
G.W. Valentine et al. / Psychiatry Research: Neuroimaging 191 (2011) 122–127 125

illness was 21.2 years (S.D. 17.4). The mean lifetime number of interaction was not observed (F (6,104) = 1.32, P = 0.26), post-hoc
depressive episodes was 3.1 (S.D. 1.7) and the mean number of tests demonstrated that ketamine treatment resulted in lower BPRS
adequate antidepressant treatments (not including adjunctive med- scores at 60 min (P b 0.005), 80 min (P b 0.005), 110 min (P b 0.005)
ications), as determined by self-report, pharmacy records and and 210 min (P b 0.01). However, an analysis of the BPRS positive
prescription histories from prescribing clinicians, was 2.2 (S.D. 1.9). symptom subscale using nonparametric K–W tests on data from the
Eight subjects met criteria for recurrent MDD, and two subjects for a 20 min, 60 min and 80 min time points were not significantly
single episode. Nine subjects met criteria for moderate severity and different between treatments (all P N 0.19).
one for severe with mood congruent psychotic features. Five subjects To assess whether ketamine had significant dissociative effects, an
met SCID criteria for co-morbid anxiety disorders (PTSD and social analysis of total CADSS scores was performed. While there was no
phobia); seven had a lifetime history of anxiety disorder and two overall effect of treatment (ATS = 0.95, num d.f. = 1, P = 0.33), a
subjects met for lifetime substance disorders (one for alcohol abuse significant interaction between treatment and time (ATS = 4.1, num
and one for both alcohol and substance dependence, both in sustained d.f. = 2.58, P = 0.01) was observed, explained by increased dissocia-
full remission for many years). tion at 20 min post-ketamine infusion (ATS = 12.5, num d.f. = 1,
P = 0.0004). Changes in CADDS scores at other time points were not
3.2. Effects of ketamine on depressive and anxiety symptoms significant (all P N 0.15) and the peak change did not correlate with
changes in HDRS scores at any time point.
Baseline HDRS scores measured prior to the start of the saline
(28.4 ± 2.4) and ketamine (26.8 ± 4.2) infusions were not significant- 3.4. Effects of ketamine on vital signs
ly different (P N 0.1). Analysis of HDRS scores over time revealed both a
main effect of treatment (F (1,131) = 11.84, P b 0.0008) and time Analysis of systolic blood pressure detected a main effect of
(F (7,131) = 2.75, P b 0.011). Although the pattern of change in HDRS treatment (F (1,103) = 90.2, P b .0001) but not time (F (1,103) = 1.84,
scores did not significantly differ across time under each treatment P = 0.1). Although a treatment × time interaction was not observed (F
condition (F (7,131) = 1.43, P = 0.2), post-hoc tests revealed that (1,103) = 1.26, P = 0.28), post-hoc tests confirmed that ketamine
significant differences in HDRS scores between conditions persisted significantly increased SBP at 10, 20, 40, 80 and 95 min with a mean
over time (Fig. 3). peak increase of 14.1 mmHg (S.D. 13.4 mmHg) at 40 min. Ketamine was
The overall effect of ketamine on BDI scores was comparable to those associated with a trend toward increased diastolic blood pressure (F
for the HDRS. Mean baseline BDI scores for the saline (26.9 ± 4.2) and (1,103)= 3.6, P = 0.06) and had no effect on heart rate (F (1,100) = 0.9,
ketamine (24.1 ± 5) were similar (P N 0.2). Main effects of treatment (F P = 0.35).
(1,133)= 9.9, adj P = 0.004) and time (F (1,133) = 2.81, adj P = 0.018),
were detected. Although a treatment by time interaction was not 3.5. Effect of ketamine on AANt content in the occipital cortex
observed (F (1,133) = 1.44, adj P = 0.38), post-hoc tests found
significant decreases in BDI scores after ketamine treatment at 60 min The standard deviations of individual metabolite measurements,
(P b 0.001), 210 min (P b 0.05), 24 h (P b 0.01), 48 h (P b 0.05) and 72 h equivalent to the Cramer–Rao lower bounds calculated by the
(P b 0.05) after treatment, but not at 5 or 7 days. standard LCModel program (Provencher, 1993), had the following
Because HARS scores at the start of the placebo (15.7 ± 1.6) mean ± variance: GABA = 0.038 ± 0.044, glutamate = 0.380 ± 0.454,
and ketamine (12.9 ± 2.3) infusions were significantly different glutamine = 0.144 ± 0.156, NAA = 0.033 ± 0.040, myoinosi-
(F (1,98) = 7.43, P b 0.01), an analysis of the absolute percent change tol = 0.063 ± 0.066, creatine = 0.052 ± 0.060, choline = 0.020 ±
from baseline scores was performed and failed to detect an effect of 0.022. As shown in Fig. 4, 1H-MRS measurements of glutamate,
treatment (F (1,76) = 1.8, adj P N 0.2) or time F (4,76) = .25, adj GABA and glutamine content in the occipital cortex were highly
P N 0.8). reproducible across scans. However, a main effect of treatment was
not observed on measures of glutamate (F (1,39) = 2.11, adj P = 0.4),
3.3. Psychomimetic and dissociative effects of ketamine in MDD subjects GABA (F (1,36) = 0.72, adj P N 0.99), or glutamine (F (1,39) = 0.12, adj
P N 0.99). In addition, significant main effects of time (all P N 0.09) or a
Ketamine had significant effects on a range of psychiatric symptom treatment × time interaction (all P N 0.47) were not detected. Second-
constructs as determined by significant main effects of treatment (F ary analyses of ketamine's effect on all other metabolites (see
(1,104) = 13.3, P = 0.0004) and time (F (6,104) = 7.47, P b 0.0001), on Methods) and, of the glutamine/glutamate ratio, were also without
the total score of the 18-item BPRS. Although a treatment by time main effects.
To determine whether the antidepressant effect we observed at
2 days was associated with alterations in AANt content at that time
point, correlation analyses of changes in HDRS scores and changes in
AANt content were performed. Results indicated that changes in
glutamate (r = 0.36, adj P N 0.99), GABA (r = −0.23, adj P N 0.99), and
glutamine (r = 0.77, adj P = 0.21) content were not significantly
correlated with depressive symptoms. Because it was also possible
that the clinical effect at 2 days might be associated with earlier
changes in AANt content (3 h after infusion), secondary correlation
analyses were performed. Again, associations between glutamate
(r = 0.52, adj P = 0.54), GABA (r = 0.17, adj P N 0.99) and glutamine
(r = 0.38, adj P N 0.99) and changes in HDRS scores on day 2 were not
significant.

3.6. Correlation of baseline occipital amino acid content and


clinical response
Fig. 3. Scores on 25-item Hamilton Depression Rating Score (HDRS) over 7 days after
each infusion (n = 10). The difference in baseline scores was not significant. Values are Baseline measures of glutamate, GABA and glutamine were not
represented as mean scores ± s.e.m. * indicates P b 0.05; + P b 0.01; ‡ P b 0.001. correlated with changes in HDRS scores at any time point.
126 G.W. Valentine et al. / Psychiatry Research: Neuroimaging 191 (2011) 122–127

is consistent with the report of negligible effects on BPRS positive


symptom scores in the majority of MDD subjects treated with the
same ketamine dosing parameters used in this study (Mathew et al.,
2009), but contrary to the significant effect reported by (Krystal et al.,
1994). Additional studies, such as those examining the relationship
between pharmacokinetic variability or genetic variation in NMDA
subunits within specific clinical populations, and clinical responses,
are needed to clarify the basis for such contradictory findings. Overall,
ketamine treatment was well tolerated and was without persisting
negative effects. Although ketamine induced a statistically significant
rise in systolic blood pressure, it was without clinical significance in
this study.
The second objective of this study was to characterize the effects of
ketamine on occipital AANt content as measured by 1H-MRS. Because
we have previously shown that a subset of MDD subjects have low
GABA and elevated glutamate content in the occipital cortex, and
furthermore, that GABA alterations appear to normalize after
successful treatment with either chemical antidepressants (Sanacora
et al., 2002) or ECT (Sanacora et al., 2003), we hypothesized that
decreased depression scores (reflecting clinical improvement) fol-
lowing ketamine treatment would correlate negatively with a change
in GABA levels, and positively with a change in glutamate levels. We
did not observe any such change in AANt content following ketamine
treatment, and correlations between clinical response and cortical
AANt content were not observed. Moreover, we found no evidence to
support the possibility that baseline occipital cortex AANt levels can
serve as a predictive biomarker of clinical response to ketamine.
The small sample size of this study limits our ability to draw firm
conclusions regarding the effects of ketamine on occipital GABA and
glutamate content. However, because this study used a very similar
MRS methodology and sample size to those that previously detected
relatively large effect sizes on occipital GABA content after treatment
with SSRIs (d = 1.3) (Sanacora et al., 2002) and ECT (d = 2.6)
(Sanacora et al., 2003), we believe the small effect size at 3 h
(d = 0.21), and 2 days (d = 0.28) post-infusion indicates that keta-
mine does not produce the same magnitude of effect at these early
time points as compared to the effect of chronic treatment with SSRIs
or ECT. However, it is also possible that the absence of experimental
Fig. 4. [1H]-MRS measurements of amino acid neurotransmitter content in the occipital
cortex of MDD subjects before and after ketamine infusion. Values represent mean
support for our hypotheses is due to baseline differences in AANt
values of the ratio of each metabolite to creatine (Cr). Error bars = ±s.e.m. profiles between study populations, as there was no control group to
confirm that either the GABA or glutamate levels were abnormal in
this group of MDD subjects. Although there is no specific reason to
4. Discussion assume this cohort of depressed subjects is markedly different from
those enrolled in the earlier studies, until appropriate controls are
The antidepressant effect of ketamine reported here replicates two utilized, the possibility that our negative findings are secondary to
important features described in the previous controlled studies that investigating a subgroup of depressed subjects that were without
used identical infusion parameters (40-minute infusion of racemic baseline AANt abnormalities cannot be excluded.
ketamine at 0.5 mg/kg) (Berman et al., 2000; Zarate et al., 2006). First, Additional limitations of this study are notable. The 1H-MRS
the improvement in symptoms was rapid, as measured by significant method used in this study can only measure static levels of total tissue
decreases in HDRS and BDI scores 1 h post-infusion. Second, the amino acid content, and may thereby be insensitive to dynamic
antidepressant effect persisted for up to one week as measured by a changes in other aspects of AANt neurotransmission that are induced
significant decrease in HDRS scores at day 7. Also consistent with by ketamine, such as changes in glutamate efflux or turnover.
previous reports, we found that ketamine induced perceptual However, because a previous study using 1H-MRS did detect
alterations during the infusion, as measured by the CADSS, which significant changes in glutamine content in the anterior cingulate
dissipated prior to the emergence of antidepressant effects. The cortex of healthy subjects during ketamine infusion (Rowland et al.,
absence of a correlation between peak perceptual changes and 2005), it is possible that we may have missed significant changes in
changes in HDRS scores in this study suggests that ketamine's AANt content that developed at earlier time points, or within different
antidepressant effect is not dependent on the induction of a brain regions, following ketamine treatment. Future imaging studies
dissociative state, a possibility consistent with the finding that the on the neurobiological mechanisms underlying ketamine's antide-
rapid antidepressant effect of the NR2B subunit-selective receptor pressant effect may benefit from dynamic measures of AANt function
antagonist traxoprodil is also not dependent on a dissociative reaction and the assessment of other brain regions.
in research subjects (Preskorn et al., 2008). Consequently, it is likely In summary, while we replicated the rapid antidepressant-like
that NMDA receptor antagonists constitute a class of drugs whose effects of ketamine, we did not observe ketamine-induced changes in
antidepressant efficacy is independent of their dissociative potential. AANt content in the occipital cortex at several time points associated
In contrast to the significant changes on CADDS scores, we did not with clinical improvement. Further studies on the neurochemical
observe changes in the BPRS positive symptom subscale. This finding basis of NMDAR antagonists' antidepressant activity are warranted
G.W. Valentine et al. / Psychiatry Research: Neuroimaging 191 (2011) 122–127 127

given that the rapid and robust improvement in symptoms they can swimming test and increases BDNF levels in the rat hippocampus. Progress in
Neuropsychopharmacology & Biological Psychiatry 32 (1), 140–144.
induce in ‘treatment-resistant’ depressed subjects may reflect Gould, E., McEwen, B.S., Tanapat, P., Galea, L.A., Fuchs, E., 1997. Neurogenesis in the
unidentified brain mechanisms that contribute to both treatment dentate gyrus of the adult tree shrew is regulated by psychosocial stress and NMDA
resistance and antidepressant efficacy. receptor activation. The Journal of Neuroscience 17 (7), 2492–2498.
Hamilton, M. (Ed.), 1969. Diagnosis and rating of anxiety: Br J Psychiatry Spec Publ.
Henry, P.G., Dautry, C., Hantraye, P., Bloch, G., 2001. Brain GABA editing without
Acknowledgments macromolecule contamination. Magnetic Resonance in Medicine 45 (3), 517–520.
Keilhoff, G., Bernstein, H.G., Becker, A., Grecksch, G., Wolf, G., 2004. Increased neurogenesis
in a rat ketamine model of schizophrenia. Biological Psychiatry 56 (5), 317–322.
We would like to thank Ms. Yael Levin and Ms. Jessica Howell for Krystal, J.H., Karper, L.P., Seibyl, J.P., Freeman, G.K., Delaney, R., Bremner, J.D., Heninger,
their assistance on this project, and Ms. Lisa Roach for her assistance G.R., Bowers Jr., M.B., Charney, D.S., 1994. Subanesthetic effects of the noncom-
with data management and manuscript preparation. This study was petitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual,
cognitive, and neuroendocrine responses. Archives of General Psychiatry 51 (3),
supported by the Yale/Pfizer imaging alliance and from NIH 199–214.
5K02MH076222-05 (GS). Additional funding was provided by R01 Li, N., Lee, B., Liu, R.J., Banasr, M., Dwyer, J.M., Iwata, M., Li, X.Y., Aghajanian, G., Duman,
DA021785 (GM) and K02 AA13430 (GM). Dr. Sanacora has received R.S., 2010. mTOR-dependent synapse formation underlies the rapid antidepressant
effects of NMDA antagonists. Science 329 (5994), 959–964.
consulting fees form AstraZeneca, Bristol-Myers Squibb, Evotec, Eli
Machado-Vieira, R., Manji, H.K., Zarate, C.A., 2009. The role of the tripartite
Lilly & Co., Johnson & Johnson, Roche, Novartis and Sepracor Inc. He glutamatergic synapse in the pathophysiology and therapeutics of mood disorders.
has also received additional grant support from AstraZeneca, Bristol- The Neuroscientist 15 (5), 525–539.
Myers Squibb, Merck & Co., Roche, and Sepracor Inc. In addition, he is Maeng, S., Zarate Jr., C.A., Du, J., Schloesser, R.J., McCammon, J., Chen, G., Manji, H.K.,
2008. Cellular mechanisms underlying the antidepressant effects of ketamine: role
a co-inventor with Dr. Krystal on a filed patent application by Yale of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors. Biolog-
University (PCTWO06108055A1) concerning the use of glutamate ical Psychiatry 63 (4), 349–352.
modulating drugs as antidepressants and anxiolytics. Dr. Krystal has Mathew, S.J., Murrough, J.W., Aan Het Rot, M., Collins, K.A., Reich, D.L., Charney, D.S.,
2009. Riluzole for relapse prevention following intravenous ketamine in treatment-
received consulting and advisory fees from Abbott Laboratories, resistant depression: a pilot randomized, placebo-controlled continuation trial. The
AstraZeneca Pharmaceuticals, Bristol-Myers Squibb, Eli Lilly and Co., International Journal of Neuropsychopharmacology 1–12.
Forest Laboratories, GlaxoSmithKline, Janssen Pharmaceuticals, Loho- Mazure, C., Nelson, J.C., Price, L.H., 1986. Reliability and validity of the symptoms of
major depressive illness. Archives of General Psychiatry 43, 451–456.
cla Research Corporation, Merz Pharmaceuticals, Pfizer Pharmaceu- Moghaddam, B., Adams, B., Verma, A., Daly, D., 1997. Activation of glutamatergic
ticals, SK Holdings Co., Ltd, Takeda Industries, Teva Pharmaceutical neurotransmission by ketamine: a novel step in the pathway from NMDA receptor
Industries, Ltd., and Transcept Pharmaceuticals. He has also received blockade to dopaminergic and cognitive disruptions associated with the prefrontal
cortex. The Journal of Neuroscience 17 (8), 2921–2927.
additional research support from the Janssen Research Foundation Nacher, J., Rosell, D.R., Alonso-Llosa, G., McEwen, B.S., 2001. NMDA receptor antagonist
(provided drug and some study support to the Department of treatment induces a long-lasting increase in the number of proliferating cells, PSA-
Veterans Affairs), and holds Exercisable Warrant Options with NCAM-immunoreactive granule neurons and radial glia in the adult rat dentate
gyrus. The European Journal of Neuroscience 13 (3), 512–520.
Tetragenex Pharmaceuticals (value less than $100). Dr. Krystal's
Nacher, J., Alonso-Llosa, G., Rosell, D.R., McEwen, B.S., 2003. NMDA receptor antagonist
relevant patents and inventions are related to 1) Patent #5,447,948. treatment increases the production of new neurons in the aged rat hippocampus.
September 5, 1995. Dopamine and noradrenergic reuptake inhibitors Neurobiology of Aging 24 (2), 273–284.
in treatment of schizophrenia, and 2) Intranasal Administration of Paul, I.A., Trullas, R., Skolnick, P., Nowak, G., 1992. Down-regulation of cortical beta-
adrenoceptors by chronic treatment with functional NMDA antagonists. Psycho-
Ketamine to Treat Depression (pending). M. Fasula and Drs. Valentine, pharmacology (Berl) 106 (2), 285–287.
Mason, Watzl, Gomez, and Pittman are without potential conflicts of Preskorn, S.H., Baker, B., Kolluri, S., Menniti, F.S., Krams, M., Landen, J.W., 2008. An
interest as related to the content of this report. 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. Journal of Clinical Psycho-
References pharmacology 28 (6), 631–637.
Provencher, S.W., 1993. Estimation of metabolite concentrations from localized in vivo
Beck, A.T., WC, Mendelson, M., Mock, J., Erbaugh, J., 1961. An inventory for measuring proton NMR spectra. Magnetic Resonance in Medicine 30 (6), 672–679.
depression. Archives of General Psychiatry 4, 53–63. Rothman, D., Behar, K., Petroff, O.A., 1994. Improved quantitation of shor TE 1H NMR
Behar, K., Rothman, D.L., Spencer, D.D., Petroff, O.A.C., 1994. Analysis of macromolecule human brain spectra by removal of short T1 macromolecule resonances. Soc Magn
resonances in 1H NMR spectra of human brain. Magnetic Resonance in Medicine Reson, 2nd Meeting, p. 47.
32, 294–302. Rowland, L.M., Bustillo, J.R., Mullins, P.G., Jung, R.E., Lenroot, R., Landgraf, E., Barrow, R.,
Berman, R.M., Cappiello, A., Anand, A., Oren, D.A., Heninger, G.R., Charney, D.S., Krystal, Yeo, R., Lauriello, J., Brooks, W.M., 2005. Effects of ketamine on anterior cingulate
J.H., 2000. Antidepressant effects of ketamine in depressed patients. Biological glutamate metabolism in healthy humans: a 4-T proton MRS study. The American
Psychiatry 47 (4), 351–354. Journal of Psychiatry 162 (2), 394–396.
Breier, A., Malhotra, A.K., Pinals, D.A., Weisenfeld, N.I., Pickar, D., 1997. Association of Sanacora, G., Mason, G.F., Rothman, D.L., Krystal, J.H., 2002. Increased occipital cortex
ketamine-induced psychosis with focal activation of the prefrontal cortex in GABA concentrations in depressed patients after therapy with selective serotonin
healthy volunteers. The American Journal of Psychiatry 154 (6), 805–811. reuptake inhibitors. The American Journal of Psychiatry 159 (4), 663–665.
Bremner, J.D., Krystal, J.H., Putnam, F.W., Southwick, S.M., Marmar, C., Charney, D.S., Sanacora, G., Mason, G.F., Rothman, D.L., Ciarcia, J.J., Ostroff, R.B., Krystal, J.H., 2003.
Mazure, C.M., 1998. Measurement of dissociative states with the clinician- Increased occipital cortex GABA concentrations following electroconvulsive
administered dissociative states scale (CADSS). J Trauma Stress 11 (1), 125–136. therapy in depressed patients. The American Journal of Psychiatry 160 (3), 577–579.
Brunner, E., Domhof, S., Langer, F., 2002. Nonparametric Analysis of Longitudinal Data in Sanacora, G., Gueorguieva, R., Epperson, C.N., Wu, Y.-T., Appel, M., Rothman, D.L.,
Factorial Experiments. John Wiley and Sons, New York, NY. Krystal, J.H., Mason, G.F., 2004. Subtype-specific alterations of GABA and glutamate
Buck, N., Cali, S., Behr, J., 2006. Enhancement of long-term potentiation at CA1- in major depression. Archives of General Psychiatry 61 (7), 705–713.
subiculum synapses in MK-801-treated rats. Neuroscience Letters 392 (1–2), 5–9. Sanacora, G., Zarate, C.A., Krystal, J.H., Manji, H.K., 2008. Targeting the glutamatergic
Dall'Olio, R., Gaggi, R., Bonfante, V., Gandolfi, O., 1999. The non-competitive NMDA system to develop novel, improved therapeutics for mood disorders. Nature
receptor blocker dizocilpine potentiates serotonergic function. Behavioural Reviews. Drug Discovery 7 (5), 426–437.
Pharmacology 10 (1), 63–71. Shen, J., Yang, J., Choi, I.Y., Li, S.S., Chen, Z., 2004. A new strategy for in vivo spectral
First, M., Spitzer, R., Gibbon, M., Williams, J., 2001. Structured clinical interview for editing. Application to GABA editing using selective homonuclear polarization
DSM-IV-TR axis I disorders, research version, patient edition. (SCID-I/P). Biometrics transfer spectroscopy. Journal of Magnetic Resonance 170 (2), 290–298.
Research, New York State Psychiatric Institute, New York. Zarate Jr., C.A., Singh, J.B., Carlson, P.J., Brutsche, N.E., Ameli, R., Luckenbaugh, D.A.,
Garcia, L.S., Comim, C.M., Valvassori, S.S., Reus, G.Z., Barbosa, L.M., Andreazza, A.C., Charney, D.S., Manji, H.K., 2006. A randomized trial of an N-methyl-D-aspartate
Stertz, L., Fries, G.R., Gavioli, E.C., Kapczinski, F., Quevedo, J., 2008. Acute antagonist in treatment-resistant major depression. Archives of General Psychiatry
administration of ketamine induces antidepressant-like effects in the forced 63 (8), 856–864.

You might also like