You are on page 1of 13

EXPERT REVIEW OF NEUROTHERAPEUTICS

https://doi.org/10.1080/14737175.2019.1640604

DRUG PROFILE

Esketamine for treatment resistant depression


Jennifer Swainsona,b, Rejish K Thomasa,c, Shaina Archera, Carson Chreneka,b, Mary-Anne MacKaya, Glen Bakera,
Serdar Dursuna,c, Larry J. Klassend, Pratap Chokkaa,c and Michael L Demasa,c
a
Department of Psychiatry, University of Alberta, Edmonton, AB, Canada; bMisericordia Community Hospital, Edmonton, AB, Canada; cGrey Nuns
Community Hospital, Edmonton, AB, Canada; dEden Mental Health Center, Winkler, MB, Canada

ABSTRACT ARTICLE HISTORY


Introduction: Treatment Resistant Depression (TRD) is a common and burdensome condition with poor Received 11 April 2019
outcomes and few treatment options. Esketamine is the S-enantiomer of ketamine and has recently Accepted 3 July 2019
been FDA approved in the United States for treating depression that has failed to respond to trials of KEYWORDS
two or more antidepressants. Esketamine; treatment
Areas covered: This review will briefly discuss current treatment options for TRD, then review esketa- resistant depression;
mine. Relevant literature was identified through online database searches, and clinical trial data were ketamine; glutamate; nmda
provided by Janssen Pharmaceuticals. Pharmacology, including kinetics and dynamics, is discussed, receptor
then clinical data regarding efficacy and safety for esketamine from Phase 2–3 trials are reviewed.
Expert opinion: In the expert opinion, the authors discuss multiple factors including patient, physician,
and social factors that will influence the use of esketamine. While the efficacy of esketamine compared
to off-label use of racemic ketamine remains unclear, both esketamine’s approval for use in TRD and
longer-term safety data may position it preferentially above racemic ketamine, although factors such as
cost and monitoring requirements may limit its use. While questions remain regarding duration and
frequency of treatment, as well as addictive potential, esketamine is a novel treatment option offering
new hope for TRD.

1. Introduction focusing on serotonin, noradrenaline and dopamine systems,


recent interest has focused on targeting the glutamate system.
1.1. The problem of treatment-resistant depression
Glutamatergic agents have offered the promise of a rapid anti-
(TRD)
depressant response via a novel mechanism. One of these agents
Major depressive disorder (MDD) is a common disease with a is esketamine, which is the S-enantiomer of the parent drug
lifetime prevalence of 9.9% [1]. It terms of total disability-adjusted ketamine. This paper will briefly review current TRD treatment
life years (DALYs), MDD represents the most disabling disease in options, then focus on esketamine in terms of its properties and
the Americas [2]. The Sequenced Alternatives to Relieve pharmacology, review evidence for use of esketamine as a treat-
Depression (STAR*D) report revealed that despite many antide- ment for TRD, and discuss future directions, including the
pressant trials, a large portion of patients develop TRD and authors’ opinion as to where this drug may fit in the field of
patients who require more treatment steps to reach remission psychiatry moving forward.
carry a greater burden of depressive illness as well as more psy-
chiatric and medical comorbidities [3]. As each trial of a traditional 2. Overview of current treatment options for TRD
antidepressant takes several weeks to be considered adequate,
2.1. Standard pharmacotherapy
patients suffer the burden of this illness for long periods of time.
Similarly, partial response may lead to polypharmacy with adjunc- Currently, treatment guidelines for depression guide clinicians
tive medications or combination strategies, which carry an to either switch or combine antidepressants, or to use adjunc-
increased risk of side effects. The generally accepted definition of tive medications such as antipsychotics, stimulants, or mood
TRD in the literature is a failure to respond to 2 or more antide- stabilizers when remission is not achieved with an antidepres-
pressants. One problem with this definition, however, is that it fails sant [5,6]. As even these strategies are unsuccessful with a
to address real-world considerations such as patients who partially subset of patients, novel treatment options are desirable.
respond and those who have tried adjunctive strategies [4].
High levels of treatment resistance even with currently avail-
2.2. Neurostimulation
able treatments have led to a call for alternate antidepressant
strategies that could offer a rapid response with a better long- When traditional antidepressants fail to offer relief, neurostimu-
term side effect profile. While traditional antidepressants have lation offers an alternate treatment option. Electroconvulsive
focused on monoamine systems, with various mechanisms therapy (ECT) has long been considered the gold standard

CONTACT Jennifer Swainson jns1@ualberta.ca 3rd floor Cabrini Center, 16940 87 Avenue, Edmonton, AB, Canada
© 2019 Informa UK Limited, trading as Taylor & Francis Group
2 J. SWAINSON ET AL.

period and found a response rate of 70.8% [20]. IV ketamine


Article highlights has subsequently been demonstrated to have a large effect
● TRD carries a large burden of illness.
size and response rate in even severe TRD, both in research
● Currently available treatments have limited efficacy and/or a lengthy and clinical settings [21,22].
time to response. While response rate has been reported as 40–90% in
● The addition of esketamine intranasally to a new antidepressant
increases rates of response and remission in TRD.
patients with TRD [21] and 44% in patients with ultra-resistant
● IN esketamine will offer a new adjunctive treatment option for depression (URD) [23], the time to depressive relapse averages
patients who fail to respond to trials of two or more antidepressants, 18 days after the last infusion [20]. URD has been defined as a
but due to its requirement of a risk evaluation and mitigation
strategy (REMS) for distribution and use, as well as potential limita-
failure of over five antidepressants ± ECT [22]. At this time,
tions in third party coverage, access to this treatment may prove there are no robust data to support ongoing ketamine treat-
limited for patients. ment in terms of long-term safety and efficacy, or to guide
● IN esketamine appears to be safe, and well tolerated in the short
term, but longer-term studies are required, particularly to look at
frequency and duration of treatment. One retrospective case
urinary and cognitive side effects, and to evaluate addictive potential. series of patients with URD who had exhausted other treat-
● IN esketamine may offer patients relief from an otherwise burden- ment options suggested that ongoing maintenance treat-
some illness.
● Other agents targeting the glutamate system may prove novel anti-
ments with IV ketamine may be of benefit in sustaining
depressants/adjuncts and offer promise within the field of psychiatry. response in this population, although potential risks and ben-
● Given the strong data from animal studies suggesting that R-keta- efits should be considered on an individual basis [23].
mine is a good antidepressant with a better adverse effect profile
than esketamine, clinical studies on R-ketamine as an antidepressant
are warranted.
● Future head-to-head clinical comparisons of racemic ketamine, eske- 2.4. Intranasal ketamine
tamine and R-ketamine will be important.
The intranasal (IN) delivery of racemic ketamine was first studied
in an RCT of 18 patients with TRD in 2014. The treatment was
tolerable and demonstrated positive results, with a 44%
response rate [24]. Interestingly, this response rate coincides
treatment for TRD, with an estimated 71% efficacy. While effi-
with that seen in the use of IV ketamine with URD patients in
cacy is promising at 71% [7], use of ECT is limited by the need for
real-world clinical settings [22,24]. A more recent randomized
general anesthesia, hospital resources, the potential for side-
controlled trial of IN racemic ketamine was aborted early due to
effects and stigma [7,8]. Deep Brain Stimulation (DBS) and
extremely poor tolerability of the treatment, suggesting this
Vagal Nerve stimulation (VNS), are other neurostimulation tech-
would not be a viable treatment option moving forward.
niques, both of which carry limitations of requiring surgery and
Patients in this study experienced extreme side effects that
increased latency of response compared to traditional antide-
prevented several patients from continuing to self-administer
pressants [9–11] Repetitive transcranial magnetic stimulation
the nasal sprays of the drug [25]. However, other clinical experi-
(rTMS) is a non-invasive neuromodulation option that does not
ence has found IN ketamine to be well tolerated, suggesting this
require general anesthesia. It does, however, require a patient to
could be a viable treatment option for some patients [26].
attend 20–30 treatment sessions over the course of several
weeks. The frequency of appointments for the treatment itself
may serve as a barrier to the most ill depressed patients. Clinical 2.5. Esketamine
use is also limited by the low availability of treatment [12–15].
In response to the positive results combined with the chal-
lenges of ketamine treatments for TRD, IN esketamine has
2.3. Intravenous ketamine been studied with hopes of offering another treatment option.
Esketamine (the S enantiomer of ketamine) is a NMDA gluta-
Ketamine is considered a novel antidepressant, acting primarily mate receptor antagonist that has been demonstrated to
as a non-selective and non-competitive antagonist at glutamate produce rapid antidepressant and anti-suicidal efficacy that is
N-methyl-D-aspartate (NMDA) receptors by binding to the allos- sustained well beyond its half-life [27]. This review will focus
teric phencyclidine (PCP) site within the ionotropic channel pore on chemical properties of esketamine, clinical data and safety
[16,17]. NMDA receptor antagonism is believed to be the most data, and explore future directions and implications for the
important molecular target underlying the antidepressant effect field of psychiatry.
of ketamine, but this remains under investigation and other
mechanisms of action have been hypothesized [18].
In 2000, Berman et al. reported on the first randomized 3. Esketamine
controlled clinical trial of IV ketamine on nine depressed
3.1. Chemistry
patients after a two-week medication washout period [19].
Patients were randomized to receive two IV infusions over a Esketamine is the S(+) enantiomer of ketamine (Figure 1), an
week of either a saline solution or a 0.5mg/kg ketamine HCl arylcyclohexylamine derivative. Numerous drugs used in psychia-
solution over 40 min. IV ketamine produced a rapid and robust try have one or more chiral centers (center of asymmetry), result-
antidepressant response. The effects were transient, however, ing in the existence of stereoisomers termed enantiomers (non-
with relapse over one to two weeks. There are few multi-dose superimposable mirror images). Though these two enantiomers
studies with IV ketamine. The largest open-label trial treated make up one molecule, each may differ significantly with regard to
24 TRD patients with six IV ketamine doses in a twelve-day pharmacokinetics and pharmacodynamics. Pharmacologic activity
EXPERT REVIEW OF NEUROTHERAPEUTICS 3

markedly increases the antidepressant response rate with keta-


mine, raising questions as to the role of TORC1 in ketamine’s
action [49].
Specific metabolites of ketamine have also been studied for
their antidepressant behavioral effects in animal models [50–53].
The active metabolite of ketamine (2R,6R)-hydroxynorketamine
Figure 1. Molecular structure of ketamine – chiral center marked with *. (HNK) has been reported to exert antidepressant-like effects
without side-effect behaviors in mice and apparently targets
receptor binding sites other than NMDA receptors that currently
may reside primarily in one enantiomer, and the two enantiomers
remain unknown [50]. However, more recent reports have sug-
may influence each other’s pharmacokinetics [28,29].
gested that this metabolite is not necessary for the antidepres-
sant activity (reviewed in [42,51]). Further safety and efficacy
3.2. Pharmacology clinical trials are required to translate these findings to the clinical
condition of depression in humans.
Esketamine is a more potent NMDA receptor antagonist than Additional cellular targets of ketamine include binding to
its enantiomer R-ketamine and it has higher analgesic potency opioid (mu, delta, and kappa) receptors, monoaminergic recep-
than both R-ketamine and the racemic ketamine [29–32]. tors and transporters, and muscarinic and nicotinic cholinergic
There is considerable disagreement in the literature about receptors [52–54]. Interestingly, it has been suggested that
the degree of adverse effects such as dissociation, psychoses, ketamine’s antidepressant effect, but not its dissociative effect,
and cognitive effects with esketamine [29–45]and readers are may depend on activation of the opioid system [53]. However,
referred especially to two comprehensive review articles on based on animal experiments, Zhang and Hashimoto [55] sug-
this matter [30,40]; it should be remembered that some of the gested that the opioid system may not play a role in ketamine’s
studies reported were at anesthetic doses. There are now antidepressant effects. Voltage-gated sodium channels and L
numerous animal studies that report R-ketamine has a rapid and T-type voltage-dependent calcium channels have also
onset of antidepressant effects and a better side effect profile been suggested to be involved in the therapeutic benefit of
than esketamine [35–43]. Large head-to-head clinical compar- ketamine [56]; for a review see [40].
isons of esketamine with R-ketamine and racemic ketamine
have not yet been reported, but several researchers have
commented on the importance of doing so [e.g. 35,40,42] . 3.4. Pharmacokinetics and metabolism
Bioavailability of ketamine, and thus esketamine, differs with the
mode of administration. While intravenous administration pro-
3.3. Pharmacodynamics
vides most predictable dosing with 100% bioavailability, alter-
There are two neuronal NMDA receptor binding site hypoth- nate routes of administration include IN, sublingual, oral,
eses of ketamine. In the disinhibition hypothesis, subanaes- intramuscular and rectal preparations Zhang and Hashimoto
thetic doses of ketamine have a preferential affinity for NMDA [42] have included a table [acknowledged to be from 52] com-
receptors on gamma-aminobutyric acid (GABA)-secreting inhi- paring the pharmacokinetic properties of racemic ketamine in
bitory interneurons, resulting in pyramidal cell disinhibition humans after various routes of administration; bioavailability of
and an increase in glutamate release and extracellular gluta- IN ketamine was reported to vary from 8% to 45%. Intranasally
mate concentration [46]. This presynaptic pyramidal neuron administered esketamine is absorbed quickly through the rich
excitatory effect preferentially activates α-amino-3-hydroxy-5- vascular bed within the IN cavity, reaching maximum plasma
methylisoxazole-4-propionic acid (AMPA) receptors. The block- concentration (Tmax) within 10–14 min of administration [57].
ade of NMDA receptors and enhanced activation of AMPA Mean absolute bioavailability of IN esketamine is equal to that of
receptors then drive neurotropic effects such as the release ketamine [52]. Unlike orally administered esketamine and race-
of brain-derived neurotrophic factor (BDNF) and activation of mic ketamine formulations, IN esketamine is not subject to
downstream trophic signaling cascades. This leads to dendritic extensive hepatic first-pass metabolism [58,59]. Plasma protein
sprouting and synaptogenesis in corticolimbic brain regions binding of IN esketamine is 27% [60] and it is extensively dis-
that seems to be responsible for the antidepressant effect of tributed throughout perfused tissues in the body. Yanagihara et
ketamine [47,48]. al. compared plasma concentrations of ketamine in healthy
In the second hypothesis, ketamine directly inhibits extra- Japanese volunteers after various routes of administration and
synaptic NMDA receptors containing GluN2B subunits on pyrami- found that bioavailability after IN administration (45%) was
dal neurons. Basal cortical activity activates these receptors by higher than after administration by oral tablet, sublingual tablet
extracellular ambient concentrations of glutamate. Direct inhibi- or suppository (20%, 30%, and 30%, respectively) [57].
tion by ketamine is thought to disrupt the basal activation of Esketamine undergoes rapid and extensive metabolism by
pyramidal neurons, resulting in a GluN2B-dependent mechanism hepatic cytochrome P450 (CYP) liver enzyme CYP2B6 and
involving eukaryotic elongation factor (eEF2) dephosphorylation, CYP3A4 systems through N-demethylation to the active metabo-
an increase in BDNF translation and activation of homeostatic lite S-norketamine. S-norketamine is then further metabolized by
mTOR synaptic plasticity, also in a protein-synthesis dependent CYP-dependent pathways to metabolites such as S-5-hydroxynor-
manner [18]. Interestingly, it has recently been reported that rapa- ketamine (HNK) and S-5,6-dehydronorketamine (DHNK) which are
mycin, an immunosuppressant drug and inhibitor of mTORC1, further metabolized by glucuronidation [61]. It has been proposed
4 J. SWAINSON ET AL.

that S-norketamine may also be a useful antidepressant with relationship. It was also found that doses of 56 mg and 84 mg
fewer adverse effects than esketamine (see [43] for review). produced plasma levels equivalent to IV ketamine dosing of eske-
Esketamine has a faster systemic clearance when it is administered tamine 0.2mg/kg, which was previously shown to have antide-
alone as opposed to a racemic mixture [62], with a rapid decline in pressant efficacy [64]. A second phase 2 study (SUI2001) looked at
plasma concentration within the initial 2–4-h period following the reduction of depressive symptoms and suicidality in acutely
administration. depressed patients presenting to the emergency room. The pri-
mary endpoint was mean change from baseline MADRS and this
was significant at 4-h post esketamine dose, and at 2 and 81 days
3.5. Intranasal drug delivery system post esketamine dose, but not at 25 days [65].
IN esketamine HCl (SpravatoTM) is supplied in a single-use nasal
spray device containing 200 µl of vehicle solution (2 sprays),
4.1.2. Phase 3 studies
with each device delivering a total of 28 mg (14 mg of esketa-
The phase 3 efficacy studies for IN esketamine have been
mine base per 100 µl of spray) [63]. The device has an indicator
named the TRANSFORM studies. TRANSFORM 1 investigated
system that informs the user of when the device is full (2 green
fixed doses of 56 mg and 84 mg esketamine versus placebo
dots), one spray remaining (one green dot) and when the
for antidepressant effect [66]. Both treatment arms involved a
device is empty (no green dots, 28 mg delivered). Esketamine
56 mg initial dose on day 1, then on day 4 patients either
is only available through a restricted distribution system where
continued 56 mg or were increased to 84 mg, which was
certified administration sites are equipped to monitor and
continued twice weekly for a month. All three arms included
observe the patient for at least 2 h following dosing for safety
the initiation of a new oral antidepressant two weeks prior to
purposes.
initiation of the study. The study’s primary endpoint of reduc-
Before esketamine administration, the patient is instructed
tion in Montgomery-Asberg Depression Rating Scale (MADRS)
to blow their nose (only before the first device is administered)
did not reach statistical significance (p = 0.088 for the 84 mg
and then assisted to recline their head to 45° (semi-reclined
arm), but results were stated to be clinically significant based
position) during administration to contain the medication
on the change seen in subjects and not compared to placebo,
within the nasal cavity. For each spray, the device tip is inserted
which is a difficult assertion without statistical significance
straight into the nostril until the nose rest touches the skin. The
[67,68]. The study also yielded response rates over 50% for
patient closes the opposite nostril and is instructed to breathe
both treatments arms – which was felt to be clinically signifi-
in while pushing the plunger up until it completely stops.
cant but again did not separate statistically from placebo.
Sniffing gently after administration is encouraged to maintain
Authors noted three times the number of study dropouts
the medication inside the nose. It is recommended to wait 5
with the highest dose, suggesting the dose increase of 56
min between each device (28 mg) application and to keep the
mg to 84 mg from Day 1 to Day 4 may have been too fast
patient in a semi-reclined position for improved tolerability.
for some patients [66]. The sample size (315) may have been
too small considering a higher than predicted placebo
4. Clinical efficacy response, which may have been a result of frequent therapeu-
tics contacts during the study.
4.1. Intranasal esketamine studies
TRANSFORM 2 had a similar study design, but with flexible
Janssen pharmaceuticals have now conducted a series of stu- dosing of IN esketamine. Over the span of a month, two-thirds
dies of esketamine for TRD involving patients with nonpsycho- of the intervention arm reached the maximum dose allowed
tic, unipolar depression with a history of failure of greater than of 84 mg. This study did meet primary outcomes with clinical
two previous antidepressant treatments. Studies will be sum- and statistically significant results. Response rate was 69.3% in
marized below, but for further detail please refer to Tables 1 the treatment arm vs 52.5% in the placebo group. Remission
and 2. rate was 52.0% in the esketamine group, compared to 31.0%
in the placebo group. With a smaller sample size than
4.1.1. Phase 2 studies TRANSFORM 1, it was thought that the change to the flexible
The SYNAPSE 1 study was a fixed dose trial giving patients 14, 28, dosing strategy in this study may have been a factor in leading
56, and 84 mg of IN esketamine or IN saline placebo. Treatments to a statistically significant positive trial [66,69].
were given on days 1 and 4. The study yielded statistically sig- TRANSFORM 3 followed a similar study to TRANSFORM 2,
nificant and clinically meaningful improvement at all doses aside with flexible dosing, however the population was elderly
from 14 mg, and there appeared to be a dose-response patients without neurocognitive issues and a higher level of

Table 1. Completed PHASE II intranasal esketamine trials.


Sample
Study ID Trial Design Comparator Population size Phase Primary outcome and results References
NCT01998958 RCT, 5 arm Fixed Esketamine 14mg MDD, failed ≥2 AD 108 II Change from baseline MADRS at 1 week; [64]
SYNAPSE 1 dosing Acute phase vs. 28mg vs. (at least one in 14mg had a negative result. All other doses
Phase 2 56mg vs. 84mg current episode), yielded positive results with a dose-
vs. placebo IDS-C30 ≥ 34 response relationship
SUI2001 RCT, 2 Arm, fixed Dose 84 mg esketamine MDD with suicidal 66 II Mean change from baseline MADRS at 4 hours [65]
vs placebo ideation post dose on day 1; yielded positive results.
Table 2. Phase III – completed intranasal esketamine trials.
Sample
Study ID Trial Design Comparator Population size Phase Primary outcome and results References
NCT02417064 RCT, 3 arm Fixed dosing Esketamine 56mg + AD vs. Esketamine 84mg + AD vs. MDD, failed 5≥ AD≥1 and no 346 III Change from baseline MADRS at 4 weeks; not [66]
TRANSFORM 1 Acute phase Placebo + AD ECT in current episode IDS- significant.
Phase 3 C30 ≥34 Response and remission rates better with 56
mg and 84 mg, not significant but
described as clinically meaningful
NCT02417064 RCT, 3 arm Fixed dosing Esketamine 56mg + AD vs. Esketamine 84mg + AD vs. MDD, failed 5≥ AD≥1 and no 346 III Change from baseline MADRS at 4 weeks; not [66]
TRANSFORM 1 Acute phase Placebo + AD ECT in current episode IDS- significant.
Phase 3 C30 ≥34 Response and remission rates better with 56
mg and 84 mg, not significant but
described as clinically meaningful
NCT02418585 RCT, 2 arm Flexible dosing Esketamine +AD vs. Placebo +AD MDD, failed 5≥ AD≥1 and no 227 III Change from baseline MADRS at 4 weeks: [69]
TRANSFORM 2 Acute phase ECT in current episode IDS- positive results
Phase 3 C30 ≥34
NCT02422186 RCT, 2 arm Flexible dosing Esketamine +AD vs. Placebo +AD MDD, Elderly MDD failed 8≥ 139 III Change from baseline in MADRS at 4 weeks: [70]
TRANSFORM 3 Acute phase AD≥1 and no ECT in current did not meet statistical significance in
Phase 3 episode No MCI IDS-C30 primary endpoint
≥31
NCT02493868 RCT, 4 arm Optimization phase Maintenance phase: Esketamine weekly dosing vs. MDD, in remission, failed 5≥ 718 III Time to relapse in patients with remission (up [73]
SUSTAIN 1 for all arms: AD + esketamine biweekly dosing vs. switch to placebo AD ≥1 and no ECT in to 104 weeks): remitters and responders on
Phase 3 esketamine Flexible dosing and weekly dosing vs. switch to placebo and current episode, response both weekly and biweekly maintenance
Maintenance phase biweekly dosing to esketamine MADRS ≥28 with esketamine had a lower risk of relapse
at original screening compared to placebo

EXPERT REVIEW OF NEUROTHERAPEUTICS


NCT02497287 Open label Single group, AD + Esketamine 2x/wk for a month, then once MDD, failed ≥2 AD in current 802 III Safety outcomes at the end of 56 weeks: 6.9% [74]
SUSTAIN 2 Flexible dosing in induction weekly for a month and then flexible to weekly or episode, MADRS ≥22 patients had serious TEAE. No clinical
Phase 3 Maintenance phase biweekly for 44 weeks and one month follow up meaningful changes to cognition. No cases
without esketamine of cystitis. No major withdrawal symptoms.

5
6 J. SWAINSON ET AL.

Table 3. Intranasal esketamine trials in progress.


Sample
Study ID Trial Design Comparator Population size Phase Primary outcome
NCT02782104 Open label None MDD, patients enter from 1150 III Safety outcome up to
SUSTAIN-3 Induction: 4 weeks of 2x/week other esketamine ~5 years and 3
Flexible dosing studies months.
Maintenance: 4 weeks of weekly
dosing and then dosed weekly
or biweekly based on CGI-S
weekly dosing

NCT03434041 RCT, 2-arm plus initiation of new Esketamine + AD vs. MDD, failed 5≥ AD≥1 and 234 III Change from baseline
Includes US and Chinese sites oral antidepressant Flexible Placebo + AD no ECT in current MADRS at 4 weeks
dosing Acute phase episode,
MADRS ≥28

NCT02918318 RCT, 4-arm Add on to AD after Esketamine 28mg vs. MDD, AD response in 183 II Change from baseline
Only Japanese sites response to AD documented 56mg vs. 84mg vs. current episode, MADRS MADRS at 4 weeks
Acute phase placebo ≥28 at screening.
TRD not defined
Abbreviations: AD = oral antidepressant; IDS-C30 = Inventory of Depressive Symptoms-Clinician rated, 30-item total score; MADRS = Montgomery–Åsberg
Depression Rating Scale total score; RCT = randomized controlled trial; CGI-S = Clinical Global Impression-Severity scale total score; ECT = electroconvulsive
therapy; MCI = mild cognitive impairment; TEAE = treatment emergent adverse event.
Note: All studies excluded psychosis. AD used in trials were new ADs started 2 weeks prior to the trial.

treatment resistance was allowed. This study included patients flexibly dosed, twice a week for a month. These patients were
who had failed up to eight antidepressant trials compared to then treated with weekly IN esketamine for a month and then
the maximum of five previous antidepressant trials allowed in flexibly dosed to a frequency of weekly or biweekly for 44
TRANSFORM 1 and 2. This trial did not meet the primary weeks, followed by a one month follow up period. The primary
endpoint of change from baseline MADRS at 4 weeks statisti- outcomes were safety outcomes which will be commented on
cally by a narrow margin. Secondary endpoints of response later in this review. In terms of efficacy, 76.5% of patients
and remission rates were 27.0% versus 13.3% and 17.5% ver- remained responders at the end of study. Remission rates
sus 6.7%, respectively, for intervention versus control arms, increased from 47.2% after the 1-month induction phase to
demonstrating clinical significance [70]. 58.2% by the end of this trial [74]. This is an interesting finding
It is notable that patients in the TRANSFORM 1 and 3 in that it suggests the possibility that some individuals may
studies had an average longer period of illness in the current benefit further with longer-term, cumulative treatment.
depressive episode, compared to TRANSFORM 2. This may be Table 3 summarizes studies ongoing. SUSTAIN 3 is a 5
important because only TRANSFORM 2 met its primary end- year study to evaluate long term safety of esketamine.
point, and it has been established that a longer duration of Two other efficacy studies are currently in the recruitment
illness is a poor prognostic factor in the management of phase. NCT02918318 involves Japanese sites and is pro-
unipolar MDD [71,72]. TRANSFORM 1 and 3 may have been posed to be a phase 2 trial with 183 patients with the
treating a more chronically ill population. primary outcome of change in baseline MADRS at 4
Maintenance treatment studies for IN esketamine have weeks. This will build on SYNAPSE 1, which was a phase
been completed and named SUSTAIN 1 and 2. SUSTAIN 1 2 study with a primary outcome with an endpoint of 1
was a large study (n = 708) that followed patients that had week. It is also unique in that the inclusion criteria limit
either responded or remitted with esketamine through 16 the sample to patients that have already had a response
weeks of treatment. Total duration of treatment and examina- to an oral antidepressant in the acute phase and IN eske-
tion went up to 88 weeks. In this study, both weekly and every tamine will be added on for further efficacy. They will be
second week maintenance dosing with esketamine had a studying fixed dosing of 28 mg versus 56 mg versus 84
lower risk of relapse compared to placebos administered mg versus saline placebo in a 4-arm RCT design.
weekly or every second week. Determination of weekly vs NCT03434041 is a phase 3 trial with sites in the United
every second-week maintenance was made on the basis of States and China, with a similar design, sample size and
‘severity of depressive symptoms’, suggesting more ill patients primary outcome measure to TRANSFORM 2.
received weekly maintenance. As this was not randomized, the There currently are no head-to-head comparison stu-
efficacy of weekly vs every second-week maintenance cannot dies of IN racemic ketamine to IN esketamine. To date, a
be compared in this study [73]. protocol for one small non-inferiority study [75] compar-
SUSTAIN 2 was an open-label design following patients on ing single infusions of IV ketamine and IV esketamine have
maintenance therapy that responded to IN esketamine for up been published but results are not yet reported in the
to one year. In this study, 802 patients were followed who peer-reviewed literature. Large comparison studies of mul-
were responders to an initial trial of a new oral antidepressant tiple dosing of IV racemic ketamine versus IV esketamine
two weeks prior to the study initiation and then IN esketamine would be of future interest.
EXPERT REVIEW OF NEUROTHERAPEUTICS 7

5. Safety and tolerability A total of six deaths have been reported as of January 2019
in Janssen’s esketamine clinical program, all of which have
5.1. Common adverse effects
occurred in patients receiving esketamine. Three of these
Seven studies have reported on the safety profile of IN eske- were due to suicide, and they occurred 4, 12, and 20 days
tamine ([64,66,69,70,73,74]) . Common adverse effects have after the last dose of esketamine. Patients who died after 4
been summarized in Table 4. Across studies, these adverse and 12 days had been improving based on their MADRS
effects tend to be of mild to moderate severity, and typically scores, and the patient who died after 20 days was experien-
occur during and immediately after treatment, with resolution cing a worsening of symptoms. Due to lack of consistency, the
also occurring the same day. The longest study of side effects FDA report on esketamine indicates it is difficult to attribute
and tolerability (SUSTAIN 2) followed 802 patients receiving suicides to the drug, however a recent commentary on eske-
esketamine treatment for one year [74] and noted that nearly tamine [77] aptly points out that high relapse rates have been
all patients (90.1%) did experience at least one adverse effect demonstrated on discontinuation of esketamine, and cautions
during the course of the study. that protracted withdrawal from the drug, rather than expo-
These reported acute side effects are similar to those sure to the drug itself, may have played a role. Other deaths
reported for racemic ketamine, but is it is difficult to compare included a motorcycle accident 26 h after receiving esketa-
the frequency of occurrence as reports of ketamine side mine, and the other two were in elderly patients with meta-
effects in the literature tend not to be systematically collected bolic comorbidities, one experiencing sudden death and the
[75]. A recent systematic review [76] looked at the side effects other a myocardial infarction. These were not felt to be related
of ketamine treatments for depression and, similar to esketa- to esketamine as all vitals had been stable during treatments.
mine, found that transient acute side effects were common.
The most common ketamine side effects included headache,
dizziness, dissociation, blurred vision, transient hypertension, 5.3. Specific side effects of concern
and anxiety (the most common psychiatric side effect). The
5.3.1. Hypertension
authors point out that there is currently insufficient data in the
Transient hypertension has been noted frequently, with peak
literature to comment on side effects with repeated dosing or
blood pressure elevation at approximately 40 min following
with long-term treatment with ketamine.
administration, with blood pressure typically returning to
baseline within 2 h. Mean systolic increase was 7–9 mm Hg
and diastolic was 4–6 mm Hg. However, a subset of patients
5.2. Serious adverse events
(8–17%) may have a more clinically significant blood pressure
Across all esketamine studies, serious treatment emergent increase, in the realm of 40 mm Hg systolic and/or 25 mm Hg
adverse effects occurred in less than 5% of the population diastolic [63]
receiving IN esketamine [64,66,69,70,73,74]. Adverse events
reported in patients during treatment with esketamine 5.3.2. Dissociation
included hip fracture, significant blood pressure elevation, As noted in Table 4, rates of dissociation with esketamine
ventricular extrasystoles, hypothermia, lacunar stroke, partial treatment varied from 11.1% to 31.4% across studies, and
seizure, syncope, anxiety, agitation, aggression, sedation, dis- attenuated over time with repeated treatments [74].
orientation, and suicidal ideation. Although these adverse Comparatively, 24% of patients reported feeling ‘strange or
events were reported, it was not clear that any of the adverse unreal’ within 120 min of racemic IV ketamine infusion [78]. A
events were directly related to receiving the drug. Throughout previous study on IN ketamine found mild increases in disso-
one year of maintenance esketamine treatment [74], five ciation and psychotic symptoms, but did not comment on the
patients (0.6% of sample) experienced serious adverse effects frequency of occurrence [24], and a subsequent IN ketamine
that were deemed related to IN esketamine. These included trial was aborted early due to the severity of dissociative side
anxiety, delusional content, delirium, suicidal ideation, and a effects [25]. Authors attributed this difference to variations in
suicide attempt. nasal vasculature and anatomy, differences in the device, for-
mulation, and individual variability. As such, it is difficult to
Table 4. Common side effects of intranasal esketamine treatments from seven determine whether intranasally administered esketamine is
studies. better tolerated than IN ketamine.
Adverse Effect Frequency
Dizziness 20.4–39.0% 5.3.3. Sedation
Nausea 16.0–37.1%
Dissociation 11.1–31.4% Sedation has been noted as a common side effect. In the
Headache 12.5–31.4% general adult population, it tends to peak at 30-min post
Dysgeusia 5.6–31.4% dose and is resolved by 60 min. Latest time to resolve was
Vertigo 11.1–26.1%
Somnolence 11.4–21.1% noted to be 210 min. Interestingly, sedation in the geriatric
Paresthesia 5.6–17.1% population was less severe and resolved sooner. Two subjects
Vomiting 6.6–20.0% in all studies experienced loss of consciousness – in one sub-
Oral hypoesthesia 6.9–13.2%
Increased blood pressure 6.6–12.5% ject this occurred once and they were transferred to the
Anxiety 2.8–17.1% emergency room. The other subject experienced this on five
Hypoesthesia 5.6–13.3% different visits, lasting 15–35 min. The FDA report noted that
8 J. SWAINSON ET AL.

‘experience of previous visits cannot accurately predict future esketamine, when used therapeutically in the treatment of
onset, peak, resolution time, or severity, and this has implica- depression. Unpublished data from Janssen found that poly-
tions for monitoring and labeling’ [79]. drug users equally rated ‘Drug Liking’ when comparing experi-
ences with therapeutic doses of IV ketamine and IN
5.3.4. Driving esketamine, both of which were higher than placebo [88]. It
Given the nature of the common adverse effects, including has been suggested that there is a theoretical risk for patients
transient dissociative symptoms, dizziness, vertigo, and somno- to develop tolerance, dependence, craving, and withdrawal
lence, the impact of esketamine on the ability to safely operate a related to esketamine usage [89]. In one review of several
motor vehicle is an important clinical question. One study found substances of abuse, ketamine was identified as having a
that individuals who had received 84 mg of IN esketamine were relatively high ‘harm score’, similar to substances such as
not impaired on a driving safety test administered 8 h later [80]. alcohol and benzodiazepines [90]. Although it is known that
The product monograph for esketamine advises patients not to naltrexone blocks the antidepressant effects of ketamine, it is
drive until the day following esketamine treatment, after a restful unclear whether this is due to binding to mu opioid receptors
night’s sleep. Given the short half-life of esketamine, further or ketamine-mediated release of endogenous opioids [54],
studies to elucidate the time window to allow safe driving per- which may be important in considering addictive potential.
formance would be helpful. Certainly, as esketamine is used in real-world settings, close
monitoring and evaluation of its addictive potential, and the
5.3.5. Urinary symptoms possibility of harm will be crucial to monitor.
A ketamine-induced uropathy or ulcerative cystitis has been
previously reported in chronic ketamine abusers [81–84]. 5.3.8. Special populations
However, there is no clear evidence that an equivalent risk is Safety and efficacy of IN esketamine have not been studied in
present in patients utilizing therapeutic doses of either race- special populations such as in pregnancy, breastfeeding, or for
mic ketamine or esketamine. In a long-term study, three cases use in patients under the age of 18.
of nephritis occurred during one year of esketamine treat-
ment, but all cases resolved without clinical sequelae and
6. Regulatory affairs
whilst continuing esketamine treatment. There were no
reported cases of interstitial or ulcerative cystitis [74]. Further IN esketamine HCl, marketed by Janssen Pharmaceuticals as
research on the long term risks of esketamine on the lower Spravato, was approved for use in conjunction with an oral
urinary tract will be of importance. antidepressant on 5 March 2019 by FDA of the United States
of America for the treatment of depression resistant to prior
5.3.6. Cognition antidepressant medication treatments. The application for
In one study specifically evaluating cognitive function, healthy approval was given Fast Track and Breakthrough Therapy
participants received esketamine, and cognition was evaluated designations. Ketamine, including its enantiomer esketamine,
using Cogstate®, a computerized test battery evaluating multi- does not have an approved indication for Major Depressive
ple domains including attention, visual and working memory, Disorder or Suicidality at present in other countries. However,
and executive functioning. This study demonstrated acute in Canada, esketamine is being reviewed under the Priority
transient impairment in cognition. Significant impairment Review Policy of Health Canada, which fast tracks the review of
was noted 40 min after administration, but completely dissi- the compound by the agency. Janssen also has a current
pated by 2-h posttreatment [85]. Esketamine studies that have Marketing Authorization Application for the European Union
evaluated cognitive measures as part of safety and tolerability for esketamine.
have found that cognition (including measures of spatial
memory, executive function, and processing speed) has either
7. Conclusion
remained stable or improved after treatment for up to 1 year
[76,77]. However, cognition remains an area worth further There is a need in psychiatry for novel treatment options for
study and monitoring, as cognitive impairment has been TRD. Some of the current options include neurostimulation
noted in chronic ketamine abusers [86]. When used therapeu- and adjunct medications, both of which can carry burdensome
tically to treat depression, it has been suggested ketamine adverse effects. New antidepressants targeting the glutamate
actually improves cognition, likely owing to the improvement system offer promise as rapid antidepressants with a novel
of depressive symptoms with treatment [87]. The long-term mechanism. The entrance of esketamine into the field of
cognitive impacts of racemic ketamine, as well as esketamine, psychiatry comes on the heels of early excitement from initial
remain unclear. studies on racemic ketamine, which have been short term in
nature. It has been clear that ketamine has antidepressant
5.3.7. Abuse/addictive potential properties, but long-term safety and dosing remain unclear.
Serious concerns relate to the addictive potential of esketa- Esketamine data have filled gaps missing in the ketamine
mine. Ketamine itself is a well-known substance of abuse, literature. To date, 3/5 studies on IN esketamine met all end-
though the recent FDA report on esketamine cites ketamine points with positive results, while the other two failed to meet
abuse as being relatively uncommon with a lifetime preva- endpoint but showed a favorable trend towards benefit. The
lence of 1.2% [79]. To date, there are no published data on the failure to meet endpoint in these studies may have been a
risk of abuse or misuse for either racemic ketamine or reflection of study design (fixed vs flexible dosing), power, and
EXPERT REVIEW OF NEUROTHERAPEUTICS 9

high placebo response of an IN treatment. Similarly, it should account factors such as individual response, preference, toler-
be noted that esketamine studies have generally been con- ability, addictive potential, and other treatment options.
ducted on more severely depressed patients than would be
typical in phase 3 studies for FDA approved antidepressants
8.2. Practitioner considerations
and adjunctive treatment medications for depression [79]
Tolerability and safety data appear promising, with data now The role of glutamatergic agents to treat depression has heralded
extending up to a year, which are not currently available for very exciting treatment options for clinicians, but there exists
ketamine itself. To date, data regarding potential concerns controversy, even among psychiatrists regarding the use of keta-
with urinary and cognitive toxicity are reassuring. As esketa- mine in depression. The 2017 UK consensus statement on keta-
mine is used in the real world, more data will be required mine for depression suggests that it be used only as part of
regarding addictive potential and real-world efficacy, as well research studies, but that use outside formal studies should be
as the optimal duration of treatment. supported by a second psychiatrist opinion, include informed
consent, and incorporate a collection of data from a mood mon-
itoring program [91]. The American Psychiatric Association’s posi-
8. Expert opinion tion statement on the use of ketamine points to lack of long-term
data and provides many judicious suggestions for patient mon-
Esketamine provides an exciting and novel treatment option for
itoring [92]. It would be wise for any clinician prescribing esketa-
TRD. Given that trials were conducted in severely depressed
mine to also review and implement the suggestions detailed there
patients, esketamine may be an alternative or even provide
[92]. The Canadian Mood and Anxiety Disorder Network
greater efficacy than currently available adjunctive medications.
(CANMAT) has placed IV ketamine as a third line adjunctive treat-
The transient nature of esketamine side effects may confer an
ment for depression in its 2018 Bipolar Disorder Treatment guide-
advantage over the metabolic and extrapyramidal side effects
lines [93], but the 2016 Depression treatment guidelines still list
that may be associated with atypical antipsychotics commonly
ketamine as an ‘experimental’ treatment [6], which has limited its
used as treatment adjuncts. This commentary section will
use. Regulatory approval of esketamine for TRD with failure of two
address esketamine, its use, and role from the perspective of
or more antidepressants will support clinicians in offering a rapid-
the patient, practitioner, society and long -term implications.
acting antidepressant to less treatment-resistant populations, and
may decrease the overall burden of illness if a glutamatergic agent
is used earlier in the course of illness. Efficacy and tolerability of
8.1. Patient considerations
esketamine compared to racemic ketamine remains uncertain. To
Esketamine provides TRD patients an option that has been date, there is discrepancy across the literature as to which enan-
FDA approved and eliminates the uncertainty associated with tiomer (R or S) offers the best antidepressant effect and most
racemic ketamine used in some clinical settings. Although tolerability.
access and cost may play a role for some patients, the efficacy There has been a spectrum of overpromising efficacy and at
data from esketamine trials provide hope for the difficult to times underdelivering in some of the clinical trials. Nevertheless,
treat patients with depression. Stigma associated with neuro- a body of literature now exists to support ongoing use of eske-
stimulation treatments may lead to patient preference for tamine, whereas ketamine studies have tended to focus only on
esketamine as an alternative. single infusions or a series of 6–8 treatments. ‘Maintenance’
This treatment is unique in that it causes varying acute psy- treatment data for racemic ketamine is limited to trials lasting
chological side effects, so patients must be given proper psy- only several weeks, or retrospective case reports. The esketamine
choeducation prior to treatment and supportive care during trials met primary endpoint objectives in three trials, but failed to
treatment. While ketamine has been used to treat depression achieve primary endpoint in two others. There may be several
in a number of settings, by a number of medical specialties, our reasons for this. First, in the TRANSFORM 1 acute dose trial, the 84
clinical experience is that patients both require and benefit from mg group was analyzed before the 56 mg dose, and it failed to
mental health support during ketamine treatments, and the separate from placebo. As such, the 56 mg dose could not
same standard of care would be advised for esketamine treat- officially be analyzed, but did separate from placebo in ‘explora-
ments, so as to minimize any risk of psychological harm. tory analysis’. It was noted that the patients receiving 84 mg were
Esketamine should not be positioned as a panacea for likely more ill than the 56 mg group. Similarly, the TRANSFORM 3
resistant depression, but as potentially a very good option study looking at the age 65 plus population also failed to meet its
for short-term treatment with favorable outcomes for up to primary endpoint. Nevertheless, it should be noted that the
one year. Short-term side effects tend to be of a transient enrolled patients for these studies were severely ill and it is
nature, likely improving acceptability of side effect burden encouraging that there is positive short term and 1-year data
for patients compared to other adjunctive medication options. with esketamine for this population.
Clinically the adverse effects of esketamine should be weighed At this time, future comparative trials with other adjunct treat-
against alleviating of the burden of depression. While a pro- ments for depression such as atypical antipsychotics are needed.
mising agent, esketamine dosing and side effects in the long Finally, comparative trials with different formulations, and delivery
term remain unknown. Beyond one year, ongoing use of systems with esketamine and racemic ketamine could provide
esketamine should be considered on a case by case basis, some clarity in achieving much more precision in patient-focused
considering potential risks and benefits of both continuing treatments. Further, Phase 4 trials should be conducted to gather
and stopping the treatment. This evaluation should take into longer-term data to address safety, efficacy, tolerability, and
10 J. SWAINSON ET AL.

addictive potential. Physicians prescribing esketamine must dili- oral agent that acts on the glutamate system. Future studies
gently monitor patients for adverse effects and patient response to may bring more understanding as to mechanisms of action of
facilitate weighing the risk/benefit of treatment for each patient. ketamine and esketamine and can guide targeted drug
development.
8.3. Societal considerations
Funding
Although the evidence for the role of ketamine for treating
depression is growing, there is still considerable misunderstanding This paper was not funded.
from a societal perspective on its therapeutic role. Direction of
acceptability seems likely to be dictated by societal cues. There has
been extensive media and social media coverage of ketamine/
Declaration of interest
esketamine due to their novel antidepressant nature offering J Swainson has received speaking honoraria from Otsuka and Lundbeck
rapid relief of suffering, combined with controversy as substances and has acted on advisory boards for Otsuka, Lundbeck, and Janssen. R
Thomas, S Dursun and M Demas have acted on advisory boards for
of potential abuse. The FDA has taken a cautionary approach and
Janssen. C Chrenek has received speaking honoraria from Otsuka and
has made esketamine available through its restricted distribution has acted on advisory boards for Otsuka, Lundbeck, and Janssen. LJ
system, Risk Evaluation Management System, ‘REMS’. The FDA has Klassen has received research grants and speaker’s honoraria from and
also stated that esketamine ‘must be administered in a certified has acted on an advisory board for Shire. They have also received speak-
medical office where the health-care provider can monitor the er’s honoraria from and acted on advisory boards for Janssen, Lundbeck,
Otsuka, Pfizer, Purdue, and Sunovion. They have also acted on an advisory
patient’. This restricted distribution, while put in place as a societal
board for Allergan and received speaker’s honoraria from CPA and The
protection, may limit practitioners from choosing this as a ther- Canadian ADHD Resource Alliance. P Chokka received speaker’s honoraria
apeutic option due to practical obstacles inability to administer and acted on advisory boards for Allergan, Lundbeck, Janssen, Purdue,
treatments in the health-care setting. The REMS for esketamine Sunovion, and Shire. They have also received research grants from
may also create a new standard of care, frowning heavily on Lundbeck, Janssen, and Shire. The authors have no other relevant affilia-
tions or financial involvement with any organization or entity with a
prescription of take-home ketamine in any form. While concerns
financial interest in or conflict with the subject matter or materials dis-
for diversion and abuse are not to be taken lightly, the addictive cussed in this manuscript apart from those disclosed.
potential of ketamine or esketamine could be considered similar
to other commonly used psychiatric medications such as stimu-
lants or benzodiazepines. Reviewer disclosures
A peer reviewer on this manuscript in an inventor holding a patent of R-
8.4. Long-term view
ketamine in the treatment of depression. Peer reviewers on this manu-
The FDA approval of esketamine brings forth a novel treat- script have no other relevant financial relationships or otherwise to
disclose.
ment option for treatment-resistant depression. Due to multi-
ple influences outlined previously involving patient, physician
and societal factors, there will be many issues at play in References
determining where IN esketamine itself will eventually settle
Papers of special note have been highlighted as either of interest (•) or of
within the psychiatric practice. Various formulations of racemic
considerable interest (••) to readers.
ketamine have been used off label for some time to treat 1. Patten SB, Williams JVA, Lavorato DH, et al. Descriptive epidemiol-
depression, but this treatment has been limited in availability ogy of major depressive disorder in Canada in 2012. Can J
within public health-care systems, or limited to those who can Psychiatry. 2015;60:23–30.
cover the cost in private settings. With esketamine approved 2. Ustün TB, Ayuso-Mateos JL, Chatterji S, et al. Global burden of
depressive disorders in the year 2000. Br J Psychiatry.
for the TRD indication, it seems likely that more third party
2004;184:386–392.
payers may cover the cost, though it will prove more costly 3. Rush AJ, Trivedi MH, Wisniewski SR, et al. Acute and longer-term
than ketamine itself. outcomes in depressed outpatients requiring one or several treat-
Interestingly, while it is well known that racemic ketamine has ment steps: a STAR*D report. Am J Psychiatry. 2006;163:1905–1917.
antidepressant properties, IN esketamine has been studied by 4. McIntyre RS, Filteau M-J, Martin L, et al. Treatment-resistant depres-
Janssen only as an adjunct, accompanied by a change in anti- sion: definitions, review of the evidence, and algorithmic approach.
J Affect Disord. 2014;156:1–7.
depressant. Antidepressants themselves carry long-term side 5. Cleare A, Pariante CM, Young AH, et al. Evidence-based guidelines
effects including weight gain and sexual dysfunction. As esketa- for treating depressive disorders with antidepressants: A revision of
mine begins to be prescribed by psychiatrists, it seems likely that the 2008 British Association for Psychopharmacology guidelines. J
clinical experience may be gained using esketamine as mono- Psychopharmacol (Oxford). 2015;29:459–525.
6. Kennedy SH, Lam RW, McIntyre RS, et al. Canadian Network for
therapy, particularly in the subset of patients who have tried
Mood and Anxiety Treatments (CANMAT) 2016 clinical guidelines
nearly every class of antidepressant with no or limited benefit. for the management of adults with major depressive disorder:
Further research including Phase IV clinical trials should be com- section 3. pharmacological treatments. Can J Psychiatry.
pleted for determination of long-term effects of IN esketamine, 2016;61:540–560.
which can lead future therapeutic directions. 7. Folkerts HW, Michael N, Tölle R, et al. Electroconvulsive therapy vs.
paroxetine in treatment-resistant depression — a randomized
The first approval of a rapid-acting antidepressant is an
study. Acta Psychiatrica Scand. 1997;96:334–342.
exciting development and is likely to be followed by the 8. UK ECT Review Group. Efficacy and safety of electroconvulsive
development of other glutamatergic agents. Due to limitations therapy in depressive disorders: a systematic review and meta-
with IN delivery, it seems likely that there will be a push for an analysis. Lancet. 2003;361:799–808.
EXPERT REVIEW OF NEUROTHERAPEUTICS 11

9. Berlim MT, McGirr A, Van den Eynde F, et al. Effectiveness and 28. Baker GB, Prior TI. Stereochemistry and drug efficacy and develop-
acceptability of deep brain stimulation (DBS) of the subgenual ment: relevance of chirality to antidepressant and antipsychotic
cingulate cortex for treatment-resistant depression: a systematic drugs. Ann Med. 2002;34:537–543.
review and exploratory meta-analysis. J Affect Disord. 29. Andrade C. Ketamine for depression, 3: does chirality matter? J Clin
2014;159:31–38. Psychiatry. 2017;78:e674–e677.
10. Martin JLR, Martín-Sánchez E. Systematic review and meta-analysis 30. Muller J, Pentyala S, Dilger J, et al. Ketamine enantiomers in the rapid
of vagus nerve stimulation in the treatment of depression: variable and sustained antidepressant effects. Ther Adv Psychopharmacol.
results based on study designs. Eur Psychiatry. 2012;27:147–155. 2016;6:185–192.
11. Schlaepfer TE, Frick C, Zobel A, et al. Vagus nerve stimulation for 31. Peltoniemi MA, Hagelberg NM, Olkkola KT, et al. Ketamine: a review
depression: efficacy and safety in a European study. Psychol Med. of clinical pharmacokinetics and pharmacodynamics in anesthesia
2008;38:651–661. and pain therapy. Clin Pharmacokinet. 2016;55:1059–1077.
12. Berlim MT, Van Den Eynde F, Jeff Daskalakis Z. Clinically meaningful 32. Pfenninger EG, Durieux ME, Himmelseher S. Cognitive impairment after
efficacy and acceptability of low-frequency repetitive transcranial small-dose ketamine isomers in comparison to equianalgesic racemic
magnetic stimulation (rTMS) for treating primary major depression: ketamine in human volunteers. Anesthesiology. 2002;96:357–366.
a meta-analysis of randomized, double-blind and sham-controlled 33. Singh JB. Antidepressant efficacy and dosing comparisons of keta-
trials. Neuropsychopharmacology. 2013;38:543–551. mine enantiomers: response to Hashimoto. Am J Psychiatry.
13. Berlim MT, van Den Eynde F, Tovar-Perdomo S, et al. Response, 2016;173:1045–1046.
remission and drop-out rates following high-frequency repetitive 34. White PF, Schüttler J, Shafer A, et al. Comparative pharmacology of
transcranial magnetic stimulation (rTMS) for treating major depres- the ketamine isomers. Studies in volunteers. Br J Anaesth.
sion: a systematic review and meta-analysis of randomized, double- 1985;57:197–203.
blind and sham-controlled trials. Psychol Med. 2014;44:225–239. 35. Fukumoto K, Toki H, Iijima M, et al. Antidepressant potential of (R)-
14. Cao X, Deng C, Su X, et al. Response and remission rates following ketamine in rodent models: comparison with (S)-ketamine. J
high-frequency vs. low-frequency repetitive transcranial magnetic Pharmacol Exp Ther. 2017;361:9–16.
stimulation (rTMS) over right DLPFC for treating major depressive 36. Hashimoto K, Kakiuchi T, Ohba H, et al. Reduction of dopamine D2/
disorder (MDD): a meta-analysis of randomized, double-blind trials. 3 receptor binding in the striatum after a single administration of
Front Psychiatry. 2018;9:413. esketamine, but not R-ketamine: a PET study in conscious monkeys.
15. Senova S, Cotovio G, Pascual-Leone A, et al. Durability of antidepres- Eur Arch Psychiatry Clin Neurosci. 2017;267:173–176.
sant response to repetitive transcranial magnetic stimulation: sys- 37. Yang C, Shirayama Y, Zhang J-C, et al. R-ketamine: a rapid-onset
tematic review and meta-analysis. Brain Stimul. 2019;12:119–128. and sustained antidepressant without psychotomimetic side
16. Anis NA, Berry SC, Burton NR, et al. The dissociative anaesthetics, effects. Transl Psychiatry. 2015;5:e632.
ketamine and phencyclidine, selectively reduce excitation of cen- 38. Yang C, Han M, Zhang J-C, et al. Loss of parvalbumin-immunor-
tral mammalian neurones by N-methyl-aspartate. Br J Pharmacol. eactivity in mouse brain regions after repeated intermittent admin-
1983;79:565–575. istration of esketamine, but not R-ketamine. Psychiatry Res.
17. Lodge D, Anis NA, Burton NR. Effects of optical isomers of ketamine 2016;239:281–283.
on excitation of cat and rat spinal neurones by amino acids and 39. Zhang J-C, Li S-X, Hashimoto K. R (-)-ketamine shows greater
acetylcholine. Neurosci Lett. 1982;29:281–286. potency and longer lasting antidepressant effects than S (+)-keta-
18. Miller OH, Moran JT, Hall BJ. Two cellular hypotheses explaining the mine. Pharmacol Biochem Behav. 2014;116:137–141.
initiation of ketamine’s antidepressant actions: direct inhibition and 40. Zanos P, Moaddel R, Morris PJ, et al. Ketamine and ketamine
disinhibition. Neuropharmacology. 2016;100:17–26. metabolite pharmacology: insights into therapeutic mechanisms.
19. Berman RM, Cappiello A, Anand A, et al. Antidepressant effects of Pharmacol Rev. 2018;70:621–660.
ketamine in depressed patients. Biol Psychiatry. 2000;47:351–354. • This is an excellent review of ketamine pharmacology
• Seminal efficacy study demonstrating efficacy of IV ketamine for 41. Tian Z, Dong C, Fujita A, et al. Expression of heat shock protein
major depressive disorder HSP-70 in the retrosplenial cortex of rat brain after administration
20. Murrough JW, Perez AM, Pillemer S, et al. Rapid and longer-term of (R,S)-ketamine and (S)-ketamine, but not (R)-ketamine.
antidepressant effects of repeated ketamine infusions in treatment- Pharmacol Biochem Behav. 2018;172:17–21.
resistant major depression. Biol Psychiatry. 2013;74:250–256. 42. Zhang K, Hashimoto K. An update on ketamine and its two enan-
21. Coyle CM, Laws KR. The use of ketamine as an antidepressant: a tiomers as rapid-acting antidepressants. Expert Rev Neurother.
systematic review and meta-analysis. Hum Psychopharmacol. 2019;19:83–92.
2015;30:152–163. •• This paper is an excellent review of ketamine, R ketamine and S
22. Thomas RK, Baker G, Lind J, et al. Rapid effectiveness of intravenous ketamine.
ketamine for ultraresistant depression in a clinical setting and evi- 43. Chang L, Zhang K, Pu Y, et al. Comparison of anatidepressant and
dence for baseline anhedonia and bipolarity as clinical predictors of side effects in mice after intranasal administration of (R,S)-keta-
effectiveness. J Psychopharmacol (Oxford). 2018;32:1110–1117. mine, (R)-ketamine and (S)-ketamine. Pharmacol Biochem Behav.
23. Archer S, Chrenek C, Swainson J. Maintenance ketamine therapy for 2019;181:53–59.
treatment-resistant depression. J Clin Psychopharmacol. 44. Mathisen LC, Skjelbred P, Skoglund LA, et al. Effect of ketamine, an
2018;38:380–384. NMDA receptor inhibitor, in acute and chronic orofacial pain. Pain.
24. Lapidus KAB, Levitch CF, Perez AM, et al. A randomized controlled 1995;61:215–220.
trial of intranasal ketamine in major depressive disorder. Biol 45. Hashimoto K. Ketamine’s antidepressant action: beyond NMDA
Psychiatry. 2014;76:970–976. receptor inhibition. Expert Opin Ther Targets. 2016;20:1389–1392.
25. Gálvez V, Li A, Huggins C, et al. Repeated intranasal ketamine for 46. Moghaddam B, Adams B, Verma A, et al. Activation of glutamater-
treatment-resistant depression - the way to go? Results from a pilot gic neurotransmission by ketamine: a novel step in the pathway
randomised controlled trial. J Psychopharmacol (Oxford). from NMDA receptor blockade to dopaminergic and cognitive
2018;32:397–407. disruptions associated with the prefrontal cortex. J Neurosci.
26. Lee V, Archer S, Chrenek C, et al. A response to: repeated intranasal 1997;17:2921–2927.
ketamine for treatment resistant depression: the way to go? Results 47. Autry AE, Adachi M, Nosyreva E, et al. NMDA receptor blockade at
from a pilot randomised controlled trial. J Psychopharmacol rest triggers rapid behavioural antidepressant responses. Nature.
(Oxford). 2019;33:258–259. 2011;475:91–95.
27. Abdallah CG, Adams TG, Kelmendi B, et al. Ketamine’s mechanism 48. Li N, Lee B, Liu R-J, et al. mTOR-dependent synapse formation
of action: a path to rapid-acting antidepressants. Depress Anxiety. underlies the rapid antidepressant effects of NMDA antagonists.
2016;33:689–697. Science. 2010;329:959–964.
12 J. SWAINSON ET AL.

49. Abdallah CG, Averill LA, Gueorguieva R, et al. Rapamycin, an immu- corresponding to the definition of remission on the Hamilton rating
nosuppressant and mTORC1 inhibitor, triples the antidepressant scale for depression. J Psychiatr Res. 2004;38:577–582.
response rate of ketamine at 2 weeks following treatment. A 69. Popova V, Daly EJ, Trivedi M, et al. Efficacy and safety of flexibly dosed
double-blind, placebo-controlled, cross-over, randomized clinical esketamine nasal spray combined with a newly initiated oral anti-
trial. bioRxiv preprint first posted online. 2018 Dec 19. depressant in treatment-resistant depression: a randomized double-
DOI:10.1101/500959. blind active-controlled study. Am J Psychiatry. 2019;176:428–438.
50. Zanos P, Moaddel R, Morris PJ, et al. NMDAR inhibition-indepen- •• Phase 3 trial demonstrating statistical significant superiority
dent antidepressant actions of ketamine metabolites. Nature. over placebo
2016;533:481–486. 70. Ochs-Ross R, Daly EJ, Zhang Y, et al. Efficacy and safety of intranasal
51. Chaki S. Is metabolism of (R)-ketamine essential for the antidepres- esketamine plus an oral antidepressant in elderly patients with
sant effects? Int J Neuropsychopharmacol. 2018;21:154–156. treatment-resistant depression. Biol Psychiatry. 2018;83:S391.
52. Li L, Vlisides PE. Ketamine: 50 years of modulating the mind. Front •• Phase 3 trial demonstrating benefits to maintenance therapy
Hum Neurosci. 2016;10:612. versus placebo up to 104 weeks post response
53. Mion G, Villevieille T. Ketamine pharmacology: an update (pharma- 71. Joyce PR, Mulder RT, Luty SE, et al. Patterns and predictors of remis-
codynamics and molecular aspects, recent findings). CNS Neurosci sion, response and recovery in major depression treated with fluox-
Ther. 2013;19:370–380. etine or nortriptyline. Aust N Z J Psychiatry. 2002;36:384–391.
54. Williams NR, Heifets BD, Blasey C, et al. Attenuation of antidepres- 72. Mynors-Wallis L, Gath D. Predictors of treatment outcome for major
sant effects of ketamine by opioid receptor antagonism. Am J depression in primary care. Psychol Med. 1997;27:731–736.
Psychiatry. 2018;175:1205–1215. 73. Daly E, Trivedi M, Janik A, et al. A randomized withdrawal, double-
55. Zhang K, Hashimoto K. Lack of opioid system in the antidepressant blind, multicenter study of esketamine nasal spray plus an oral
actions of ketamine. Biol Psychiatry. 2019;85:e25–e27. antidepressant for relapse prevention in treatment-resistant
56. Yang Y, Cui Y, Sang K, et al. Ketamine blocks bursting in the lateral depression. Poster/abstract presented at Annual Meeting of the
habenula to rapidly relieve depression. Nature. 2018;554:317–322. American Society of Clinical Psychopharmacology, Hollywood,
57. Yanagihara Y, Ohtani M, Kariya S, et al. Plasma concentration Florida; May 30, 2018. Poster W68.
profiles of ketamine and norketamine after administration of var- 74. Wajs E, Aluisio L, Morrison R, et al. Long-term safety of esketamine
ious ketamine preparations to healthy Japanese volunteers. nasal spray plus an oral antidepressant in patients with treatment-
Biopharm Drug Dispos. 2003;24:37–43. resistant depression: phase 3, open label safety and efficacy study
58. Chong C, Schug SA, Page-Sharp M, et al. Development of a sub- (SUSTAIN-2). Poster presented at: The American Society of Clinical
lingual/oral formulation of ketamine for use in neuropathic pain: Psychopharmacology Meeting; May 2018; Miami, Florida. 2018.
preliminary findings from a three-way randomized, crossover study. •• Phase 3 trial demonstrating 6.9% TEAE, an attenuation of dis-
Clin Drug Investig. 2009;29:317–324. sociation symptoms over time and no meaningful effect on
59. Fanta S, Kinnunen M, Backman JT, et al. Population pharmacokinetics cognition over 56 weeks of maintenance treatment
of S-ketamine and norketamine in healthy volunteers after intrave- 75. Correia-Melo FS, Leal GC, Carvalho MS, et al. Comparative study of
nous and oral dosing. Eur J Clin Pharmacol. 2015;71:441–447. esketamine and racemic ketamine in treatment-resistant depres-
60. Brunton L, Hilal-Dandan R, Knollman B Goodman & Gilman’s The sion: protocol for a non-inferiority clinical trial. Medicine
Pharmacological Basis of Therapeutics, 13e. Minion Pro by Cenveo® (Baltimore). 2018;97:e12414.
Publisher Services; 2018. 76. Short B, Fong J, Galvez V, et al. Side-effects associated with keta-
61. Portmann S, Kwan HY, Theurillat R, et al. Enantioselective capillary mine use in depression: a systematic review. Lancet Psychiatry.
electrophoresis for identification and characterization of human 2018;5:65–78.
cytochrome P450 enzymes which metabolize ketamine and norke- 77. Schatzberg AF. A word to the wise about intranasal esketamine.
tamine in vitro. J Chromatogr A. 2010;1217:7942–7948. Am J Psychiatry. 2019;176:422–424.
62. Ihmsen H, Geisslinger G, Schüttler J. Stereoselective pharmacoki- 78. Wan L-B, Levitch CF, Perez AM, et al. Ketamine safety and toler-
netics of ketamine: R(-)-ketamine inhibits the elimination of S ability in clinical trials for treatment-resistant depression. J Clin
(+)-ketamine. Clin Pharmacol Ther. 2001;70:431–438. Psychiatry. 2015;76:247–252.
63. Janssen Pharmaceuticals inc. SpravatoTM (esketamine hydrochlor- 79. FDA report on esketamine for treatment resistant depression. 2019;
ide) [Package insert]. Copyright Janssen Pharmaceuticals; Titusville, Accessed April 9, 2019. Available from: https://www.fda.gov/down
NJ; 2019. Available from: http://www.janssenlabels.com/package- loads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/
insert/product-monograph/prescribing-information/SPRAVATO-pi. PsychopharmacologicDrugsAdvisoryCommittee/UCM630970.pdf.
pdf 80. van de Loo AJAE, Bervoets AC, Mooren L, et al. The effects of
64. Daly EJ, Singh JB, Fedgchin M, et al. Efficacy and safety of intranasal intranasal esketamine (84 mg) and oral mirtazapine (30 mg) on
esketamine adjunctive to oral antidepressant therapy in treatment- on-road driving performance: a double-blind, placebo-con-
resistant depression: A randomized clinical trial. JAMA Psychiatry. trolled study. Psychopharmacology (Berl.). 2017;234:3175–3183.
2018;75:139–148. 81. Chu PS-K, Ma W-K, Wong SC-W, et al. The destruction of the
65. Canuso CM, Singh JB, Fedgchin M, et al. Efficacy and safety of lower urinary tract by ketamine abuse: a new syndrome?. BJU
intranasal esketamine for the rapid reduction of symptoms of Int. 2008(102):1616-1622.
depression and suicidality in patients at imminent risk for suicide: 82. Shahani R, Streutker C, Dickson B, et al. Ketamine-associated ulcera-
results of a double-blind, randomized, placebo-controlled study. tive cystitis: A new clinical entity. Urology. 2007;69:810–812.
Am J Psychiatry. 2018;175:620–630. 83. Tam Y-H, Ng C-F, Pang KK-Y, et al. One-stop clinic for keta-
66. Fedgchin M, Trivedi M, Daly EJ, et al. Randomized, double-blind mine-associated uropathy: report on service delivery model,
study of fixed-dosed intranasal esketamine plus oral antidepressant patients’ characteristics and non-invasive investigations at
vs. active control in treatment-resistant depression. Presented at baseline by a cross-sectional study in a prospective cohort of
the 9th Biennial Conference of the International Society for 318 teenagers and young adults. BJU Int. 2014;114:754–760.
Affective Disorders (ISAD) and the Houston Mood Disorders 84. Tsai J-H, Tsai K-B, Jang M-Y. Ulcerative cystitis associated with
Conference. Houston, TX, USA, 2018 September 21. ketamine. Am J Addictions. 2008;17:453.
67. Lam RW, Michalak E, Swinson R. Assessment scales in depression, 85. Morrison RL, Fedgchin M, Singh J, et al. Effect of intranasal
mania and anxiety. London: Taylor and Francis; 2005. esketamine on cognitive functioning in healthy participants: a
68. Zimmerman M, Posternak MA, Chelminski I. Derivation of a definition randomized, double-blind, placebo-controlled study.
of remission on the Montgomery-Asberg depression rating scale Psychopharmacology (Berl). 2018;235:1107–1119.
EXPERT REVIEW OF NEUROTHERAPEUTICS 13

86. Morgan CJA, Riccelli M, Maitland CH, et al. Long-term effects of 91. Royal College of Psychiatrists. Statement on ketamine to treat depres-
ketamine: evidence for a persisting impairment of source memory sion. Position statement cert02/17. Approved by the Royal College of
in recreational users. Drug Alcohol Depend. 2004;75:301–308. Psychiatrists, Committee on ECT and related treatments: 2017.
87. Shiroma PR, Albott CS, Johns B, et al. Neurocognitive performance 92. Sanacora G, Frye MA, McDonald W, et al. A consensus statement on
and serial intravenous subanesthetic ketamine in treatment-resis- the use of ketamine in the treatment of mood disorders. JAMA
tant depression. Int J Neuropsychopharmacoly. 2014;17:1805–1813. Psychiatry. 2017;74:399–405.
88. Unpublished data: clinical registry number 108104: a single-center, •• The APA Consensus statement on use of ketamine contains an
single-dose, double blind, double dummy, placeo controlled, ran- appendix (online version) that makes practical suggestions for
domized crossover study to evaluate the abuse potential of intra- judicious use of ketamine. This appendix contains many clini-
nasal esketamine compared to racemic intravenous ketamine in cally applicable suggestions for use and monitoring when pre-
nondependentm, recreational users of perception-altering drugs. scribing esketamine.
89. Kolar D. Addictive potential of novel treatments for refractory depres- 93. Yatham LN, Kennedy SH, Parikh SV, et al. Canadian Network for
sion and anxiety. Neuropsychiatr Dis Treat. 2018;14:1513–1519. Mood and Anxiety Treatments (CANMAT) and International
90. Nutt D, King LA, Saulsbury W, et al. Development of a rational scale to Society for Bipolar Disorders (ISBD) 2018 guidelines for the
assess the harm of drugs of potential misuse. Lancet. 2007;369:1047– management of patients with bipolar disorder. Bipolar Disord.
1053. 2018;20(2):97–170.

You might also like