Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

CNS Drugs

https://doi.org/10.1007/s40263-018-0519-3

REVIEW ARTICLE

Antidepressant Efficacy and Tolerability of Ketamine


and Esketamine: A Critical Review
P. Molero1 • J. A. Ramos-Quiroga2,3 • R. Martin-Santos4 • E. Calvo-Sánchez2,3 •

L. Gutiérrez-Rojas5 • J. J. Meana6,7

Ó Springer International Publishing AG, part of Springer Nature 2018

Abstract Ketamine and its enantiomer S-ketamine (es- metabolism to R-hydroxynorketamine (R-HNK). The
ketamine) are promising candidates to produce a rapid- enantiomer S-ketamine (esketamine) displays approxi-
onset antidepressant effect in treatment-resistant mately fourfold greater affinity for the glutamate NMDA
depression. Ketamine causes continued blockade of the receptor in vitro than R-ketamine. Proof-of-concept
glutamate N-methyl-D-aspartate (NMDA) receptor, single-dose and repeat-dose studies with intravenous
though this might not primarily mediate the antidepres- ketamine show a significant antidepressant and probably
sant effect. Alternative hypotheses include selectivity for antisuicidal effect in the short term, with response rates
the NMDA receptor subtype containing the NMDA over 60% as early as 4.5 h after a single dose, with a
receptor subunit 2B (NR2B), inhibition of the phospho- sustained effect after 24 h, and over 40% after 7 days.
rylation of the eukaryotic elongation factor 2 (eEF2) This response can be further sustained over several
kinase, increased expression of brain-derived neu- weeks with repeated doses (two to three doses per week).
rotrophic factor (BDNF) and tropomyosin receptor Tolerability seems acceptable in the short term, with
kinase B (TrKB), and activation of the mammalian target transient elevation of blood pressure and mild and tran-
of rapamycin (mTOR) signaling pathway, alongside sient dissociative and psychotomimetic effects. Intrana-
other independent actions attributed to the ketamine sal esketamine has shown a comparable antidepressant
effect, which has resulted in the US FDA granting the
& P. Molero
drug a ‘‘breakthrough therapy’’ designation, and theo-
pmolero@unav.es retically it may offer an improved tolerability profile.
However, major concerns remain regarding an effective
1
Department of Psychiatry, Instituto de Investigación Sanitaria protocol to maintain the clinical antidepressant effect of
de Navarra (IdiSNA), University Clinic of Navarra, 31008
Pamplona, Spain
ketamine seen with acute administration and the safety
2
of ketamine and esketamine in the long term, specifically
Department of Psychiatry, Hospital Universitari Vall
d’Hebron, CIBERSAM, Barcelona, Catalonia, Spain
related to potential neurocognitive and urologic toxicity,
3
together with the potential induction of substance use
Department of Psychiatry and Forensic Medicine, Universitat
Autònoma de Barcelona, Barcelona, Catalonia, Spain
disorders. Ketamine and esketamine are not currently
4
approved treatments for depression, but the clinical use
Department of Psychiatry and Psychology, Hospital Clinic,
IDIBAPS, CIBERSAM, Institute of Neuroscience, University
of ketamine is increasing in a variety of practice settings
of Barcelona, Barcelona, Spain internationally.
5
Psychiatry and Neurosciences Research Group (CTS-549),
Institute of Neurosciences, University of Granada, Granada,
Spain
6
Department of Pharmacology, University of the Basque
Country, UPV/EHU, CIBERSAM, Leioa, Bizkaia, Spain
7
Biocruces Health Research Institute, Barakaldo, Bizkaia,
Spain
P. Molero et al.

pharmacological inhibition of reuptake or monoamine


Key Points oxidase activities. The most recent large real-world effec-
tiveness trials indicated that only one-third of patients had
Ketamine and esketamine are promising candidates achieved remission by the end of 12 weeks of antidepres-
to produce a rapid-onset antidepressant effect in sant drug therapy and that the remission rate approached
treatment-resistant depression. 70% after four sequential pharmacological treatments [1].
Therefore, improvement of remission rates and shortening
Antidepressant effects of ketamine may be mediated of the latency period before onset of drug action remain
by a range of mechanisms beyond unspecific N- unmet clinical needs in MDD treatment.
methyl-D-aspartate (NMDA) receptor antagonism, In recent years, ketamine has become a promising can-
including the selective antagonism of NMDA didate to produce a rapid-onset antidepressant effect in
receptor subtypes containing the NMDA receptor MDD, including treatment-resistant depression (TRD). We
subunit 2B (NR2B), inhibition of the provide a critical overview of ketamine clinical pharma-
phosphorylation of the eukaryotic elongation factor 2 cology, the controversy around its mechanism of action and
(eEF2) kinase, increased expression of brain-derived clinical experience in TRD and discuss the clinical
neurotrophic factor (BDNF) and tropomyosin potential of the S-ketamine enantiomer esketamine in
receptor kinase B (TrKB), and activation of the MDD.
mammalian target of rapamycin (mTOR) signaling
pathway, alongside other independent actions
attributed to the ketamine metabolite 2 Clinical Pharmacology of Ketamine
R-hydroxynorketamine (R-HNK). Esketamine has a
higher affinity for the NMDA receptors than Ketamine was developed in the 1960s as a short-acting
ketamine, resulting in the S-enantiomer being about analog of phencyclidine to be used as an anesthetic drug
two to four times as potent as the R-enantiomer. with less emergence delirium [2]. Dissociative anesthesia
In clinical proof-of-concept studies, ketamine and was the term selected for a state where patients appear
esketamine showed a rapid, significant awake, do not respond to sensory inputs but do preserve
antidepressant effect and probable antisuicidal effect spontaneous respiratory activity. Standard initial doses for
that was transient when limited to a single anesthesia are in the ranges of 1–4.5 and 6.5–13 mg/kg via
administration. intravenous and intramuscular routes, respectively, with
further repeated infusions of half the initial dose for
Major concerns remain regarding both the evidence
anesthesia maintenance. Ketamine can also induce a pro-
for efficacious strategies to maintain an
found analgesia and sustained sedation at lower doses.
antidepressant effect and the safety of the long-term
Analgesia and sedation are achieved at intravenous doses
use of ketamine or esketamine.
of 0.2–0.8 mg/kg, and infusion at 0.5 mg/kg/h provides
continuous effect [3]. In addition to anesthesia, analgesia
and sedative actions, ketamine induces central sympath-
omimetic effects leading to tachycardia, hypertension and
palpitations. Higher doses (20 mg/kg) can induce a direct
1 Introduction myocardial depressant effect.
Other neuropsychiatric side effects of ketamine in
The treatment of major depressive disorder (MDD) and anesthesia have been described, although the frequency and
bipolar depression has classically targeted modulation of severity are low [3]. In the treatment of depression, more
monoamine neurotransmission systems. Blockade of detailed side effects associated with ketamine have been
noradrenaline, dopamine and/or serotonin reuptake by described [4] (see Sect. 4.4). The clinical condition termed
acting on their selective transporters (NET, DAT and ‘‘ketamine bladder’’ is described in daily recreational users
SERT, respectively) and inhibition of monoamine break- as an ulcerative cystitis characterized by frequent and
down by the monoamine oxidase enzyme are the most painful urination [5].
established mechanisms of pharmacological action for The psychoactive effects that maintain concerns about
antidepressant drugs. Typically, current antidepressants ketamine use in clinical therapeutics have been evaluated
require several weeks to promote clinical response. This under experimental conditions and are described as a
reflects that the mechanism of therapeutic activity differs, feeling of intoxication, visual, auditory and somatosensory
and probably is more complex, than the simple perceptual distortions, referential thinking disorders and
negative symptoms [6]. Recreational users describe
Ketamine and Esketamine in Depression

dissociative symptoms with hallucinations and distortion of More recently, it was proposed that the antidepressant
time and space that—under long-term use conditions—can activity of ketamine could be related to the R-enantiomer
reproduce persistent schizophrenia-like symptoms [3]. The of the HNK metabolite [18]. According to this study, the
development of a compulsive pattern of ketamine use is a pharmacological activity of (2R,6R)-HNK did not seem to
potential harm reported by recreational users [7]. Tolerance be mediated by NMDA receptor blockade because HNK
develops rapidly to ketamine, leading to the use of an neither binds to nor inhibits NMDA receptor function [18].
increased dose over time [5]. Despite the controversy, these findings need to be con-
Parenteral intravenous or intramuscular administration firmed in the human brain and under clinical conditions.
is the most used route for ketamine because of the limited Indeed, the ratio between the four R- and S-HNK enan-
bioavailability from extensive first-pass metabolism. In tiomers could differ between normal and pathological
oral formulations, increasing the dose is a feasible method human brains and those of rodents. On the other hand,
to surmount the lower bioavailability but does pose the risk evidence that esketamine provides better antidepressant
of tolerability issues and erratic residual effects related to efficacy than racemic ketamine under controlled conditions
active metabolites. This pharmacokinetic profile has led to has not yet been provided. The superiority of esketamine
the development of more convenient routes than parenteral has been assumed from anesthetic and analgesic activity
administration, such as an intranasal formulation (8–45% but needs further demonstration [19]. Moreover, the pos-
bioavailability) [3]. Ketamine is highly soluble in lipids, sibility that different mechanisms of action could be
which determines a distribution volume of 3 L/kg and involved in obtaining anesthetic, analgesic, sedative and
different half-life values between the acute (anesthesia) and antidepressant activity with ketamine remains debatable.
maintained (potential use for depression) conditions of
administration. Thus, intravenous ketamine at a standard
dose provides 5–10 min of surgical anesthesia, whereas 3 Mechanism of Action of Ketamine
repeated sub-anesthetic doses of intravenous ketamine are
being delivered in clinical trials every 2–3 days with pro- The pharmacological and therapeutic mechanism of action
longed antidepressant effects (see Sect. 4.2). The existence of ketamine has been debated [20]. Ketamine binds to the
of active metabolites of ketamine with longer half-lives channel pore on the glutamate NMDA receptor and causes
than the mother compound also contributes to reduced continued blockade. This functional antagonism of NMDA
administration frequency. The dissociative effects of acute receptors expressed by c-aminobutyric acid (GABA)
ketamine administration resolve in approximately inhibitory neurons has been postulated as the mechanism
80–120 min [8], but the analgesic effects last longer [9]. by which ketamine induces psychosis-like effects [21].
With oral administration, ketamine undergoes liver However, other NMDA-receptor antagonist drugs fail to
metabolism mainly by cytochrome P450 (CYP)-3A4, produce sustained antidepressant properties. Alternative
CYP2C9 and CYP2B6 to the active metabolite norke- hypotheses have been proposed, wherein the NMDA
tamine, which retains one-third of the pharmacological receptor subtype expressing the NMDA receptor subunit
activity of ketamine with a plasma half-life of 12 h [10] or 2B (NR2B) seems to be the target for antidepressant
less [11, 12]. Other metabolites of ketamine are dehy- activity [22], which could explain the lack of activity and
dronorketamine, hydroxyketamine and hydroxynorke- neurotoxicity associated with non-selective NMDA recep-
tamine (HNK). The clinical relevance of potential tor antagonists. The NMDA receptor subtype containing
pharmacokinetic interactions between ketamine and other the NR2B subunit is mostly expressed at extrasynaptic sites
antidepressant drugs has not been sufficiently evaluated, and seems to mediate the phosphorylation of the eukaryotic
but available data seem to suggest the absence of important elongation factor 2 (eEF2) kinase and subsequent sup-
interactions [13]. pression of brain-derived neurotrophic factor (BDNF)
Ketamine is a racemic mixture of different R- and S- translation. The increased expression of BDNF and its
enantiomers. S-ketamine (esketamine) displays approxi- receptor tropomyosin receptor kinase B (TrKB) have been
mately fourfold greater affinity for the glutamate N-methyl- related to the antidepressant activity of ketamine. BDNF
D-aspartate (NMDA) receptor in vitro than R-ketamine also activates the mammalian target of rapamycin (mTOR)
[14–16]. In human studies, esketamine has greater anal- signaling pathway, a critical intracellular effect of keta-
gesic and anesthetic activity with less psychotomimetic mine administration [23]. This extrasynaptic NMDA
effects than the racemic mixture and R-isomer [9]. There- receptor-mediated signaling pathway activation clearly
fore, emerging interest in the potential efficacy of ketamine differs from the neurochemical consequences of synaptic
in MDD is focused on esketamine. In contrast, in animal NMDA receptor activation, where the NR2A is the pre-
models of depression, R-ketamine shows higher and dominant subunit [23]. Therefore, the NMDA receptor
longer-lasting antidepressant potency than esketamine [17]. subunit composition and anatomical location could
P. Molero et al.

underlie the singular activity and therapeutic effects of ketamine dose without an augmentation or prolongation
ketamine in MDD. More recently, separate intracellular effect after administration of riluzole. The second,
pathways involving mTOR and ERK have been proposed involving an active placebo (midazolam) in a double-blind,
for R-ketamine and S-ketamine, respectively [24]. Finally, parallel-arm study with a larger sample (73 patients with
the possibility that the antidepressant activity of ketamine TRD), yielded a comparable response rate in terms of
might depend on R-HNK pharmacological activity at an Montgomery-Asberg Depression Rating Scale (MADRS)
NMDA receptor-independent target has been also pre- reduction (64% with ketamine monotherapy vs. 28% with
sented in animal models [18]. However, these exciting midazolam) with evidence of a specific antidepressant
preclinical findings await translation to human conditions effect that differed from the nonspecific effect of an
[25]. Alternative or complementary mechanisms of phar- anesthetic agent such as midazolam [31].
macological action have been suggested, such as an effect In brief, these studies (Table 1) have globally assessed
of ketamine on mu opiate receptors and voltage channels responses to a single dose of intravenous ketamine in 166
[3, 20, 26]. patients with TDR with multiple treatment failures,
including electroconvulsive therapy (ECT). The findings
provide evidence of improvement in depressive symptoms
4 Clinical Experience in Treatment-Resistant within hours, with a response rate [ 60% in the first 4.5
Depression (TRD) and 24 h, and [ 40% after 7 days, with a big effect size in
comparison with placebo (Cohen’s d 1.3–1.7) or active
4.1 Single-Dose, Proof-of-Concept Studies placebo (midazolam, d = 0.8). These figures, though pre-
with Ketamine liminary, contrast with the average effect size of conven-
tional antidepressants (Cohen’s d 0.53–0.81 in patients
The first randomized, double-blinded clinical study with intense symptoms) [32] and their response latency
specifically designed to determine the antidepressant (about 4–7 weeks) [1].
effects of ketamine was published by Berman et al. [27] in
2000. A single dose of intravenous ketamine 0.5 mg/kg 4.2 Studies of Repeated Doses of Intravenous
monotherapy in seven patients with MDD yielded a sig- Ketamine
nificantly greater reduction in depressive symptoms (as
measured with the Hamilton Depression Rating Scale With prior evidence that a different glutamatergic modu-
[HDRS]) than saline treatment after 72 h [27]. This work lator (riluzole) apparently did not confer augmentation or
revitalized the hypotheses of NMDA dysfunction in prolongation effects to the single dose [29, 30], repeated
depression and of the potential therapeutic effects of doses of intravenous ketamine monotherapy 0.5 mg/kg
NMDA antagonism. Six years later, Zarate et al. [28] were tested. An open-label study of three doses per week
conducted a randomized, double-blind, placebo-controlled over 12 days in 24 patients with TDR yielded a stable re-
crossover study to test the same single dose of intravenous sponse rate in terms of MADRS score reduction from day 1
ketamine monotherapy in 18 patients with unipolar TRD. to day 12, with a median time to relapse of 18 days [33].
Their results were similar: [ 70% response rate (change Interestingly, the response after 4 h following the first dose
of C 50% from baseline in HDRS) and almost 30% predicted the response at day 12, with a positive predictive
remission rate to ketamine after 24 h. This response was value (PPV) of 0.89. A similar scheme in a different series
maintained for 1 week in 35% of patients [28]. These two of 12 patients yielded a response rate of 91.6% after six
seminal studies laid the foundations of the clinical evidence doses and a comparable time to relapse (mean time of
supporting the hypothesis of a putative antidepressant 16 days in six subjects, and five subjects with maintained
effect from ketamine. Two important aspects of this response after 28 days of follow-up) [34]. A briefer open-
hypothesis have since been tested with the single-dose label study of two doses per week of intravenous ketamine
methodology. The first was how to prolong the initial, rapid 0.5 mg/kg adjunctive with conventional antidepressants
antidepressant effect without resorting to a repeated dose of until remission, with a total maximum of four doses, in ten
ketamine, given the expected tolerability issues. Two pilot patients with TRD achieved a response rate of 80% and a
studies addressed this matter with the proper methodology: remission rate of 50%, with two patients sustaining the
open-label, single-dose ketamine monotherapy followed by remission status for 4 weeks after the last dose [35]. The
randomization to riluzole (a glutamatergic modulator issue of the optimal dose frequency has been studied in a
already incorporated in clinical practice without major double-blind, randomized, placebo-controlled trial evalu-
tolerability issues) or placebo in patients with TRD ating the efficacy of twice- and thrice-weekly repeated
[29, 30]. Both studies confirmed the rapid, significant doses of intravenous ketamine 0.5 mg/kg over 40 min for
improvement of depressive symptoms following a up to 4 weeks, in a larger sample (67 patients with TRD).
Ketamine and Esketamine in Depression

Table 1 Summary and main results of the seminal, single-dose intravenous ketamine (0.5 mg/kg) studies in unmedicated patients with major
depressive disorder (DSM-IV criteria)
Study Sample Study design Efficacy Effect size (Cohen’s d) Conclusion/most significant results
measure

Berman 7 pts DB ran clinical trial of KET HDRS NR Pts experienced significant improvement of
et al. vs. PL depressive symptoms within 72 h after KET but
[27] not PL infusion
Zarate 18 pts DB ran clinical trial of KET HDRS 1.46 (95% CI 0.91–2.01) KET showed significant improvement of
et al. with vs. PL after 24 h; 0.68 (95% depression vs. PL. 71% response rate and 29%
[28] TRD CI 0.13–1.23) after 1 remission rate the day following KET infusion.
wk 35% of pts maintained response for at least 1 wk
Price et al. 26 pts OL trial of KET plus ran MADRS d = 2.11 (95% CI 24 h after a single infusion, MADRS-Suicidality
[37] with DB continuation trial of 1.25–2.97) item scores reduced by an average 2.08 points;
TRD RIL 100–200 mg/d or PL reductions were sustained for 12 days by
repeated-dose KET
Mathew 26 pts OL trial of KET plus ran MADRS d = 2.11 (95% CI 65% response rate 24 h after a single infusion.
et al. with DB continuation trial of 1.25–2.97) RIL did not prevent relapse
[29] TRD RIL 100–200 mg/d or PL
Ibrahim 42 pts DB, ran, parallel, PC trial of MADRS 1.02 (day 2); 0.46 (day 62% response rate 4–6 h after infusion. Average
et al. with KET vs. KET plus RIL 28) time to relapse: 13.2 days. RIL did not prevent
[30] TRD relapse
Murrough 73 pts Two-site DB ran clinical MADRS 0.81 MADRS score 7.95 points lower with KET vs.
et al. with trial of KET vs. active PL MID (95% CI 3.20–12.71) with response rates
[31] TRD (MID) assigned in 2:1 of 64 and 28%, respectively
ratio
CI confidence interval, DB double-blind, HDRS Hamilton Depression Rating Scale, KET ketamine, MADRS Montgomery-Asberg Depression
Rating Scale, MID midazolam, NR not reported, OL open label, PC placebo-controlled, PL placebo, pt(s) patient(s), ran randomized, RIL
riluzole, wk week

This study yielded response rates of 68.8% for twice- induced reduction of nocturnal wakefulness has been sug-
weekly dosing and 53.8% for thrice-weekly dosing at day gested as a possible mediator of a putative antisuicidal
15, without apparent significant differences in tolerability, effect [39]. Whether ketamine promotes a specific
favoring the twice-weekly regimen to induce a robust and antisuicidal effect and whether this effect is sustained in the
maintained antidepressant effect with less patient and clinic long term are yet to be determined, as are its neurobio-
burden and costs [36]. logical underpinnings [40].

4.3 Possible Antisuicidal Effects with Ketamine 4.4 Tolerability of Ketamine

Aside from its antidepressant effects, intravenous ketamine Current published data on tolerability indicate that intra-
(0.5 mg/kg over 40 min) produced a significant reduction venous ketamine (0.5 mg/kg over 40 min) for the treatment
in suicidality 24 h after the single dose (monotherapy) in of MDD is safe and well tolerated in the short term. A
26 patients with TRD, measured as a reduction in both review of 205 doses in 97 patients [41] yielded an overall
suicidal cognition using the Implicit Association Test and attrition rate of 3.1% and a profile of common, non-vital-
in the suicidality item of MADRS (mean reduction 2.08 risk-associated and reversible effects during the 4 h after
points, with 81% of patients receiving a rating of 0 or 1 infusion (drowsiness, dizziness, poor coordination, blurred
post-infusion). This effect was sustained for 12 days with vision, feeling strange or unreal). About 30% of patients
thrice-weekly repeated doses [37]. A recent midazolam- presented hemodynamic changes (transient increases in
controlled randomized clinical trial of adjunctive ketamine pulse and mean blood pressure, with a mean systolic
in patients with MDD yielded a significant antisuicidal increase of 19.6 ± 12.8 mmHg and a mean diastolic
response rate (proportion of patients experiencing a increase of 13.4 ± 9.8 mmHg). In that review, ketamine
reduction C 50% on the Scale of Suicide Ideation score) of was associated with mild and reversible but significant
55% at 24 h after infusion; this effect was partially inde- dissociative and psychotomimetic effects in the 40-min
pendent of the antidepressant effect [38]. Ketamine- evaluation. Four subjects (1.95%) discontinued infusions
P. Molero et al.

because of adverse effects: two experienced elevated blood [47]. In this trial, reductions in dosing frequency from
pressure, up to 180/115 and 187/91 mmHg, respectively, twice weekly to weekly and to every 2 weeks showed a
with normalization shortly after discontinuation; one sustained response up to 2 months after cessation of dos-
experienced increased anxiety; and another experienced ing. Interestingly, another study found that a single dose of
transient hypotension and bradycardia with venipuncture. intranasal esketamine 84 mg caused no significant
Side effects occurred in the first 2 h after infusion and had impairment in road-driving performance [48]. Further
generally resolved by the 4 and 24-h evaluations. clinical trials to prove this hypothesis of advantages of
A lower infusion rate of the intravenous dose (100 intranasal esketamine over ketamine are needed.
instead of 40 min) showed improved tolerability, without The clinical evidence favoring an antidepressant effect
significant increases on the Brief Psychiatric Rating Scale with intranasal esketamine has led the US FDA to grant
(BPRS) or Young Mania Rating Scale (YMRS), with this drug a ‘‘breakthrough therapy’’ designation [49] (in-
similar efficacy [35]. tended to expedite the development and review of drugs for
serious or life-threatening conditions based on preliminary
4.5 Clinical Studies with Esketamine: The clinical evidence [50]). Approval is expected if this evi-
Intranasal Route dence is supported by completed and recruiting clinical
trials.
Esketamine has a higher affinity for the NMDA receptors A search in ClinicalTrials.gov (on 25 January 2018)
than ketamine, resulting in the S-enantiomer being about identified 14 trials of esketamine in depression (13 intra-
two to four times as potent as the R-enantiomer [14–16]. It nasal, one intravenous), of which six are still recruiting
has been suggested that intravenous ketamine 0.5 mg/kg is (two in TRD, four in MDD), two are active but not
comparable to esketamine 0.4 mg/kg, taking into account recruiting (both in TRD) and six are completed (four in
both the higher potency of esketamine and its greater TRD, two in MDD). Of these, two are focused on the long-
clearance in the absence of R-ketamine [16, 42]. Eske- term safety and efficacy of esketamine in TDR (one is still
tamine monotherapy was tested in 29 patients with TRD active), four are focused on the rapid reduction of suicide
randomly assigned to receive intravenous esketamine 0.20 ideation in adult and pediatric patients with MDD, and one
or 0.40 mg/kg (over 40 min) or placebo [43]. Both doses is focused on the effects on on-road driving of single-dose
yielded similar response rates (67 and 64%, respectively), and repeated administration of esketamine. The results of
comparable to the reported efficacy of intravenous keta- these trials will probably answer some of the unresolved
mine. However, the tolerability profile was better with the queries regarding the efficacy and safety of esketamine in
low dose (0.20 mg/kg), which might represent the optimal depressive disorder.
risk/benefit ratio of intravenous esketamine for TRD. A
retrospective report found that 0.25 mg/kg (over 10 min in
conjunction with conventional antidepressants) yielded a 5 Unresolved Issues and Limitations
significant increase in mild/severe dissociative effects in 23 of the Current Evidence
patients with TRD and four with bipolar depression [44].
Intravenous administration involves inherent difficulties Both the pharmacodynamics and the neurobiological
for both patients and clinicians. Oral administration results underpinnings of the antidepressant effects of ketamine/
in low bioavailability of around 20% [3, 45], whereas the esketamine remain largely unknown. Regarding the phar-
intranasal route offers a more acceptable bioavailability of macodynamics, NMDA receptor antagonism leads to
up to 45% [3, 45]. Results from a pilot trial of a single dose induction of mTORC1 and ERK cellular pathways, con-
of intranasal ketamine 50 mg in conjunction with con- tributing to neuroplasticity and synaptogenesis through
ventional antidepressants in 18 patients with MDD were indirect mechanisms, which may involve the release of
comparable to those with intravenous ketamine with min- BDNF [51], but a definite molecular model that explains
imal adverse effects [46]. Taking this evidence all together, the antidepressant action remains missing. Preliminary
it seems plausible to anticipate that intranasal esketamine evidence from a positron emission tomography (PET)
may offer an efficacy outcome comparable to that with study in 20 patients with TRD showed decreased metabo-
intravenous ketamine, with an improved tolerability pro- lism in the right habenula and the extended medial and
file. Indeed, a recent placebo-controlled randomized trial of orbital prefrontal networks in association with rapid
intranasal esketamine 28, 56 or 84 mg in conjunction with antidepressant response to ketamine [52]. A recent review
oral antidepressant therapy in 67 patients with TRD of the brain effect of ketamine measured in neuroimaging
showed a dose-related, robust, rapid antidepressant effect studies implicated the subgenual anterior cingulate cortex,
with 56 and 84 mg and a safety profile comparable to that among other brain areas, alongside a variety of hypothe-
previously obtained with intravenous ketamine 0.5 mg/kg sized mechanisms such as increased activity in reward
Ketamine and Esketamine in Depression

processing and emotional blunting and decreased ability to symptoms [56]. As suggested by Murrough et al. [31],
self-monitor [53]. Limitations include the high hetero- future studies comparing the antidepressant effect of
geneity in the selected studies (imaging procedure, limited ketamine/esketamine with that of established active com-
sample sizes, sex bias, time to antidepressant effect, use of parators, such as ECT or antidepressant–antipsychotic
different regimens and doses of both racemic ketamine or medication combinations, are also needed. Indeed, a
esketamine). Future research may address these limitations Patient-Centered Outcomes Research Institute (PCORI)-
to improve our knowledge of ketamine’s antidepressant funded comparative effectiveness clinical trial of ECT
mechanisms of action. versus ketamine for patients with TRD is currently ongoing
Future studies should investigate several safety con- (ClinicalTrials.gov, NCT03113968).
cerns. A major concern involves the potential for neuro- Finally, an important aspect to consider in the global
toxicity associated with long-term use of ketamine, leading evaluation of the efficacy and tolerability of ketamine and
to neurocognitive impairment [54]. There is evidence of esketamine as antidepressants is the inherent limitations of
cognitive impairment in recreational ketamine users, albeit a research area that has continued to evolve since the
at much higher doses than used for TRD, and it is critical seminal work by Berman et al. [27] in 2000. Given that
this aspect be tested in long-term, prospective, controlled only 6 of the 14 trials of esketamine in depression listed on
studies [55]. Caution is also needed regarding the potential ClinicalTrials.gov have been completed, the conclusions
for physical side effects as seen in frequent users of keta- presented herein might change considerably in the near
mine, such as ulcerative cystitis with concerns of an future. A considerable concern in the application of the
increased risk of bladder cancer [54]. In addition, two other evidence reviewed herein to actual treatment for patients is
main issues arise. On one hand, further studies are needed the lack of established consensus or clinical guidelines for
to assess the tolerability profile of repeated doses of keta- the use of ketamine in depression, as it remains an off-label
mine or esketamine in the long term, especially regarding indication. However, of note, the American Psychiatric
the systemic and cardiovascular response and dissociative Association (APA) Council of Research Task Force on
and psychotomimetic reactions. On the other hand, whether Novel Biomarkers and Treatments has published a con-
this treatment may induce substance abuse in general sensus statement on this topic with useful and detailed
should be clarified, as should how to control the risk of recommendations on key aspects such as patient selection,
ketamine/esketamine abuse specifically by means of new clinician experience and training, treatment setting, medi-
galenical forms. cation delivery, and follow-up and assessments [57].
The possibility of nasal injuries or olfactory dysfunction
associated with ketamine or esketamine administered via
this route in the long term has yet to be confirmed. Overall, 6 Conclusions
further clinical evidence to prove the putative advantages
of intranasal esketamine over ketamine are needed. Proof-of-concept single-dose and repeat-dose studies with
Further unresolved issues include whether repeated ketamine and esketamine show a significant antidepressant
doses of ketamine/esketamine in the longer term (i.e. and probably antisuicidal effect in the short term, with
9–12 months) maintain a significant antidepressant effect response rates [ 60% as early as 4.5 h after a single dose,
and, if so, whether psychotomimetic and dissociative with a sustained effect after 24 h, and [ 40% after 7 days.
reactions are attenuated or aggravated. Possible strategies This response can be further sustained over several weeks
to prolong the acute antidepressant effect of single-dose with repeated doses (two to three doses per week). Toler-
ketamine (other than repeated doses) are needed after the ability seems acceptable in the short term, with transient
negative trials with riluzole. For instance, evidence elevation of blood pressure and mild and transient disso-
regarding the role of existing psychotropic drugs (e.g., ciative and psychotomimetic effects. Intranasal esketamine
lithium or other mood stabilizers—except lamotrigine, theoretically might offer an improved tolerability profile.
which produced negative results as a post-ketamine However, major concerns remain in terms of establishing
enhancer [29]) and new agents (such as different gluta- an effective protocol to maintain the clinical antidepressant
matergic modulators) is needed. The anticipated response effect of ketamine seen with acute administration while
to this treatment in patients with psychotic symptoms managing long-term safety, specifically regarding the
(present in some forms of severe depression) remains potential for neurocognitive and urologic toxicity and the
unclear because psychotic symptoms are a common induction of substance use disorders.
exclusion criterion in the proof-of-concept clinical trials.
However, a recent small case series provided preliminary
evidence of an acceptable safety and efficacy profile with Author Contributions All authors made a substantial contribution to
the conception, writing and structure of the review article. The first
esketamine in the treatment of TRD with psychotic
P. Molero et al.

draft of the manuscript was prepared by PM (clinical aspects) and depression. Drugs [Internet]. 2017;77:381–401. Available from:
JJM (pharmacological aspects). All authors proofread the manuscript. http://link.springer.com/10.1007/s40265-017-0702-8.
9. Muller J, Pentyala S, Dilger J, Pentyala S. Ketamine enantiomers
Compliance with Ethical Standards in the rapid and sustained antidepressant effects. Ther Adv Psy-
chopharmacol [Internet]. 2016;6:185–92. Available from: http://
Funding No sources of funding were used to conduct this study or journals.sagepub.com/doi/10.1177/2045125316631267.
prepare this manuscript. 10. Niciu MJ, Henter ID, Luckenbaugh DA, Zarate CA, Charney DS.
Glutamate receptor antagonists as fast-acting therapeutic alter-
Conflict of interest Three of the six authors are principal investi- natives for the treatment of depression: ketamine and other
gators or subinvestigators on several Janssen-supported studies of compounds. Annu Rev Pharmacol Toxicol [Internet].
esketamine for depression. PM is supported by Clinica Universidad 2014;54:119–39. Available from: http://www.annualreviews.org/
de Navarra and has received research grants from the Ministry of doi/10.1146/annurev-pharmtox-011613-135950.
Education (Spain), the Government of Navarra (Spain), the Spanish 11. Peltoniemi MA, Hagelberg NM, Olkkola KT, Saari TI. Ketamine:
Foundation of Psychiatry and Mental Health and Astrazeneca. a review of clinical pharmacokinetics and pharmacodynamics in
Without any relevance to this work, PM declares that he is a clinical anesthesia and pain therapy. Clin Pharmacokinet [Internet].
consultant for MedAvante-ProPhase and has received speaker hono- 2016;55:1059–77. Available from: http://link.springer.com/10.
raria from Scienta, AB-Biotics and Janssen. PM is the principal 1007/s40262-016-0383-6.
investigator at Clinica Universidad de Navarra of several studies 12. Fanta S, Kinnunen M, Backman JT, Kalso E. Population phar-
supported by Janssen about the efficacy and safety of esketamine. macokinetics of S-ketamine and norketamine in healthy volun-
JARQ is the principal investigator for Hospital Universitari Vall teers after intravenous and oral dosing. Eur J Clin Pharmacol
d’Hebron in several studies about the efficacy and safety of eske- [Internet]. 2015;71:441–7. Available from: http://link.springer.
tamine supported by Janssen-Cilag. ECS is cooperating as subinves- com/10.1007/s00228-015-1826-y.
tigator at Hospital Universitari Vall d’Hebron in several studies about 13. Andrade C. Ketamine for depression, 5: potential pharmacoki-
the efficacy and safety of esketamine for the treatment of depression netic and pharmacodynamic drug interactions. J Clin Psychiatry
supported by Janssen-Cilag. LGR has been speaker and advisory [Internet]. Physicians Postgraduate Press, Inc.; 2017;78:e858–61.
board member for Janssen, Rovi, Servier, Lundbeck, Otsuka, Pfizer Available from: http://www.psychiatrist.com/JCP/article/Pages/
and Exeltis. RMS and JJM have no conflicts of interest. 2017/v78n07/17f11802.aspx.
14. Moaddel R, Abdrakhmanova G, Kozak J, Jozwiak K, Toll L,
Jimenez L, et al. Sub-anesthetic concentrations of (R,S)-ketamine
metabolites inhibit acetylcholine-evoked currents in a7 nicotinic
References acetylcholine receptors. Eur J Pharmacol [Internet].
2013;698:228–34. Available from: http://linkinghub.elsevier.
1. Rush AJ, Trivedi MH, Wisniewski SR, Nierenberg AA, Stewart com/retrieve/pii/S0014299912009545.
JW, Warden D, et al. Acute and longer-term outcomes in 15. Zeilhofer HU, Swandulla D, Geisslinger G, Brune K. Differential
depressed outpatients requiring one or several treatment steps: a effects of ketamine enantiomers on NMDA receptor currents in
STAR*D report. Am J Psychiatry [Internet]. 2006;163:1905–17. cultured neurons. Eur J Pharmacol [Internet]. 1992;213:155–8.
Available from: http://psychiatryonline.org/doi/abs/10.1176/ajp. Available from: http://www.ncbi.nlm.nih.gov/pubmed/1386806.
2006.163.11.1905. 16. Oye I, Paulsen O, Maurset A. Effects of ketamine on sensory
2. Domino EF. Taming the ketamine tiger. 1965. Anesthesiology perception: evidence for a role of N-methyl-D-aspartate receptors.
[Internet]. 2010;113:678–84. Available from: http:// J Pharmacol Exp Ther [Internet]. 1992;260:1209–13. Available
anesthesiology.pubs.asahq.org/Article.aspx?doi=10.1097/ALN. from: http://www.ncbi.nlm.nih.gov/pubmed/1312163.
0b013e3181ed09a2. 17. Yang C, Shirayama Y, Zhang J, Ren Q, Yao W, Ma M, et al.
3. Li L, Vlisides PE. Ketamine: 50 years of modulating the mind. R-ketamine: a rapid-onset and sustained antidepressant without
Front Hum Neurosci. [Internet]. 2016;10:612. Available from: psychotomimetic side effects. Transl Psychiatry [Internet].
http://journal.frontiersin.org/article/10.3389/fnhum.2016.00612/ 2015;5:e632. Available from: http://www.nature.com/doifinder/
full. 10.1038/tp.2015.136.
4. Short B, Fong J, Galvez V, Shelker W, Loo CK. Side-effects 18. Zanos P, Moaddel R, Morris PJ, Georgiou P, Fischell J, Elmer GI,
associated with ketamine use in depression: a systematic review. et al. NMDAR inhibition-independent antidepressant actions of
Lancet Psychiatry [Internet]. 2018;5:65–78. Available from: ketamine metabolites. Nature [Internet]. 2016;533:481–6. Avail-
http://linkinghub.elsevier.com/retrieve/pii/S2215036617302729. able from: http://www.nature.com/doifinder/10.1038/
5. Singh I, Morgan C, Curran V, Nutt D, Schlag A, McShane R. nature17998.
Ketamine treatment for depression: opportunities for clinical 19. Zanos P, Moaddel R, Morris PJ, Wainer IW, Albuquerque EX,
innovation and ethical foresight. Lancet Psychiatry [Internet]. Thompson SM, et al. Reply to: Antidepressant Actions of Keta-
2017;4:419–26. Available from: http://linkinghub.elsevier.com/ mine Versus Hydroxynorketamine. Biol Psychiatry [Internet].
retrieve/pii/S2215036617301025. 2017;81:e69–71. Available from: http://linkinghub.elsevier.com/
6. Pomarol-Clotet E, Honey GD, Murray GK, Corlett PR, Absalom retrieve/pii/S0006322316328323.
AR, Lee M, et al. Psychological effects of ketamine in healthy 20. Zanos P, Gould TD. Mechanisms of ketamine action as an
volunteers. Phenomenological study. Br J Psychiatry [Internet]. antidepressant. Mol Psychiatry [Internet]. 2018;23:801–11.
2006;189:173–9. Available from: http://bjp.rcpsych.org/cgi/doi/ Available from: http://www.nature.com/doifinder/10.1038/mp.
10.1192/bjp.bp.105.015263. 2017.255.
7. Short B, Fong J, Galvez V, Shelker W, Loo CK. Side-effects 21. Homayoun H, Moghaddam B. NMDA receptor hypofunction
associated with ketamine use in depression: a systematic review. produces opposite effects on prefrontal cortex interneurons and
Lancet Psychiatry [Internet]. 2017; Available from: http:// pyramidal neurons. J Neurosci [Internet]. 2007;27:11496–500.
linkinghub.elsevier.com/retrieve/pii/S2215036617302729. Available from: http://www.jneurosci.org/cgi/doi/10.1523/
8. Lener MS, Kadriu B, Zarate CA. Ketamine and beyond: inves- JNEUROSCI.2213-07.2007.
tigations into the potential of glutamatergic agents to treat
Ketamine and Esketamine in Depression

22. Lang E, Mallien AS, Vasilescu A-N, Hefter D, Luoni A, Riva from: http://linkinghub.elsevier.com/retrieve/pii/
MA, et al. Molecular and cellular dissection of NMDA receptor S0165032713007787.
subtypes as antidepressant targets. Neurosci Biobehav Rev [In- 35. Rasmussen KG, Lineberry TW, Galardy CW, Kung S, Lapid MI,
ternet]. 2017; Available from: http://linkinghub.elsevier.com/ Palmer BA, et al. Serial infusions of low-dose ketamine for major
retrieve/pii/S0149763417303226. depression. J Psychopharmacol [Internet]. 2013;27:444–50.
23. Chan SY, Matthews E, Burnet PWJ. ON or OFF?: modulating the Available from: http://journals.sagepub.com/doi/10.1177/
N-methyl-D-aspartate receptor in major depression. Front Mol 0269881113478283.
Neurosci [Internet]. 2016;9:169. Available from: http://journal. 36. Singh JB, Fedgchin M, Daly EJ, De Boer P, Cooper K, Lim P,
frontiersin.org/article/10.3389/fnmol.2016.00169/full. et al. A double-blind, randomized, placebo-controlled, dose-fre-
24. Yang C, Ren Q, Qu Y, Zhang J-C, Ma M, Dong C, et al. quency study of intravenous ketamine in patients with treatment-
Mechanistic target of rapamycin-independent antidepressant resistant depression. Am J Psychiatry [Internet].
effects of (R)-ketamine in a social defeat stress model. Biol 2016;173:816–26. Available from: http://www.ncbi.nlm.nih.gov/
Psychiatry [Internet]. 2018;83:18–28. Available from: http:// pubmed/27056608.
linkinghub.elsevier.com/retrieve/pii/S0006322317316189. 37. Price RB, Nock MK, Charney DS, Mathew SJ. Effects of intra-
25. Zanos P, Thompson SM, Duman RS, Zarate CA, Gould TD. venous ketamine on explicit and implicit measures of suicidality
Convergent mechanisms underlying rapid antidepressant action. in treatment-resistant depression. Biol Psychiatry [Internet].
CNS Drugs [Internet]. 2018; Available from: http://link.springer. 2009;66:522–6. Available from: http://linkinghub.elsevier.com/
com/10.1007/s40263-018-0492-x. retrieve/pii/S0006322309005198.
26. Williams NR, Schatzberg AF. NMDA antagonist treatment of 38. Grunebaum MF, Galfalvy HC, Choo T-H, Keilp JG, Moitra VK,
depression. Curr Opin Neurobiol [Internet]. 2016;36:112–7. Parris MS, et al. Ketamine for rapid reduction of suicidal
Available from: http://linkinghub.elsevier.com/retrieve/pii/ thoughts in major depression: a midazolam-controlled random-
S0959438815001750. ized clinical trial. Am J Psychiatry [Internet].
27. Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, 2017;appi.ajp.2017.1. Available from: http://www.ncbi.nlm.nih.
Charney DS, et al. Antidepressant effects of ketamine in gov/pubmed/29202655.
depressed patients. Biol Psychiatry [Internet]. 2000;47:351–4. 39. Vande Voort JL, Ballard ED, Luckenbaugh DA, Bernert RA,
Available from: http://www.ncbi.nlm.nih.gov/pubmed/10686270. Richards EM, Niciu MJ, et al. Antisuicidal response following
28. Zarate CA, Singh JB, Carlson PJ, Brutsche NE, Ameli R, ketamine infusion is associated with decreased nighttime wake-
Luckenbaugh DA, et al. A randomized trial of an N-methyl-D- fulness in major depressive disorder and bipolar disorder. J Clin
aspartate antagonist in treatment-resistant major depression. Arch Psychiatry [Internet]. 2016; Available from: http://www.ncbi.
Gen Psychiatry [Internet]. 2006;63:856–64. Available from: nlm.nih.gov/pubmed/27929610.
http://archpsyc.jamanetwork.com/article.aspx?doi=10.1001/ 40. Price RB, Mathew SJ. Does ketamine have anti-suicidal proper-
archpsyc.63.8.856. ties? Current status and future directions. CNS Drugs [Internet].
29. Mathew SJ, Murrough JW, aan het Rot M, Collins KA, Reich DL, 2015;29:181–8. Available from: http://link.springer.com/10.
Charney DS. Riluzole for relapse prevention following intra- 1007/s40263-015-0232-4.
venous ketamine in treatment-resistant depression: a pilot ran- 41. Wan L-B, Levitch CF, Perez AM, Brallier JW, Iosifescu DV,
domized, placebo-controlled continuation trial. Int J Chang LC, et al. Ketamine safety and tolerability in clinical trials
Neuropsychopharmacol [Internet]. 2010;13:71–82. Available for treatment-resistant depression. J Clin Psychiatry [Internet].
from: https://academic.oup.com/ijnp/article-lookup/doi/10.1017/ 2015;76:247–52. Available from: http://www.psychiatrist.com/
S1461145709000169. jcp/article/pages/2015/v76n03/v76n0302.aspx.
30. Ibrahim L, Diazgranados N, Franco-Chaves J, Brutsche N, Henter 42. Persson J, Hasselström J, Maurset A, Oye I, Svensson JO,
ID, Kronstein P, et al. Course of improvement in depressive Almqvist O, et al. Pharmacokinetics and non-analgesic effects of
symptoms to a single intravenous infusion of ketamine vs add-on S- and R-ketamines in healthy volunteers with normal and
riluzole: results from a 4-week, double-blind, placebo-controlled reduced metabolic capacity. Eur J Clin Pharmacol [Internet].
study. Neuropsychopharmacology [Internet]. 2002;57:869–75. Available from: http://www.ncbi.nlm.nih.gov/
2012;37338:1526–33. Available from: http://www.nature.com/ pubmed/11936706.
doifinder/10.1038/npp.2011.338. 43. Singh JB, Fedgchin M, Daly E, Xi L, Melman C, De Bruecker G,
31. Murrough JW, Iosifescu D V, Chang LC, Al Jurdi RK, Green CE, et al. Intravenous esketamine in adult treatment-resistant
Perez AM, et al. Antidepressant efficacy of ketamine in treat- depression: a double-blind, double-randomization, placebo-con-
ment-resistant major depression: a two-site randomized con- trolled study. Biol Psychiatry [Internet]. 2016;80:424–31.
trolled trial. Am J Psychiatry [Internet]. 2013;170:1134–42. Available from: http://linkinghub.elsevier.com/retrieve/pii/
Available from: http://ajp.psychiatryonline.org/doi/10.1176/appi. S0006322315009142.
ajp.2013.13030392. 44. Correia-Melo FS, Argolo FC, Araújo-de-Freitas L, Leal GC,
32. Fournier JC, DeRubeis RJ, Hollon SD, Dimidjian S, Amsterdam Kapczinski F, Lacerda AL, et al. Rapid infusion of esketamine for
JD, Shelton RC, et al. Antidepressant drug effects and depression unipolar and bipolar depression: a retrospective chart review.
severity. JAMA [Internet]. 2010;303:47. Available from: http:// Neuropsychiatr Dis Treat [Internet]. 2017;13:1627–32. Available
www.ncbi.nlm.nih.gov/pubmed/20051569. from: https://www.dovepress.com/rapid-infusion-of-esketamine-
33. Murrough JW, Perez AM, Pillemer S, Stern J, Parides MK, Aan for-unipolar-and-bipolar-depression-a-ret-peer-reviewed-article-
Het Rot M, et al. Rapid and longer-term antidepressant effects of NDT.
repeated ketamine infusions in treatment-resistant major depres- 45. Yanagihara Y, Ohtani M, Kariya S, Uchino K, Hiraishi T,
sion. Biol Psychiatry [Internet]. 2013;74:250–6. Available from: Ashizawa N, et al. Plasma concentration profiles of ketamine and
http://linkinghub.elsevier.com/retrieve/pii/S0006322312005574. norketamine after administration of various ketamine prepara-
34. Shiroma PR, Johns B, Kuskowski M, Wels J, Thuras P, Albott tions to healthy Japanese volunteers. Biopharm Drug Dispos
CS, et al. Augmentation of response and remission to serial [Internet]. John Wiley & Sons, Ltd.; 2003;24:37–43. Available
intravenous subanesthetic ketamine in treatment resistant from: http://doi.wiley.com/10.1002/bdd.336.
depression. J Affect Disord [Internet]. 2014;155:123–9. Available 46. Lapidus KAB, Levitch CF, Perez AM, Brallier JW, Parides MK,
Soleimani L, et al. A randomized controlled trial of intranasal
P. Molero et al.

ketamine in major depressive disorder. Biol Psychiatry [Internet]. 52. Carlson PJ, Diazgranados N, Nugent AC, Ibrahim L, Lucken-
2014;76:970–6. Available from: http://linkinghub.elsevier.com/ baugh DA, Brutsche N, et al. Neural correlates of rapid antide-
retrieve/pii/S0006322314002273. pressant response to ketamine in treatment-resistant unipolar
47. Daly EJ, Singh JB, Fedgchin M, Cooper K, Lim P, Shelton RC, depression: a preliminary positron emission tomography study.
et al. Efficacy and safety of intranasal esketamine adjunctive to Biol Psychiatry [Internet]. 2013;73:1213–21. Available from:
oral antidepressant therapy in treatment-resistant depression. http://linkinghub.elsevier.com/retrieve/pii/S0006322313001534.
JAMA Psychiatry [Internet]. 2017; Available from: http://www. 53. Ionescu DF, Felicione JM, Gosai A, Cusin C, Shin P, Shapero
ncbi.nlm.nih.gov/pubmed/29282469, BG, et al. Ketamine-associated brain changes. Harv Rev Psy-
48. van de Loo AJAE, Bervoets AC, Mooren L, Bouwmeester NH, chiatry [Internet]. 2018;1. Available from: http://www.ncbi.nlm.
Garssen J, Zuiker R, et al. The effects of intranasal esketamine nih.gov/pubmed/29465479.
(84 mg) and oral mirtazapine (30 mg) on on-road driving per- 54. Morgan CJA, Curran HV, Independent scientific committee on
formance: a double-blind, placebo-controlled study. Psy- drugs. Ketamine use: a review. Addiction [Internet].
chopharmacology (Berl). [Internet]. 2017;234:3175–83. 2012;107:27–38. Available from: http://doi.wiley.com/10.1111/j.
Available from: http://link.springer.com/10.1007/s00213-017- 1360-0443.2011.03576.x.
4706-6. 55. Mathew SJ, Shah A, Lapidus K, Clark C, Jarun N, Ostermeyer B,
49. Ionescu DF, Papakostas GI. Experimental medication treatment et al. Ketamine for treatment-resistant unipolar depression. CNS
approaches for depression. Transl Psychiatry [Internet]. Nature Drugs [Internet]. 2012;26:189–204. Available from: http://www.
Publishing Group; 2017;7:e1068. Available from: http://www. ncbi.nlm.nih.gov/pubmed/22303887.
nature.com/doifinder/10.1038/tp.2017.33. 56. Ajub E, Lacerda ALT. Efficacy of esketamine in the treatment of
50. FDA. FDA breakthrough therapy designation [Internet]. [cited depression with psychotic features: a case series. Biol Psychiatry
2018 Jan 1]. Available from: https://www.fda.gov/ [Internet]. 2017; Available from: http://linkinghub.elsevier.com/
RegulatoryInformation/LawsEnforcedbyFDA/ retrieve/pii/S0006322317316815.
SignificantAmendmentstotheFDCAct/FDASIA/ucm341027.htm. 57. Sanacora G, Frye MA, McDonald W, Mathew SJ, Turner MS,
51. Duman RS, Aghajanian GK, Sanacora G, Krystal JH. Synaptic Schatzberg AF, et al. A consensus statement on the use of keta-
plasticity and depression: new insights from stress and rapid- mine in the treatment of mood disorders. JAMA Psychiatry [In-
acting antidepressants. Nat Med [Internet]. 2016;22:238–49. ternet]. 2017;74:399. Available from: http://www.ncbi.nlm.nih.
Available from: http://www.ncbi.nlm.nih.gov/pubmed/26937618. gov/pubmed/28249076.

You might also like