Henry Ford Health Henry Ford Health
Henry Ford Health Scholarly Commons Henry Ford Health Scholarly Commons
Behavioral Health Articles Behavioral Health Services / Psychiatry
8-27-2020
Effectiveness and Safety of Ketamine for Unipolar Depression: a Effectiveness and Safety of Ketamine for Unipolar Depression: a
Systematic Review Systematic Review
Raheel I. Memon
Henry Ford Health
Sadiq Naveed
Amber Ehsan Faquih
Ania Fida
Noureen Abbas
See next page for additional authors
Follow this and additional works at: https://scholarlycommons.henryford.com/behavioralhealth_articles
Recommended Citation Recommended Citation
Memon RI, Naveed S, Faquih AE, Fida A, Abbas N, Chaudhary AMD, and Qayyum Z. Effectiveness and
Safety of Ketamine for Unipolar Depression: a Systematic Review. Psychiatr Q 2020.
This Article is brought to you for free and open access by the Behavioral Health Services / Psychiatry at Henry Ford
Health Scholarly Commons. It has been accepted for inclusion in Behavioral Health Articles by an authorized
administrator of Henry Ford Health Scholarly Commons.
Authors Authors
Raheel I. Memon, Sadiq Naveed, Amber Ehsan Faquih, Ania Fida, Noureen Abbas, Amna Mohyud Din
Chaudhary, and Zheala Qayyum
This article is available at Henry Ford Health Scholarly Commons: https://scholarlycommons.henryford.com/
behavioralhealth_articles/83
REVIEW ARTICLE
Effectiveness and Safety of Ketamine for Unipolar
Depression: a Systematic Review
Raheel Imtiaz Memon
1
& Sadiq Naveed
2
& Amber Ehsan Faquih
3
& Ania Fida
4
&
Noureen Abbas
5
& Amna Mohyud Din Chaudhary
6
& Zheala Qayyum
7
#
Springer Science+Business Media, LLC, part of Springer Nature 2020
Abstract
Major Depressive Disorder (MDD) is a common psychiatric disorder with major impli-
cations for healthcare system and socioeconomic burden. For chronic and treatment-
resistant depression, Ketamine has emerged as a possible treatment option. This system-
atic review explores the evidence for the effectiveness and tolerability of Ketamine in
patients with MDD. This systematic review was conducted following the guidelines of
Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) check-
list. Eight electronic databases were searched by using search terms: (ketamine) AND
(trial OR RCT OR clinical-trial) AND (depressive OR depression OR depressive-
disorder). After a rigorous screening process against the predetermined eligibility
criteria, 35 randomized controlled trials (RCTs) were included. Quality assessment of
included studies was done by using the Cochrane risk-of-bias tool for RCTs. Thirty-five
RCTs are included in this review article with majority of studies from United States, Iran,
and China. Intravenous (IV) Ketamine was effective in 70% (21/30) of the included
studies whereas oral and Intranasal (IN) Ketamine were effective in two and three studies,
respectively. The majority of studies (6/8) using Ketamine as anesthetic agent during
electroconvulsive therapy (ECT) failed to show an improvement compared to the partic-
ipants receiving ECT and placebo. The most common reported side effects were nausea,
vomiting, dizziness, diplopia, drowsiness, dysphoria, hallucinations, and confusion. Ke-
tamine is an effective treatment option for patients with MDD with undesirable effects
when administered via oral, IV and IN routes. Ketamine agumentation of ECT requires
further exploration in well-designed studies with adequate sample size. The short-lived
antidepressant effect of Ketamine is a potential limitation, therefore, further studies
administering multiple infusions for acute treatment and maintenance are necessary.
Keywords Ketamine
.
MDD
.
Major depressivedisorder
.
MDD
.
Treatment-resistant depression
Psychiatric Quarterly
https://doi.org/10.1007/s11126-020-09830-6
Electronic supplementary material The online version of this article (https://doi.org/10.1007/s11126-020-
09830-6) contains supplementary material, which is available to authorized users.
* Sadiq Naveed
naveed193@gmail.com
Extended author information available on the last page of the article
Introduction
Major Depressive Disorder (MDD) is a widespread and disabling illness with a lifetime
prevalence of 20% in USA. MDD affects around 264 million people throughout the world,
leading to major health system and socioeconomic burden [1,2]. The global incidence of
depression has increased by 49.86% in the last two decades and it is estimated that MDD will
become the leading cause of debility by the year 2030 [3]. Untreated depression has debili-
tating consequences for individuals, resulting in academic, interpersonal, social, and occupa-
tional impairments [4]. The clinical course of depression tends to be chronic with a high rate of
recurrence of around 80% [4]. The economic burden of depression is reported to be around
$210.5 billion with approximately 45% due to direct costs, 5% to suicide-related costs, and
50% to workplace costs [5]. Beside the financial implications, around 90% of suicidal patients
have an underlying psychiatric illness with MDD being the top of the list [6]andabout215%
of patients with MDD complete suicide [7].
The treatment of depression requires several careful considerations, such as MDD with
suicidal behaviors, early-onset depression, chronic depression, and treatment- resistant depres-
sion. With an increased emphasis on deinstitutionalization, there is a policy shift to manage
patients in a less restrictive environment with shortened length of hospital stays [8]. In this
context, pharmacological options with robust and faster response became critically important
in treatment of patients with MDD. Existing treatment options such as antidepressants,
psychotherapy, and electroconvulsive therapy (ECT) are effective but come at the cost of time
lag, so a large number of patients face the challenge of suicidal thoughts and impairing
depressive symptoms for weeks after initiation of treatment [9].
More importantly, treatment- resistant depression (TRD) continues to be a significant
public health issue. Despite multiple treatment regimens, about 6070% of patients with
MDD respond to first-line antidepressants, whereas about one-third of patients struggle with
debilitating and chronic depression [2]. Recently, in addition to psychopharmacological
treatments mediated through monoamines, the glutamate pathway and N-methyl-D-aspartate
(NMDA) receptor antagonism have become the subject of attention.
Glutamate, a neurotransmitter associated with neuroplasticity and excitotoxicity action, has
antidepressant effect by its effect on different receptors, especially N-methyl-D-aspartate
(NMDA) and α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptors.
Ketamine, a phencyclidine hydrochloride derivative and a non-competitive NMDA receptor
antagonist, was first approved in 1970 for anesthesia. It is also used during ECT since it can
intrinsically increase the seizure duration with minimal cognitive deficits [2,10,11]. The use of
Ketamine has recently expanded to its antidepressant use at s ub-anesthetic and sub-
dissociative doses, but the exact antidepressant mechanism is still unclear [12,13].
Ketamines NMDA receptor antagonism and facilitated AMPA receptor-mediated trans-
mission [14] result in a cascade of events, such as activation of the target of rapamycin
(mTOR) intracellular cascade and, specifically, upregulation of brain-derived neurotrophic
factor (BDNF). BDNF, a neurotrophic factor, is involved in neuro nal maturation,
neurogenesis, and synaptic plasticity [15]. Ketamine is a racemate compound of 2 enantio-
mers: R-(2)-ketamine e nantiomer (arketamine) and the S-(1)-ketamine enantiomer
(esketamine) which has about 34 folds higher affinity for NMDA receptor than arketamine
[16,17]. The half-life (t1/2) of Ketamine is reported to be around 34 h, but its antidepressant
action is reported to last for a week. This raises an important cue that its action is not entirely
dependent on NMDA receptors blockade but also has a long-term neuroprotective effect [18].
Psychiatric Quarterly
This systematic review provides a comprehensive overview of the use of Ketamine in the
prevention and treatment of unipolar depression.
Methods
This systematic review was conducted according to the guidelines of the Preferred Reporting
Items for Systematic Reviews and Meta-Analysis (PRISMA) checklist (Supplementary
Table 1). The protocol was developed in March 2019 and was registered with the International
Prospective Register of Systematic Reviews (PROSPERO) in April 2019
(CRD42019125801).
Eligibility Criteria
The inclusion criteria were:
1. Randomized controlled trials (RCTs) focused on the treatment of unipolar depression with
ketamine as a pharmacotherapy.
2. RCTs that focused on the adult population with MDD were included, who were either
screened for depression using validated screening instruments or diagnostic systems (such
as International Statistical Classification of Diseases and Related Health Problems or
Diagnostic and Statistical Manual).
No restriction on race, geography, sex, age, ethnicity, or language and publication
date were applied.
The exclusion criteria were:
1. Study design other than RCT (observational study, case reports, case series, letter to
editors, study protocols, thesis, reviews, commentary, conference papers, book chapter or
news articles).
2. Studies discussing the role of Ketamine for bipolar depression or diagnosis other than
MDD.
3. Overlapped data sets, unreliable information, and abstract-only articles.
4. In-vitro studies or animal studies.
Search Strategy
Eight academic databases were searched including PubMed, CINAHL, Cochrane
clinical trials registry, Web of Science, PsycINFO, POPLINE, Global Health Library,
and Virtual Health Library through September 2018, using the following search
strategy: (ketamine) AND (trial OR RCT OR Clinical-trial) AND (depressive OR
depression OR d epressive-disorder). The manual search of references and relevant
articles for included studies was performed by two independent reviewers. Any
discrepancies among reviewers were resolved by discussion or guidance f rom a senior
reviewer (SN).
Psychiatric Quarterly
Study Selection
Search results from the eight databases were imported to Endnote ×7 (Thompson Reuter, CA,
USA) to remove any duplicates. Two independent reviewers performed title and abstract
screening (when available) followed by the full-text screening of the included articles by using
the predetermined eligibility criteria. Disagreements were resolved by discussion among
reviewers or guidance from a senior reviewer (SN).
Data Extraction
The data were e xtracted by two independent reviewers and c ross-checked for accuracy by
the senior author (SN). The name of authors, sample size, site of the trial, participant
characteristics, route of administration, dose range, clinical outcomes, the adjunct treat-
ment used, and common side effects were tabulated. The meta-analysis was not performed
due to varying study d esign, population of interest, and treatment outcomes, posing
potential heterogeneity.
Risk of Bias Assessment
Two authors assessed the quality of the studies without blinding to authorship or journal, using
the Cochrane tool for randomized controlled trials against several matrices: a) sequence
generation, b) allocation concealment, c) blinding of participants and personnel, d) blinding
of outcome assessment, e) incomplete outcome data, f) selective reporting, and g) other bias.
Funding Source
There was no funding source for this study.
Results
Study Search
The initial search of databases revealed a total number of 689 non-duplicate references
predicated on their titles and abstracts screening. After the application of inclusion and
exclusion criteria, 581 citations were excluded after title and abstract screening. Later thorough
screening of 108 full texts eventually yielded 35 RCTs. Fig. 1,PRISMAFlowDiagram
summarizes the screening process.
Study Characteristics
This review article includes 35 RCTs conducted between 2002 to 2018, with most of the study
sites in the USA [14], Iran [9], and China (4) [1923]. The remaining trials were singularly
conducted in Canada, UK, Taiwan, Australia, Japan, Scotland, and Czech Republic. Daly et al.
(2017) conducted one study in USA and Belgium [1] whereas Singh et al. conducted one study
in Belgium, Germany, and Poland. There were 2183 participants in these studies with an age
range of 1880, however age range was not mentioned in two studies [24,25]. Most studies
Psychiatric Quarterly
included both men and women, except for two studies that were conducted exclusively in
women [18,26]. Tables 1 and 2 provides a summary of participant characteristics, character-
istics related to Ketamine, outcome measures, and side effects.
Fig. 1 PRISMA Flow Diagram
Psychiatric Quarterly
Table 1 Overview of randomized controlled trials included in the meta-analysis
Study Design Inclusion Criteria Primary Outcome Secondary Outcome
Alizadeh et al.,
2015
Propofol & Ketamine vs Propofol
&placebo
Age 1865, suffering from MDD, and
HDRS score of 20
Mean HDRS scores Not mentioned
Arabzadeh et al.,
2018
Sertraline & Ketamine vs Sertraline
&placebo
Age 1860, patients with moderate to
severe depression, HDRS
score 20
Change in the HDRS score from
baseline to week 2 across the two
groups.
1) Difference in the HDRS score at week 4
and 6.
2) Difference in early improvement (
20% reduction in HDRS score within
the first two weeks)
3) Response to treatment (50% reduction
in the HDRS score at the termination of
the trial)
4) Remission (HDRS score 7atthe
termina t io n of the tria l) between the two
groups.
Burger et al., 2016 Single infusion of Ketamine vs
placebo
Age 1865 years, Beck Suicidality
Scale (BSS) score > 4, Beck Hope-
lessness Scale (BHS) score > 8,
Beck Depression Inventory score >
19, and the ability to give informed
consent. Negative pre gnan cy test
for females.
Beck Suicidality Scale (BSS) Beck Hopeless-ness Scale (BHS)
Canuso et al.,
2018
Esketamine vs Placebo Age 196 4, suffering from MD D
without psychotic features, and a
score >/=22 on the
Montgomery-Åsberg Depression
Rating Scale (MADRS).
1) Change in Montgomery-Åsberg
Depression Rating Scale (MADRS)
score from baseline to 4 h after
initial dose.
2) Clinician global judgment of suicide
risk (from the Suicide Ideation and
Behavior Assessment Tool).
The primary outco me measures at 24 h
and double-blind endpoint at day 25.
Carspecken et al.,
2018
Ketamine vs Methohexital All vetera ns >18 years of age,
scheduled for an index course of
ECT for TRD
PHQ-9, HAM-D MOCA
Chen et al., 2017 Ketamine vs propofol Age 1865, with MDD, HAM-D,
scores >35.
Long-term memory, short-term
memory, and immediate memory
were assessed for all patients using
Depression was assessed by the 24-item
HAM-D interview
Psychiatric Quarterly
Table 1 (continued)
Study Design Inclusion Criteria Primary Outcome Secondary Outcome
the Wechsler Memory
Scale-Chinese Revision (WM S-RC)
Fan et al., 2017 Ketamine vs Midazolam Participants with MDD and newly
diagnosed breast cancer
(A) Suicidal ideation severity
evaluated with BSI score.
(B) Suicidal ideation severity
evaluated with MADRS-SI
Score.
(C) Overall depression severity
evaluated with MADRS score
Not mentioned
Gamble et al.,
2018
Ketamine vs Propofol Age > 17, TRD, MADRS score of 20. Number of ECT treatments required to
reach a 50% reduction in baseline
MADRS
1. Change in CADSS
2. Change in ALS-18
3. Change in ECT energy settings and
seizure quality
4. Hemodynamic instab ility and
respiratory complications
5. Time to discharge
6. Change in MADRS score 24 h after
each treatment and 30 days after final
treatment for an expected average of
2 months
7. The number of ECT sessions required to
achieve depression remission (MADRS
10)
8. The proportion of depressed patients
(MADRS >20) at 30 days after the last
ECT session
9. Change in systolic blood pressure
Ghasemi et al.,
2013
Ketamine vs ECT Participants with MDD, ages
1875 years, with a current major
depressive episode.
HDRS, BDI at baseline, 24 h after
each treatment, 72 h and one week
after the last (third)
Ketamine or ECT.
Not mentioned
Haile et al., 2013 Ketamine vs Midazolam Age 2180 years, participants with
TRD, current episode of depression.
Not mentioned
Psychiatric Quarterly
Table 1 (continued)
Study Design Inclusion Criteria Primary Outcome Secondary Outcome
Response = 50% or greater reduction
in MADRS score compa red to
baseline at 7 day post-infusion
Hu et al., 2015 Ketamine vs Placebo Age 186 0 years, patients with TRD,
24 on HAM-D
Response = 50% reduction in
baseline MADRS scores.
Remission = MADRS total score 10
- Proportion of responders and remitters in
each group
- Severity of investigator-rated depressive
symptoms (MADRS)
- Self-rated scores on Quick Inventory of
Depressive Symptomatology
Self-Report (QIDS-SR) Chinese
version,
- Suicidal ideation (QIDS-SR item 12)
- Side- effec ts and severity of manic, psy-
chotic and dissociative symptoms.
Jafarinia et al.,
2016
Ketamine vs Diclofenac Age 2055 years, patients with TRD,
chronic mild-moderate headache
HDRS at week 3 and 6, HADS Comparison of changes in HDRS scores
from baseline to each time point,
response to treatment (defined as 50%
reductionintheHDRSscore),
remission (defined as HRDS score 7),
and severity of pain intensity between
the treatment groups and evaluation of
the antidepressant effects of each drug
separately.
Jarventausta K.
et al. 2013
S-Ketamine & propofol vs Normal
saline & propofol
Age 1880 years, participants with
TRD with failure of two
antidepressants in the past.
MADRS Not mentioned
Jiang M. , et al.,
2016
Ketamine vs Control Age 1860 years, patients undergoing
orthopedic surgery
PHQ-9 Not mentioned
Tayyebi et al.,
2018
Ketamine bolus vs infusion Age 2060 years, patients with MDD HDRS, BDI Not mentioned
Lai R. et al., 2014 Ketamine at different doses vs
saline
Participants with TRD, Age
2966 years, MADRS score of
20
MADRS SAFTEE scale
Psychiatric Quarterly
Table 1 (continued)
Study Design Inclusion Criteria Primary Outcome Secondary Outcome
Lapidus et al.,
2014
Ketamine vs Placebo Age 2165 years, MDD, baseline
score of 30 on the Inventory of
Depressive
SymptomatologyClinician Rated
(IDS-C)
MADRS at 24 h after treatment Systematic Assessment for Treatment
Emergent Effects (SAFTEE)
Lenze et al., 2016 Saline infusion followed by
Ketamine
40 min infusion vs 96 h infusion
Age 1865 years, MDD, MADRS
score of
MADRS Not mentioned
Lo et al., 2016 Ketamine vs Midazolam Participants, ages 18 years , major
depressive disorder and a depressive
episode of duration 4weeks.
Change in MADRS scores BPRS, YMRS, CADSS
Murrough et al.,
2016
Ketamine vs Midazolam Age 2180, with MDD, and
inadequate response to at least three
therapeutic trials of an
antidepressant
Change in MADRS scores 24 h after
infusion
MADRS response rate, change in score on
the Quick Inventory of Depressive
SymptomatologySelf-Report, scores
on the Clinical Global Impression (CGI)
severity and improvement measures,
and durability of benefit for up to
7 days following infusion.
Fava et al., 2018 Ketamine vs Midazolam Age 1870 years old patient with TRD
MDD, current major depressive ep-
isode
HAM-D-6
2- groups comparison = Ketamine vs
Midazolam
5-groups comparison between all four
doses of Ketamine and placebo
MADRS, CGI-S, CGI-I, SDQ, and PAS
Fernie et al., 2017 Ketamine vs Propofol Age 1870 years with MDD receiving
ECT on an informal basis, consid-
ered fit by an anesthetist, had no
comorbid psychiatric diagnoses
HRSD, MADRS Cognitive function, assessed before ECT
using the Cambridge Automated
Neuropsychological Test Battery
Spatial
Recognition Memory task (CANTAB
SRM)
Daly et al., 2017 Esketamine vs Placebo Age 2064 years, TRD MDD and IDS
score 34
Change in MADRS Clinical Global Impression of Severity
scale
Severity of anxiety on the Generalized
Anxiety Disorder 7-item scale
Psychiatric Quarterly
Table 1 (continued)
Study Design Inclusion Criteria Primary Outcome Secondary Outcome
Grunebaum et al.,
2018
Ketamine vs Midazolam 18 to 65 years, adults with current
MDD, HDRS-17 score 16 and
score 4ontheScaleforSuicidal
Ideation (SSI)
SSI Differential change between groups in SSI
and depressive sympt oms (HDRS17
and 24, BDI, POMS)
Kudoh et al., 2002 Propofol, Fentanyl, Ketamine vs
Propofol and Fentanyl
Participants, ages 35 to 63 years for
intervention group and 30 to
64 years for control group), all
patients with MDD, undergoing
orthopedic surgery.
The control group did not have any
psychiatric illnesses.
Change in HDRS scores Postoperative confusion assessed using
confusion assessment method (CAM),
pain was estimated using a 100 mm
visual analog scale
Salehi et al., 2015 Ketamine vs Sodium thiopental Age 20 and 60-year-old, TRD HMDRS Blood pressure
Singh et al., 2016 Ketamine vs Placebo 1864 years old adults, recurrent
MDD- TR without psychotic
features, and a score of 34 on the
30-item Inventory of Depressive
SymptomatologyClinician rated .
Change in MADRS scores Early onset of clinical response, total
number of responders at day15, total
Change in MADRS score from baseline
through day 29, Clinical Global
Impressions severity score (CGI-S),
CGI improvement score (CGI-I),
Patient Global Impression severity
score (PGI-S), Patient Global
Impression of Change score (PGI-C)
Singh et al., 2016 Esketamine vs Placebo 1864 years old adults, recurrent
MDD- TR without psychotic fea-
tures.
Change in MADRS scores 24 h. change in MADRS total score from day 1
to day 3 and day 4 and from day 4 to
day 7, change in MADRS total score
from day 1 to day 35, change in the
Quick Inventory of Depressive
SymptomatologySelf Report, Clinical
Global ImpressionSeverity, Clinical
Global ImpressionImprovement, Pa-
tientGlobalImpression of Severity, and
Patient Global Impression of Change.
Psychiatric Quarterly
Table 1 (continued)
Study Design Inclusion Criteria Primary Outcome Secondary Outcome
Sos et al., 2013 Ketamine vs Placebo Participants with MDD, ages18 and
65 years old, MADRS score of 20
MADRS score change at day 1, 4 and
7 betwe en ketamine and placebo
Response rates
Plasma levels of ketamine and its
metabolite nor-ketamine during keta-
mine and placebo infusion at baseline,
10 mins, 30 mins of the infusion)
Su et al., 2017 Ketamine vs Placebo All patients with Treatment-resistance
MDD, score of more than 18 on
HAMD
HAMD-17 scores Response rate, dose group Responder vs
BDNF genotype, Predictor of response
and sustained effe ct
Wang et al., 2012 Ketamine vs Propofol vs Ketamine
&Propofol
Patient with MDD and score 20 on
HDRS.
Change in HDRS scale None mentioned
Xu et al., 2017 Ketamine vs Control Women age between 30 and 35 years,
who underwent modified radical
mastectomy of unilateral breast.
HAMD score of 17.
Change in HAMD scores Not mentioned
Xu et al., 2017 Ketamine vs Saline American Society of Anesthesiologists
(ASA) grade 12 and elective cae-
sarean deliverywithspinal
anaethesia.
For prevention of depression
EPDS Numeric rating scale score of pain
Yoosefi et al.,
2014
Ketamine vs Thiopental 2050 years old, MDD and
HAM-D 18.
Mean change in HAM-D scores The effects of ketamine and thiopental on
the cognitive consequences, seizure
parameters, and hemodynamic factors
of ECT
Zarate et al., 2006 Ketamine vs Placebo Participants with treatment- resistant
MDD, ages 1865 years old,
HDRS score of 18.
Mean change in HDRS-21 scores Beck Depression Inventory (BDI), Brief
Psychiatric Rating Scale (BPRS) posi -
tive symptoms subscale, Young Mania
Rating Scale (YMRS).
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
Alizadeh et al.,
2015
IV Ketamine 0.3 mg/kg per
ECT session
5 ml of normal saline Primary:
Mean HDRS scores
Baseline scores
The HDRS scores were similar among both
groups with similar recovery time and
speed.
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
Ketamine = 35.4 ± 6.7
Placebo = 36.44 ± 7.17
Study endpoint
Ketamine = 14.18 ± 11.83
Placebo = 14.33 ± 9.46
Secondary: NA
Arabzadeh et al.,
2018
Oral Ketamine 50 mg/day
All patients received
25-150 mg/day sertraline
Oral 50 mg/day placebo
All patients received
25-150 mg/day sertraline
Primary:
Change in HDRS scores = 3.41 (5.07 to
1.75)
Secondary outcome measures:
1) At 4 weeks: 2.61 (4.11 to 1.11) and at
6 weeks:
1.91 (3.34 to 0.48)
2) Ketamine (85.4%) vs Placebo (42.5%).
3) Ketamine (85.4%) vs Placebo (57.5%)
4) Ketamine (22.0%) vs Placebo (15.0%)
- Significant difference between Ketamine and
placebo group was observed at week 2, 4,
and 6.
- Early improvement and response rate was
greater in Ketamine (85.4%, 85.4%)
compared to the placebo group (42.5%,
57.5%), respectively.
- The remission rates were similar among both
groups.
Burger et al., 2016 IV Ketamine 0.2 mg/kg
over 2 min
IV Normal saline infused over
2min
Primary:
2/3 participants reported improvement in
suicidal ideations within 40 min in
suicidality while no improvement was
reported in placebo group.
Secondary: NA
Two of three who received ketamine reported
improvement in suicidality and hopelessness
compared to none among the control group.
Canuso et al.,
2018
Intranasal 84 mg
Esketamine.
Intranasal 84 mg Place b o. Primary:
1) Change in MADRS:
Esketamine group = 13.4 SD = 9.03)
Placebo group = 9.1 (SD = 8.38)
2) Esketamine VS Placebo: 21.2% and
9.7%, respectively
Secondary:
1)MADRSat24h:leastsquaremean
difference = 27.2, SE = 2.85, at day 25:
least-square mean difference = 24.5, SE =
3.14.
- The MADRS scores improved four hours
after the first dose in both groups, with a
greater improvement in Esketamine group.
- A greater improvement was reported among
Esketamine group compared to placebo
group at all stud timepoint during blinded
phase.
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
2) At 24 h
after the first dose: 40.0% and 6.5%, respectively,
and at day 25: 69% and 61% respectively
Carspecken et al.,
2018
IV racemic Ketamine
12mg/kg
IV Methohexital 1 to 2 mg/kg Primary:
HAM-D: Ketamine (F1,45 = 7.8, MSE =333.6,
P = 0.008, ηp2 = 0.16) vs Methohexital
(F1,45 = 0.43, mean square error
[MSE] = 18.4, P =0.51,ηp2 = 0.01)
PHQ-9: Ketamine (F1,47 = 6.45, MSE =140.4,
P = 0.01, ηp2 = 0.13) vs Methohexital
(F1,47 = 0.96, MSE = 21, P =0.331,
ηp2 = 0.02)
PHQ-9
Baseline scores:
Ketamine = 21.1 (±3.9)
Methohexital = 21.5 (±3.6)
Study Endpoint:
Ketamine = 7.2 (±4.4)
Methohexital = 8.7 (±5.3)
HAM-D
Baseline scores:
Ketamine = 27.6 (±8.7)
Methohexital = 28.8 (±5.2)
Study Endpoint:
Ketamine = 12.3 (±7.6)
Methohexital = 15 (±6.9)
Secondary:
MOCA
Baseline scores:
Ketamine = 27.4 (±2.0)
Methohexital = 26.7 (±3.9)
Study Endpoint:
Ketamine = 25.6 (±3.8)
- Patients in both groups reported improvement
after the ECT course.
- Plasma BDNF increased after ECT only in
the ketamine group
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
Methohexital = 24.4(±4.1)
Chen et al., 2017 IV Ketamine, 0.3 mg/kg.
All patient received ECT
treatment.
IV Normal Saline 0.3 mg/kg.
All patient received ECT
treatment.
Primary:
The occurrence global cognitive impairment in
the control group was higher than it was in
the study group.
ThedeclineintheWechslerMemory
Scale-Chinese- Revision scale was greater in
the control group than in the study group.
Secondary:
The overall remission rates in the control group
and the study group were 68% and 74%
respectively with no significant difference
among both groups.
- The overall remission rates were 74% and
68% for the Ketamine and control group
respectively.
- There was no statistically significant
difference among both groups.
- The median ECT times were 8 for ketamine
and 9 for the control group.
- The total number of ECT procedures were
511 for Ketamine and 584 for control group.
Fan et al., 2017 IV 0.5 mg/kg racemic
ketamine hydrochloride
over 40 min
IV 0.05 mg/kg
midazolam over 40 min
Primary:
BSI Scores
Baseline scores:
Ketamine = 17.06 (SD = 1.819)
Midazolam = 16.6 (SD = 2.137)
At day 3:
Ketamine: 1.69 (SD = 1.93)
Midazolam: 3.42 (SD = 1.75)
MADRS-SI
Baseline scores:
Ketamine=3.65(SD=1.173)
Midazolam = 3.65 (SD = 1.268)
At day 3:
Ketamine=1.77(SD=1.84)
Midazolam = 3.52 (SD = 1.89)
Secondary:
MADRS Scores
Baseline scores:
Ketamine = 34.89 (SD = 8.04)
Midazolam = 34.19 (SD = 10.83)
- The improvement on MADRS score was
noticed on day 1 and continued on day 3,
but it was not observable day 7 following
treatment.
- There was also improvement in suicidal
ideations on BSI and MADRS-SI.
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
At day 3:
Ketamine: 25.09 (SD = 7.07)
Midazolam: 32.03 (SD = 7.21)
At 7 days: difference was insignificant.
Gamble et al.,
2018
IV Ketamine, 0.75 mg/kg
and remifentanil 1
mcg/kg
All patients received 8 ECT
sessions
IV Propofol 1 mg/kg and
remifentan il 1 mcg/k g
All patients received 8 ECT
sessions
Primary:
All patients in the ketamine arm achieved a
50% MADRS reduction compared with 10
(83%) in the Propofol arm.
Median interquartile range [IQR] number of
ECT treatments to achieve a 50% MADRS
reduction is 2 [14] for Ketamine and 4
[27] for propofol group.
Secondary:
- All patients in the ketamine group achieved
remission compared with 7 in the propofol
group 3 ECT treatments com par ed with 7
treatments for propofol.
- Patients in the ketamine group were more than
twice as likely to ach ie ve respo ns e on
MADRS [HR]: 3.20, 95% co nfid e nce
interval [(CI)]: 2.00 to 5.13 and also were
also twice as likely to achieve remission
(HR: 3.67, 95% CI: 2.13 to 6.32) compared
with the propofol arm.
- A comparable change in ALS scores were
reported among both groups.
-Mean CADSS scores were comparab le among
both groups.
- Time from anesthesia to discharge was similar
between groups, 63.5 (18.2) vs 63.3 (15.8)
minutes in the ketamine arm and propofol
arm, respectively.
- About 100% patients in Ketamine group
achieved 50% reduction in MADRS
compared to placebo.
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
Ghasemi et al.,
2013
IV Ketamine, 0.5 mg/kg
over 45 min, 3 infusions
for45minover3days
ECT on 3 test days (every 48 h) Primary:
BDI Sco res
Baseline scores:
Ketamine = 34.66 (SD = 10.7)
ECT = 42.44 (SD = 9.53)
After one-week post treatment
Ketamine:10.88 (SD = 7.49)
ECT: 15.66 (SD = 7.51)
HDRS Scores
Baseline scores:Ketamine = 30.22 (SD = 5.78)
ECT = 35.88 (SD = 6.47)
After one-week post treatment
Ketamine:9.55 (SD = 4.98)
ECT: 14 (SD = 4.9)
Secondary: NA
- There was a significant improvement in
patient receiving Ketamine compared to
placebo group within 24 h of first infusion.
- This improvement was observed throughout
the study after second treatment and 72 h
post-treatment.
- The effect size were moderate to large
throughout the study.
Haile et al., 2013 IV Ketamine, 0.5 mg/kg,
Single dose for 40 min
for 7 days
Midazolam 0.045 mg/kg Primary:
Seven patients receiving ketamine met
response criteria at day 7, whereas two
patients receiving midazolam met response
criteria.
Secondary: NA
- Participants taking Ketamine reported an
increased in plasma BDNF levels compared
to non-responsders.
- There was highly significant negative
correlation between MADRS and BDNF
levels with ketamine (240 min post
infusion).
-BDNFcanbeusedasperipheralbiomarker
for ketamine antidepressant response.
Hu et al., 2015 IV Ketamine, 0.5 mg/kg
Oral
Escitalopram = 10 mg/-
day
IV Normal saline (placebo)
Oral Escitalopram = 10 mg/day
Primary:
Change in MADRS scores
Baseline scores:
Ketamine = 32.3 (SD = 6.5)
Placebo = 36.5 (SD = 7.8)
Afterfourweeks:
Ketamine:14.0 (SD = 10.2)
Placebo: 18.1 (SD = 8.2)
Secondary:
- Response rate was 92.3% and 57.1% for
Ketamine + Escitalopram and placebo +
Ketamine group, respectively.
- For TRD, the response rate was 88.9% and
33.3% for Ketamine + Escitalopram and
placebo + Ketamine group, respectively.
- The remission rate was 76.9% and 14.3% for
Ketamine + Escitalopram and placebo +
Ketamine group, respectively.
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
Response:
At 4 weeks:
Ketamine vs Placebo: 92.3% vs 57.1%
Remission:
At 4 weeks:
Ketamine vs Placebo: 76.9% vs 14.3%
QIDS-SR
Baseline scores:
Ketamine = 16.5 (SD = 5.3)
Placebo = 17.5 (SD = 4.2)
Afterfourweeks:
Ketamine:7.8 (SD = 5.8)
Placebo: 10.0 (SD = 4.6)
QIDS- Suicide
Baseline scores:
Ketamine = 1.9 (SD = 0.7)
Placebo = 1.4 (SD = 0.6)
Afterfourweeks:
Ketamine:0.4 (SD = 0.9)
Placebo: 0.2 (SD = 0.4)
- Patients receiving Ketamine observed quicker
improvement than placebo.
Jafarinia et al.,
2016
Oral Ketamine 50 mg three
times daily
Diclofenac 50 mg three times
daily
Primary:
HDRS M ean difference: ketamine diclofenac
at week 3: 1.85 (0.48 to 4.18) and at week
6: (95% CI): 2.85 (0 .5 4 to 5.16)
HADS Mean difference for
ketamine-dicolfenac at week 3: ketamine-
diclofenac (95% CI): 1.05 (0.45 to 1.64),
and week 6:(95% CI): 0.75 (0.18 to 1.32)
Secondary:
Mean VAS score between keta min e and
diclofenac at week 3: 2.05 (8.27 to 12.37)
and week 6:(95% CI): 2.90 (18.07 to
12.27)
- A significant dec rease in HDRS scores was
observerd with ketamine compared to
placebo.
- However, both groups were comparable at
week 3.
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
Jarventausta K.
et al. 2013
IV Ketamine, 0.4 mg/kg IV Normal saline & Propofol. Primary:
Change in MADRS score:
S-ketamine = 26.9 ± 9.5
Normal saline & propofol = 27.1 ± 7.9
Secondary: NA
- There was a comparable reduction in
MADRS scores among both groups.
- The was no difference in speed of response
and number of ECT sessions.
Jiang M. , et al.,
2016
0.5 mg/kg (0.05 ml/kg)
ketamine was
given at induction of
anesthesia, followed by
0.25 mg/kg/h
(0.025 ml/kg/h)
continuous infusion for
30 min.
0.05 ml/kg Ketamine was used at
induction of anesthesia,
followedby0.025ml/kg/h
continuous infusion of saline
for 30 min.
Primary:
Difference in PHQ-9 scores for Ketamine
group preoperatively to pos tope ra tiv ely
(days 1 and 5): (t-valu e = 2.144,
F=14.67, P <0.01)
PHQ scores
Preoperation
Control = 3.63 ± 0.14
Ketamine = 4.02 ± 0.13
Postoperative day 5
Control = 2.92 ± 0.63
Ketamine = 2.28 ± 0.61
Secondary: NA
- There was a significant decrease in PHQ-9 of
Ketamine group compared to placebo at
postoperative day 1 and 5.
- BDNF levels were higher in Ketamine group
after surgery.
Tayyebi et al.,
2018
IV Ketamine Bolus and
infusions,
0.50.75 mg/kg
NA Primary:
AccordingtoHamiltonandBeckscore,the
treatment response in investigated patients
was 64% and 60%, respectively.
About 48% participants on HDRS and 44% on
BDI responded to treatment on injection
0.5 mg/kg compared to 80% on HDRS and
76% on BDI on injection 0.75 mg/kg/
Secondary: NA
- Abo ut 64% and 60% patients reported
improvement on HDRS and BDI,
respectively.
- For dose of 0.5 mg/kg of Ketamine, 48% and
44% reported response on HDRS and BDI,
respectively.
- For dose of 0.5 mg/kg of Ketamine, 80% and
76% reported response on HDRS and BDI,
respectively.
- The positive response was highest during first
two days and at week1.
Lai R. et al., 2014 IV Ketamine,
0.10.4 mg/kg
IV Normal Saline Primary:
Baseline MADRS score, 29, 27,29, 28
Three of four subjects achieved antidepressant
response (50% decrease in MADRS scores)
- For two patients, response was noticed at
0.1 mg/kg.
- For other two, response was observed at the
highest doses.
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
Secondary: NA - All patients relapsed within a week.
Lapidus et al.,
2014
Intranasal Ketamine,
50 mg/day
Intranasal Normal saline Primary:
Estimated mean difference in MARDS score
between ketamine and placebo was 7.6
(95% CI: 3.911.3)
Secondary: NA
- At 24 h, there was significant improvement
among Ketamine (44%) compared to
placebo (6%).
- Ketamine was associated at 40 min, 4 and
48 h.
- No difference was observed at 72 h or 7 days.
Lenze et al., 2016 IV Ketamine, 0.5 mg/kg NA Primary:
Baseline score for MADRS
Saline&40-min infusion group = 34.0 (3.8)
96- h infusion group = 31.9 (5.9)
50% reduction in MADRS:
4/10 in the 96-h group and 2/10 in the 40-min
group were respond ers at week 2; of these,
2/10 in the 96-h group and 1/10 in the
40-min group maintained response out to
week 8.
Secondary: NA
- Both groups reported a significant reduction
in depressive symptoms compared to
baseline.
- Higher Ketamine concentration was
associated with a better antidepressant
response.
Lo et al., 2016 Ketamine = 0.1 mg/kg,
increasing by 0.1 mg/kg
up to 0.5 mg/kg.
Ketamine was given in three
routes including IV, IM,
SC.
Midazolam 0.01 mg/kg Primary:
12/15 participants met the criteria for both
response and remission at least at one time
point during the trial (across all dose levels
and time points).
The overall acute response/remission rates
were75% for IV, 60% for IM, and 100% for
SC groups.
Secondary:
There was no evidence of treatment emergent
mania at any of study time point, across
routes of administration and doses.
There was no clinically significant change in
BPRS or CADSS was observed in the
- 12/15 participants achieved response and
remission at least at one time point during
the trial.
- The response and remission rates were 75%
(IV), 60% (IM) and 100 % (SC).
- Dose-dependent response but also led to
increased side effects.
- The mean time to relapse was 23.2 days for
all routine, 9 days for IV group, 11.7 days
for IM group, and 34.5 days for SC group.
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
midazolam compared to different routes of
ketamin e.
Murrough et al.,
2016
Single IV infusion of
Ketamine Hydrochloride
0.5 mg/kg
Single IV infusion of Midazolam
0.045 mg/kg
Primary:
Change in MADRS scores
Baseline scores:
Ketamine = 32.6 (SD = 6.1)
Placebo = 31.1 (SD = 5.6)
After 24 h:
Ketamine = 14.77, 95% CI (11.7317 .8 0)
Placebo = 22.72, 95% CI (18.8526.59)
Secondary:
- MADRS Response rate = Ketamine 64% VS
Placebo 28%
- CGI-I=Ketam ine 62% VS Placebo 24%
- CGI-S=Ketamine 53% VS Placebo 8%
QIDS scale
Baseline scores:
Ketamine = 16.6 (SD = 4.1)
Placebo = 16.3 (SD = 4.5)
After 24 h:
Ketamine = 8.38, 95% CI (6.7110.05)
Placebo = 11.78 , 95% CI (9.6313.92)
- The response rates were 64% for Ketamine
and 28% for placebo, respectively at 24 h
with number need to treat of 2.4.
- There was a small worsening of symptoms
over time but depression scores were lower
for Ketamine group.
Fava et al., 2018 IV Ketamine 0.1 mg/kg,
0.2 mg/kg, 0.5 mg/k and
1.0 mg/kg.
IV Midazolam 0.045 mg. Primary:
Pairwise comparisons of all ketami ne group
to midazolam on HAM-D-6 at day 3
1.87, 95% CI (4.14, 0.41)
Pairwise comparisonsofeachketamine
group to midazolam on HAM-D- 6 at day
3
0.1 mg/kg = 2. 0 4, 95% CI (5. 0 4, 0.95)
0.2 mg/kg = 0. 3 6, 95% CI (3. 1 8, 2.46)
0.5 mg/kg = 3. 2 1, 95% CI (5. 9 7, 0.44)
1.0 mg/kg = 1. 8 4, 95% CI (4. 6 5, 0.96)
- IV Ketamine was superior to active placebo
within 72 h of treatment.
- The dose of 0.5 mg and 1.0 mg/kg was
superior to placebo and lower doses of
ketamine.
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
Secondary:
Statistical significance of the group*time
interaction effect was significant for only for
the SDQ (p = 0.0105) and the PAS
(p = 0.0341) in the 5-group comparison, and
the PAS (p = 0.0332) and the CGI-S
(p = 0.0204) in the 2-group comparison.
Fernie et al., 2017 IV Ketamine up to 2 mg/kg
All partic ipa nts received
ECT.
IV Propofol up to 2.5 mg/kg
All participants received ECT.
Primary:
Change in HDRS scores
Baseline severity
Ketamine = 27.19 (6.47)
Propofol = 24.79 (8.50)
After one month post-ECT
Ketamine = 14.08 (8.08)
Propofol = 12.08 (9.86)
Change in MADRS scores
Baseline severity
Ketamine = 36.38 (8.29)
Propofol = 35.68 (8.39)
After one month post-ECT
Ketamine = 17.85 (13.15)
Propofol = 17.15 (13.75)
Secondary:
CANTAB SRM
Baseline severity
Ketamine = 0.71 (0.11)
Propofol = 0.72 (0.15)
After one month post-ECT
Ketamine = 0.70 (0.11)
Propofol = 0.65 (0.12)
ketamine as an anaesthetic agent for ECT was
not associated with acceleration of the
antidepressant effect of ECT
Daly et al., 2017 Intranasal Esketamine
28 mg, 56 mg, 84 mg.
Intranasal placebo Primary:
Mean difference from placebo
Esketamine 28 mg: 4.2 (2.09)
- There was dose-dependent statistically sig-
nificant treatment among all treatment
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
Esketamine 56 mg: 6.3 (2.07
Esketamine 84 mg: 9.0 (2.13)
Secondary:
Baseline and last day
groups compared to placeb o after a week of
treatment.
- Duration of effectiveness with 28 mg dose
was shorter.
Grunebaum et al.,
2018
IV Ketamine 0.5 mg/kg IV midazolam 0.02 mg/kg in
100 ml normal saline
Primary:
Average SSI score at day 1:
Difference between Ketamine to
Midazolam = 4.96 (95% CI 2.33 to 7.59)
Secondary:
- The proportion of responders on SSI was 55%
after ketamine compa re d to Midazo lam at
day 1.
- POMS Scale improvement after ketamine
compared with
midazolam = (Estimate = 21.19 (95%
CI = 2.95 to 39.43).
- Depression = Estimate = 7.65 (95% CI = 1.36
to 13.94)
- Change in HDRS-17 = Estimate = 2.83 points
(95% CI = 0.12 to 5.77).
- Change in HDRS-24 = 3.54 points (95%
CI = 0.29 to 7.36)
Self-rated BDI = 4.66 points (95% CI = 0.0 4
to 9.36).
A single Ketamine infusion resulted in a
greater reduction in suicidal ideations at day
1 compared to active placebo. The Ketamine
groups showed a greater improvement for
depression compared to active placebo.
Kudoh et al., 2002 Group A: Single IV infusion
of 1.0 mg/kg of
Ketamine, 1.5 mg/kg of
Propofol, and 2 μg/kg of
Fentanyl
Group B: Single IV infusion of
1.5 mg/kg of Propofol and 2
μg/kg of Fentanyl
Primary:
Change in HDRS scores
Baseline scores:
Group A = 12.7 ± 5.4
Group B = 12.3 ± 6.0
Group C = 4.2 ± 1.7
After surgery:
Group A = 9.9 ± 4.1
Group B = 14.4 ± 3.8
Small-dose ketamine not only improved the
depressive symptoms postoperatively but
alsoreducedpainindepressedpatientswho
underwent orthopedic Surgery. Depressed
mood, suicidal tendencies, somatic anxiety,
and hypochondriasis significantly decreased
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
Group C = 4.8 ± 1.6
Secondary:
Postoperative pain scores in Group A at 8 and
16 h after the anesthetic effect were
significantly lower than those in Group B.
There were no significant differences in the
other postoperative pain scores between two
groups for 4 days after su rger y . P
Postoperative pain scores for Group C at 8, 16,
24, 48, and 72 h were 20.2 8.4, 18.4 7.5,
11.9 6.1, 8.7 5.5, and 5.2 3.2 ,
respectively after the end of anesthesia.
Salehi et al., 2015 IV Ketamine 0.8 mg/kg
Participants received a total
of eight ECT sessions.
IV Sodium thiopental
1mg/kg-1.5mg/kg
Participants received a total of
eight ECT sessions.
Primary:
Change in HMDRS scores
Baseline scores:
Ketamine = 29.82 ± 7.3
Sodium thiopental = 28.86 ± 7.6
After session#8:
Ketamine = 8.32 ± 5.17
Sodium thiopental = 10.53 ± 7.87
Secondary:
Systolic and diastolic blood pressure have
statistically significant difference between
ketamine and sodium thiopental in all
sessions .
The depression score s improved after each
ECT session but improvement was faster in
Ketamine group.
Singh et al., 2016 Intravenous
Ketamine (0.5 mg/kg) two
or three times weekly
Intravenous
placebo (0.9% Sodium chloride
for injection) two or three
times weekly
Primary:
Change in MADRS score
Ketamine twice daily: 218.4 (SD = 12.0)
Placebo = 25.7 (SD = 10.2)
Ketamine three times daily = 217.7 (SD = 7.3)
Placebo = 23.1 (SD = 5.7)
Secondary:
Ketamine was effective at both frequencies for
depression compared to placebo.
The mean difference in MADRS scores was
comparable among both groups.
Onset of antidepressant was noticed within first
week and was maintained through day 15
for both Ketamine group compared to
placebo.
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
The total change in MADRS score from
baseline to day 29 for both ketamine
groups
Ketamine twice a daily = 221.2 (SD = 12.9)
Placebo = 24.0 (SD = 9.1)
Ketamine three times daily = 221.1 (SD = 11.2)
Placebo = 23.6 (SD = 6.6)
MADRS response at day 15
Ketamine twice a daily vs placebo = 68.8% vs
15.4%
Ketamine three times a daily vs placebo = 6.3%
vs 53.8%
MADRS remission at day 15
Ketamine twice a daily vs placebo = 7.7% vs
37.5%
Ketamine three times a daily vs placebo = 0%
vs 23.1%
MADRS onset of response in week 1
Ketamine twice a daily vs placebo = 6.3% vs
38.9%
Ketamine three times a daily vs placebo = 0%
vs 4%
During the 2-week open-label ketamine phase,
the mean CGI-S and PGI-S scores were
similar in both groups
The treatment response was similar among
both groups during open-label phase.
Singh et al., 2016 Single IV Esketamine
0.20 mg/kg, or single IV
Esketamine 0.40 mg/kg
over 40 mins
Single IV infusion of placebo
(.9% saline solution) Over
40 mins
Primary: Change in MADRS score
compared to placebo
Placebo = 23.8 (SD = 2.97)
Esketamine .20 mg/kg = 216.8 (SD = 3.00)
Esketamine .40 mg/kg = 216.9 (SD = 2.61)
Secondary:
The total change in MADRS score from
baseline to day 3
- The depression scores improved for both
groups taking Esketamine groups compared
to plac ebo.
- The proportion of responders were 67% and
64% for both Esketamine groups. There
were no responders among placebo group.
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
Placebo = 2. 3 (SD = 3.38)
Esketamine .20 mg/kg = 16.3 (SD = 3.45)
Esketamine .40 mg/kg = 13.4 (SD = 3.03)
Responders to first dose
Esketamine 0.2 mg/kg = 67%
Esketamine 0.4 mg/kg = 64%
Placebo = 0%
Change in CGI-S score
Placebo = 0. 2 (SD = 0.63)
Esketamine .20 mg/kg = 1.3 (SD = 1.5)
Esketamine .40 mg/kg = 1.4 (SD = 1.36)
Change in PGI-S score Placebo = 0.3
(SD = 0.48)
Esketamine .20 mg/kg = 0.8 (SD = 0.97)
Esketamine .40 mg/kg = .6 (SD = 0.82)
Sos et al., 2013 Single IV loading dose of
0.27 mg/kg followed by
maintenance dose of
0.27 mg/kg
Single IV infusion of placebo
(.9% saline solution)
Primary:
Change in MADRS total score
Day 1 = 5.7 (95%CI 3.47.9)
Day 4 = 4.7 (95%CI 2.57.0)
Day 7 = 4.0 (95% CI 1.86.2
Secondary:
Response rates at day 7
Ketamine = 40.7%
Placebo = 11.1%
Ketamine serum levelsAfter
10 min = 306 ± 136 ng/ml
After 30 min =237 ± 95 ng/ml
Ketamine metabolite levels
After 10 min = 11 ± 7 ng/ml
After 30 min = 50 ± 21 ng/m..
There were no differences found between
responders and non-responders in ketamine
and/or nor-ketamine serum levels.
There was a reduction in the core symptoms of
depression in the following week with
maximum on day seven.
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
Su et al., 2017 IV Ketamine = 0.2 mg/kg,
0.5 mg/kg.
Single IV infusion of placebo
(saline solution)
Primary: HAM-D Scores
Baseline severity (Estimated Least Square
Means):
Placebo = 1.32
Ketamine 0.2 mg/kg = 1.32
Ketamine 0.5 mg/kg = 1.32
At 28 days (Estimated Least Square Means):
Placebo = 1.11
Ketamine 0.2 mg/kg = 0.91
Ketamine 0.5 mg/kg = 0.96
Secondary:
Response rates
Ketamine 0.5 mg/kg = 45.8%
Ketamine 0.2 mg/kg = 39.1%
Placebo = 12.5%
Response Rate by BDNF Val66Met
Genotype
Val/Val = 17%
Val/Met = 56.3%
Met/Met = 26.8%
There was no differences betwe en carriers of
the Met allele and Val/Val patients (re-
sponder rate: 33.9% vs 25.0%).
BDNF genotype is not a significant predictor of
the outcome.
The dose of 0.5 mg/kg was more effective
compared to 0.2 mg/kg and placebo.
There was no significant difference between
carriers of the Met allele and Val/Val pa-
tients in efficacy for Ketamine
Wang et al., 2012 Single IV dose of
0.8 mg/kg, Ketamine or
Single IV dose of
0.8 mg/kg Ketamine plus
1.5 mg/kg Propofol
All partic ipa nts received
ECT.
Single IV dose of 1.5 mg/kg
Propofol
All participants received ECT.
Primary: Change in HDRS scores
Baseline severity
Ketamine & propofol = 28.00 ± 4.88
Ketamine = 27.00 ± 3.93
Propofol = 28.16 ± 2.48
After 7 days
Ketamine & propofol = 6.94 ± 1.57
Ketamine = 6.75 ± 0.96
The HDRS scores improved quickly in
ketamine group and Ketamine and Propofol
group compared to Propofol only group.
The improvement in depression scores were
significantly greater in ketamine group and
Ketamine and Propofol group compared to
Propofol only group.
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
Propofol = 11.08 ± 2.78
Secondary:
NA
Xu et al., 2017 IV Ketamine 0.5 mg/kg. Isofibrillar saline Primary:
Change in HAMD scores
Baseline severity
Ketamine = 18.82 ± 2.82
Control group = 18.55 ± 3.21
After 7 days
Ketamine = 17.36 ± 6.25
Control group = 13.45 ± 5.21
Secondary:
NA
The HAMDS scores improved at day 1, 3 and
7 compared to placebo.
However, this change was not statistically
significant.
Xu et al., 2017 Single IV dose of
0.25 mg/kg Ketamine
Single IV infusion of placebo
(.9% saline solution)
Primary:
EDRS score s 3 days postpa rtum
Ketamine = 7.2 ± 3.9
Saline = 7.2 ± 4.2
EDRS scores 6-weeks postp ar tu m
Ketamine = 5.6 ± 3.9
Saline = 5.7 ± 4.3
Occurrence of postpartum depression
Ketamine = 16%
Saline = 17.8%
Secondary:
NRS scores 3 days postpartumKetamine = 4
(07)
Saline = 4 (08)
NRS scores 6-weeks postpartum
Ketamine = 1 (08)
Saline = 2 (09)
No significant differences were found in the
prevalence of postpartum depression
Yoosefi et al.,
2014
IV infusion of ketamine
1 mg/kg or 2 mg/kg,
IV infusion of thiopental
2 mg/kg or 3 mg/kg,
Primary:
Change in HAMD scores
Baseline severity
- There was significant in depression scores
among Ketamine group before second ECT
session compared to Thiopental.
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
ECT sessions were carried
out 3 times a week for a
total of 6 sessions over
3weeks
ECT sessions were carried out 3
times a week for a total of 6
sessions over 3 weeks
Ketamine = 23.60
Thiopental = 22.8 6
After 4 days
Ketamine = 17.07
Thiopental = 17.2 9
Secondary:
Change in MMSE scores
Baseline severity
Ketamine = 25.60
Thiopental = 24.7 9
After 4 days
Ketamine = 27.87
Thiopental = 25.2 9
Seizure parameters
Ketamine seizure duration = 27.47 to 31.87 s
Ketamine electrical dose = 28.67 to 36
Thiopental seizure duratio n = 19. 27 to 25.5 7 s
Thiopental electrical dose = 36.43 to 62.86
In both groups, baseline values of heart and
Mean Arterial Pressure were not shown to
be significantly different.
- At the end of study, both groups observed an
improvement in depre ssio n scores.
Zarate et al., 2006 Single IV dose of 0.5 mg/kg
Ketamine in two phases
Single IV infusion of placebo
(saline solution)
Primary:
Change in HDRS scores
Ketamine = 56.2 ± 20.4
Placebo = 9. 8 ± 20.1
Secondary:
BPRS positive symptoms subscale scores for
participants receiving Ketamine
compared to placebo
Drug, F1,200 = 4.23; P =.04;time,
F8,200 = 9.31; P.001; drug time,
F8,200 = 6.89; P.001)
Ketamine was effective for depression
compared to placebo. This benefit was
maintained for a week after the Ketamine
dose.
Psychiatric Quarterly
Table 1 (continued)
Study Intervention group Comparison group Mean change in primary outcome measures Summary of results.
YMRS scores for participants receiving
ketamine compare d to placebo
Days 1 to 2 (drug, F1,201 = 3.08; P =.08;time,
F8,201 = 3.54; P.001; drug time,
F8,201 = 4.68; P.001
Response rates
Ketamine = 71%
Placebo = 0%
Remission rates
Ketamine = 35%
Placebo = 0%
BHS, Beck Hopelessness Scale; BSI, Beck Suicidality Ideation; ECT, Electroconvulsive Therapy; HAM-D, Hamilton Depression Rating Scale; HRDS, Hamilton Depression Rating
Scale; IDS-CR, Inventory of Depressive SymptomatologyClinician Rated; MADRS, Montgomery-Åsberg Depression Rating Scale; MADRS-SI, Montgomery- Asberg Depression
Rating Scale; MDD, Major Depressive Disorder; MoCA, Montreal Cognitive Assessment; PHQ-9, Patient Health Questionnaire; QIDS-SR, Quick Inventory of Depressive
Symptomatology Self-Report; SAFTEE, Systematic Assessment for Treatment Emergent Effects; TRD, Treatment Resistant Depression; WMS-RC, Wechsler Memory Scale-
Chinese Revision
Psychiatric Quarterly
Table 2 Baseline characteristics of patients in the included trials
Study Groups Duration Sample
[n]
Age [years]
[Mean/Med ia n]
Single infusion vs
multiple infusion
Dose range (mg)
Alizadeh et al., 2015 Ketamine & Propofol NA 22 34.27 ± 10.66 Multiple 0.3 mg/kg
Saline & Propofol 20 35.1 ± 12.44 0.3 mg/kg
Arabzadeh et al., 2018 Ketamine 6 weeks 41 34.31 ± 6.73 NA (oral dosage) 50 mg/day
Placebo 40 33.72 ± 8.34 50 mg/day
Burger et al., 2016 Ketamine 2 weeks 3 28 Single 0.2 mg/kg
Placebo 7 27 0.2 mg/kg
Canuso et al., 2018 Esketamine 4 weeks 35 35.7 ± 13.40 NA (intranasal) 84 mg
Placebo 31 36.0 ± 12.82 84 mg
Carspecken et al., 2018 Ketamine Ketamine = 46 weeks
Methohexital = 53 weeks
23 50 ± 12 Single 12mg/kg
Methohexital 27 47 ± 12 12mg/kg
Chen et al., 2017 Ketamine 4 weeks 63 40.94 ± 15.41 Multiple infusions 0.3 mg/kg
Control 64 37.44 ± 14.16 0.3 mg/kg
Fan et al., 2017 Ketamine 7 days 20 46.75 ± 14.04 Single 0.5 mg/kg
Midazolam 17 44.65 ± 15.1 0.5 mg/kg
Fernie et al., 2017 Ketamine One month 20 51.76 ± 9.97 Multiple infusions Up to 2 mg/kg
Propofol 20 49.88 ± 12.53 Up to 2.5 mg/kg
Gamble et al., 2018 Ketamine 4 weeks 12 42 ± 16 Single infusion vs
multiple
infusion
0.75 mg/kg
Propofol 12 46.5 ± 16 0.75 mg/kg
Ghasemi et al., 2013 Ketamine 7 days 9 35.22 ± 13.63 Multiple infusions 0.5 mg/kg
ECT 9 40 ± 16.41 0.5 mg/kg
Haile et al., 2013 Ketamine 1 week 15 48.53 ± 3.30 Single 0.5 mg/kg
Midazolam 7 42.71 ± 4.85 0.5 mg/kg
Hu et al., 2015 Ketamine 4 weeks 13 36.7 ± 14 Single infusion vs
multiple
infusion
0.5 mg/day
Placebo 1 4 4 1 ± 11.1 0.5 mg/day
Jafarinia et al., 2016 Ketamine 6 weeks 20 40.7 ± 8.71 NA 150 mg
Diclofenac 20 38.95 ± 9.22 150 mg
Ja¨rventausta K. et al.
2013
S-Ketamine 2 weeks 16 48.4 Multiple 0.4 mg/kg
Propofol & normal saline 16 53.7 0.4 mg/kg
Jiang M., et al., 2016 Ketamine 5 days 60 43.38 ± 0.95 NA 0.250.5 mg/kg
Control 60 41.40 ± 0.16 0.250.5 mg/kg
Psychiatric Quarterly
Table 2 (continued)
Study Groups Duration Sample
[n]
Age [years]
[Mean/Med ia n]
Single infusion vs
multiple infusion
Dose range (mg)
Tayyebi et al., 2018 Bolus Ketamine 2 months 25 40.84 ± 11.75 Single 0.5 mg/kg
(Bolus)
25 42.84 ± 12.17 0.75 mg/kg
(Bolus)
Infusion Ketamine 25 39 ± 11.49 0.5 mg/kg
(Infusion)
25 39.72 ± 10.18 0.75 mg/kg
(Infusion)
Lai R. et al., 2014 Ketamine 5 weeks 4 NA Multiple 010. 4 mg/kg
Normal saline NA
Lapidus et al., 2014 Ketamine 2 weeks 18 48.0 ± 12.8 Single Intranasal Ketamine
50 mg/day
Placebo Intranasal 0.9%
saline (Placebo)
Lenze et al., 2016 96 h infusion 8 weeks 10 44.6 ± 12.8 Single 0.6 mg/kg/h
Saline for first 95 h
and 20 min followed
by 0.5 mg/kg
Ketamine
for final 40 min
10 42.5 ± 13.8 Saline, 40-min
Ketamine at 0.5
mg/kg
Lo et al., 2016 Ketamine 7 days 11 48.5 ± 11 Multiple infusions 0.1 mg/kg - 0.5 mg/kg
Midazolam 3 0.01 mg/kg
Murrough et al., 2016 Ketamine 7 days 47 46.9 ± 12.8 Single 0.5 mg/kg
Midazolam 25 42.7 ± 11.6 Single 0.045 mg/kg
Fava et al., 2018 Ketamine 30 days 80 43.1 ± 11.9
45.5 ± 14.6
48.6 ± 12.9
47.4 ± 10.1
Single 0.1 mg/kg
0.2 mg/kg
0.5 mg/kg
1.0 mg/kg
Midazolam 19 45.6 ± 13.8
Daly et al., 2017 Esketamine 28, 56,
84 mg
130 days 34 44.7 [10.0] Multiple doses Esketamine 28, 56,
84 mg
Placebo 33 Placebo
Psychiatric Quarterly
Table 2 (continued)
Study Groups Duration Sample
[n]
Age [years]
[Mean/Med ia n]
Single infusion vs
multiple infusion
Dose range (mg)
Grunebaum et al., 2018 Ketamine One day 40 40.7 ± 13.1 Single infusion 0.5 mg/kg
Midazolam 40 38.4 ± 13.2 0.02 mg/kg
Kudoh et al., 2002 Gro up A Pro pofo l,
Fentanyl, Ketamine
1 week 35 46.9 ± 8.8 Single infusion 1.0 mg/kg of ketamine,
1.5 mg/kg of propofol,
and 2 g/kg of
fentanyl
Group B Propofol,
Fentanyl
35 48.2 ± 7.4 1.5 mg/kg of propofol
and 2 g/kg of
fentanyl
Group C Propofol,
Fentanyl, Ketamine
20 46.2 ± 10.3 1.0 mg/kg of ketamine,
1.5 mg/kg of propofol,
and 2 g/kg of
fentanyl
Salehi et al., 2015 Ketamine 8 session 80 Not mentioned Multiple infusions 0.8 mg/kg
Sodium thiopental 80 Not mentioned 11.5 mg/kg
Singh et al., 2016 Ketamine 15 days 35 BID = 45.7 ± 9.6
TID = 43.3 ± 12.0
Multiple infusions 0.5 mg/kg twice daily
VS 0.5 mg/kg thrice
daily
Placebo 3 3 B ID = 40.3 ± 11.8
TID = 46.1 ± 10.5
Singh et al., 2016 Esketamine 4 days 20 0.2 mg/kg = 44.7 ± 13.38
0.4 mg/kg = 41.8 ± (11.63)
Multiple infusions 0.2, 0.4 mg/kg
Placebo 10 42.7 ± (10.89)
Sos et al., 2013 Ketamine 2 weeks 11 42.2 ± 15.1 Multiple infusions 0.5 mg/kg
Placebo 19 44.6 ± 10.9
Su et al., 2017 Ketamine 2 weeks 47 0.2 mg/kg = 45.0 ± 12.3
0.5 mg/kg = 48.5 ± 11
Single infusion 0.2, 0.5 mg/kg
Placebo 24 48.6 ± 8.2
Wang et al., 2012 Ketamine One week 16 56.2 ± 11.5 Single infusion 0.8 mg/kg
Ketamine & Propofol 16 58.6 ± 16.3
Propofol 16 53.8 ± 15.2
Xu et al., 2017 Ketamine 1 week 50 42.36 ± 7.28 Single infusion 0.5 mg/kg
Psychiatric Quarterly
Table 2 (continued)
Study Groups Duration Sample
[n]
Age [years]
[Mean/Med ia n]
Single infusion vs
multiple infusion
Dose range (mg)
Saline 43.27 ± 6.6
Xu et al., 2017 Ketamine 6 weeks 165 31 ± 4 Single infusion 0.25 mg/kg
Saline 165 32 ± 4
Yoosefi et al., 2014 Ketamine 6 ECT sessions over
2 weeks with 3 sessions every
week
16 40.87 Multiple infusions 1 to 2 mg/kg
Thiopental 15 47 2 to 3 mg/kg
Zarate et al., 2006 Ketamine One week 9 46.7 ± 11.2 Single infusion one
week a part in
cross-over study
0.5 mg/kg
Placebo 9
Study Male
[%]
Adjunct Medications (Mention the percentage
taking medications if any and what are those medications)
Common side effects Site of trial
Alizadeh et al., 2015 6 (27%) Patients were prescribed medications but names
were not mentioned.
No side effects were reported Iran
7(35%)
Arabzadeh et al., 2018 26 (63.4%) Sertraline Abdominal pain, nau sea, tremor, and
dissociation. Comparable side effects
between groups.
Iran
24 (60%)
Burger et al., 2016 2 (67%) Not mentioned No side effects were reported. USA
5(71%)
Canuso et al., 2018 13 (37.4%) Standard antidepressant started at day1, titrated
during 2 weeks and continued. If alre ady
on ADT, continued.
Nausea, dizziness, dysgeusia, dissociation,
headache, vomiting, anxiety, paresthesia,
sedation, somnolence, euphoric mood,
vertigo.
USA
10 (32%)
Carspecken et al., 2018 17 (74%) All patients received ECT sessions. Continued
use of ADTs.
Not reported. USA
24 (89%)
Chen et al., 2017 21 (33.3%) All patients receiv ed 12 ECT session
(three per week)
No diff eren ce in side effects among
both groups.
USA
23 (35.9%)
Fan et al., 2017 8 (40%) NA Not reported. China
4(23.5%)
Fernie et al., 2017 18 (45%) No restrictions were placed on the psychiatric
medications and or treatments prescribed
Not reported. Scotland
Psychiatric Quarterly
Table 2 (continued)
Study Male
[%]
Adjunct Medications (Mention the percentage
taking medications if any and what are those medications)
Common side effects Site of trial
either before or during the course of the trial.
All participated received ECT.
Gamble et al., 2018 6 (50%) All patients received 8 ECT sessions Hypertension, hypotension, nausea,
vomiting headache
Canada
6(50%)
Ghasemi et al., 2013 4 (44.4%) All patients received ECT sessions. Temporary non-significant increase
in pulse and systolic blood pressure.
Iran
444.4%)
Haile et al., 2013 Not mentioned Single dose for 40 min. Not reported. USA
Not mentioned
Hu et al., 2015 6 (46.2%) All patients received escitalopram 10 mg/day Nightmares, restlessness, dizziness,
nausea, headache and increasing salivation
China
4(28.6%)
Jafarinia et al., 2016 5 (25%) None Transient loss of appetite, blurred vision,
tremor, abdominal pain
Iran
5(25%)
Ja¨rventausta K. et al. 2013 8 (50%) Antidepressant, antipsychotic, antidepressants +
antipsychotics, benzodiazepines
Both groups received ECT treatment.
Posttreatment disorientation and
restlessness.
USA
5(31.2%)
Jiang M., et al., 2016 34 (57%) None Side effects were comparable among
both groups.
China
33 (55%)
Tayyebi et al., 2018 15 (60%) None Side effects were not repo rted. Iran
14 (56%)
11 (44%)
10 (40%)
Lai R. et al., 2014 2 (50%) Alprazolam, olanzapine, quetiapine, tranylcypromine Transient tachycardia, and hypertension. UK
Lapidus et al., 2014 10 (50%) Names of dugs not mentioned. Small increase in psychosis, dislocation,
and systolic blood pressure. Other side
effects were feeling strange, poor
memory, and weakness or fatigue.
USA
Lenze et al., 2016 2 (20%) Clonidine, SSRI , SNRI agents, aripiprazole Mild and transient side effects were
reported for blood pressure.
USA
4(40%)
Lo et al., 2016 4 (26.6%) Patients were prescribed psychotropic
medications but names were not
mentioned.
Mild depersonalization, derealization,
altered body and time perception.
Australia
Not mentioned
Psychiatric Quarterly
Table 2 (continued)
Study Male
[%]
Adjunct Medications (Mention the percentage
taking medications if any and what are those medications)
Common side effects Site of trial
Transient increase in pulse, systolic
and diastolic blood pressure.
Fatigue, light-headedness, dizziness,
blurred vision, dry mouth and emotional
liability
Murrough et al., 2016 21 (45%) Zolpid em Dizziness, blurred vision, headache, nausea
or vomiting, dry mouth, poor coordination,
poor concentration, and restlessness.
USA
14 (56%)
Fava et al., 2018 42 (52.5%) Benzodiazepine
Non- benzodiazepine hypnotics
SSRIs
SNRIs
TCAs
Bupropion
Mirtazap in e
Vortioxetine
High systolic and diastolic blood pressure
among participants in ketamine groups.
USA
8(42.1%)
Daly et al., 2017 29 (43.3%) Participants continued their existing
antidepr e s s an t treatme nt during the study
Dizziness, headache, and dissociative
symptoms
USA, Belgium
Grunebaum et al., 2018 18 (45%) Antidep res sants , anticonvulsants,
antipsychotics, benzodiazepines, lithium
(above medications were stopped at
least 24 h pre-infusion)
Four patients with suicide attempts
(3 after and 1 before study procedures),
and 3 inpatient admissions for suicidal
ideations.
Transient increase in blood pressure.
USA
14 (35%)
Kudoh et al., 2002 Not mentioned All depressed patients were medicated by
antidepressants for more than a year
Ventricular ectopic rhythm Japan
Salehi et al., 2015 37 (46.2%) Not mentioned Increased blood pressure, headache, nausea,
and fear with the illusion of awaken ing s
Iran
37 (46.2%)
Singh et al., 2016 11 (31.4%) Patients continued any antidepressant medications
they were receiving at screening, at the same
stable dosages throughout the study. The
antidepressants most commonly used (>10%
of patients in each treatment group) at baseline
Headache, anxiety, dissociation, nausea,
and dizz iness
USA
11 (31.4%)
Psychiatric Quarterly
Table 2 (continued)
Study Male
[%]
Adjunct Medications (Mention the percentage
taking medications if any and what are those medications)
Common side effects Site of trial
were fluoxetine, citalopram, and bupropion;
these agents were continued throughout the study.
Singh et al., 2016 8 (40%) Not mentioned The most common side effects were nausea
and headache for Esketamine 0.2 mg/kg
and headache, dissociation , and nau sea
for Esketamine 0.40 mg/kg.
Belgium, Germany, Poland
4(40%)
Sos et al., 2013 5 (45.5%) Dissociation, perceptual disturbances,
confusion, mild increases in blood
pressure, emotional blunting and
euphoria.
Czech Republic
10 (52.6%)
Su et al., 2017 9 (19.1%) Not mentioned Increased in systolic blood pressure Taiwan
9(37.5%)
Wang et al., 2012 6 (50%) SSRI, TCA, Atypical antipsychotics,
Benzodiazep ine s
Hypertension during the ECT session,
angialgia at the site of injection of
the anesthetic and sense of fear upon
awakening from anesthesia
USA
7(58%)
5(42%)
Xu et al., 2017 0 Not reported Not reported. China
0
Xu et al., 2017 0 Single Dizziness, drowsiness, diplopia,
hallucinations, headache, and
vomiting
China
0
Yoosefi et al., 2014 7 (50%) Not mentioned Increase in BP 10 min after induction
among Ketamine group
Iran
8(53.3%)
Zarate et al., 2006 0 Participants did not receive these
medications during the length of study.
Perceptual disturbances, confusion,
elevations in blood pressure,
euphoria, dizziness, and increased libido
USA
ADT, antidepressant treatment, ECT-Electroconvulsive therapy; NA, Not Applicable; SNRI, Selective Norepinephrine Re-Uptake Inhibitor; SSRI, Selective Serotonin Re-Uptake
Inhibitor
Psychiatric Quarterly
Target Population
In this review article, 35 studies were assessed for effectiveness in treating clinical depression,
while Ketamine was used for the prevention of postpartum depression in one study [18]. The
inclusion criterion was TRD (n = 13) and moderate to severe MDD (n = 22). Ketamine was
also used as anesthetic and antidepressant in participants who underwent orthopedic surgery
and unilateral mastectomy [27,28]. In one study, participants were newly diagnosed with
breast cancer [29]. Another study focused on prevention of depression in pregnant patients
undergoing elective cesarean section [26].
Scales for Outcome Measures
The primary outcome was measured using various rating scales. The breakdown of these
measure was: Hamilton Depression Rating Scale (HDRS) (n = 22), MontgomeryÅsberg
Depression Rating Scale (MADRS) (n = 18), Beck Depression Inventory (BDI) (n =5), and
Patient Health Questionnaire (PHQ) - 9 (n =3).
Dose of Ketamine
In 32 studies, Ketamine was used in the treatment arm whereas three studies used
Esketamine as the active agent. It was administered as Intravenous (IV) infusion in 30
studies, intranasal in two studies, a nd orally in three studies. One study administered
Ketamine through a subcutaneous route. Intravenous Ketamine was used in varying
dosages ranging from 0.1 mg/kg to 1 mg/kg day in 29 studies. Three studies used a dose
range of up to 2 mg/kg. Intranasal Ketamine was administered between dose ranges of 28
to84mg.DosageoforalKetaminerangedfrom50mg/dayto50mgthreetimesaday.
Studies comparing the dose-d ependent response found a greater response at doses o f
0.5 mg/kg to 1 mg/kg IV Ketmaine [24,30,31] whereas Singh et al., 2016 found similar
response for doses of 0.2 mg/kg and 0.4 mg/kg [17].
Change in Depression and Related Outcome Measures
IV Ketamine was used in 27 studies, with 19 studies reporting significant improvement
compared to the control or placebo group of participants with MDD. One study com-
paring Ketamine at 0.5 mg/kg to ECT reported similar improvement among both g roups
[32]. However, the antidepressant benefits lasted up to 72 h on BDI scale and one week
on HDRS scale after the last infusion of Ketamine. Among studies with favorable
responses, two studies used Ketamine as an anesthetic agent in patients receiving ECT
treatments. Eight studies reported lack of improvement with Ketamine compared to the
control gro up. In six of th ese studies, Ketamine was used as an antidepressant in patients
receiving ECT treatment [15,3337]. Common reasons for lack of efficiency were cited
as ECT ceiling treatment effect and ECT blunting the response to Ketamine. Other
reasons include inadequate sample size [38], trauma from breast cancer and surgical
treatment of breast cancer [26]. It is noteworthy that Ketamine resulted in faster im-
provement [36]. Intravenous Ketamine was ineffective for prevention of postpartum
depression [18].
Psychiatric Quarterly
Intranasal Ketamine was administered in two studies at doses of 50 mg [39]and 84 mg [40]
with improvement in both studies. Oral Ketamine at doses of 50 mg/day [41] and 50 mg three
times a day [42] reporting significant improvement in both studies. Loo and colleagues (2016)
compared Ketamine administered intravenously, intramuscularly, and subcutaneously to the
control group. About 75% of patients with IV Ketamine, 60% with IM Ketamine, and 100%
with SC Ketamine reported improvement in depression scores [9].
In a study, an increase in BDNF level after administration of Ketamine had a negative
correlation with depression [43]. Similar results were reported in a study comparing Ketamine
and ECT to ECT and methohexital group, indicating BDNF as a potential biomarker for
antidepressant response [31]. However, BDNF genotype was not indicated as a predictor of
response.
Side Effects
No side effects were reported in nine RCTs. The most common side effects were fatigue,
nausea, vomiting, transient increase in blood pressure, anxiety, confusion, dissociation, dizzi-
ness, and drowsiness. Other side effects were diplopia, emotional blunting, euphoria, head-
ache, increased libido, perceptual disturbances, paraesthesia, sedation, and vertigo. Ventricular
ectopic rhythm was reported in one study [28]. One study reported suicidal ideations and
attempt [10].
Quality Assessment of RCTs
Random sequence generation was at low risk among 26 studies and allocation concealment
among 26 RCTs. Frequency of studies reporting a low risk across other domains of Cochrane
risk of bias tool was: blinding of outcome assessors (n = 30), blinding of participants and
personnel (n = 27), attrition bias (n = 27), other sources of bias (n = 30), and selective reporting
(n = 34). A total of 10 studies were rated as having a high risk of overall bias i.e. 3 matrices
of risk of bias tool were rated as having unclear or high risk of bias for these studies. Figure 2.
presents a clustered bar chart exhibiting frequencies of high, unclear and low risk bias across
all matrices of Cochrane risk of bias tool. Figure 3. presents study-wise risk of bias across all
matrices of Cochrane risk of bias tool.
Fig. 2 Risk of Bias Graph
Psychiatric Quarterly
Fig. 3 Risk of Bias Summary
Psychiatric Quarterly
Discussion
This systematic review provides a comprehensive overview of the use of Ketamine in MDD
with chronic and treatment-resistant course. Overall, twenty-six studies (77%) reported sig-
nificant improvement in depressive symptoms among patients receiving Ketamine compared
to the control group. Of the remaining eight studies, participants received ECT in five studies
in both the treatment and control group. Oral and intranasal Ketamine were effective in two
and three studies, respectively. It is noteworthy that one study comparing ECT to Ketamine
reported similar improvement in both groups [32]. However, Ketamine was not found to be
effective for the prevention of postpartum depression [18]. Ketamine was associated with
higher BDNF levels in patients with MDD compared to the control group and this higher
BDNF had a negative correlation with depression scores [31,43].
In the majority of studies (21/30), IV Ketamine was effective for treatment of unipolar
depression and ineffective in prevention of postpartum depression in one study [18]. The
existing literature suggests that Ketamine is superior to placebo and is equally effective as ECT
[32]. It resulted in significantly higher rates of remission with odd ratio (OR) of 3.86 and
Number Needed to Treat (NNT) of 6 after 24 h, 3 days and 7 days [44]. The clinical response
for Ketamine was reported at OR of 4.87 and NNT of 4[44]. The clinical applicability of
Ketamine is limited by shorter duration of action that can be challenging in patients with
chronic and treatment- resistant depression. The existing evidence suggests that the clinical
response is observable in 40 to 120 min in about 50% of patients. However, this response
lasted for two hours (51.1% Ketamine group VS 2% control group), one day (52.6% Ketamine
group VS 7% control group), three days (46.6% Ketamine group VS 7.1% control group), and
seven days (31% Ketamine group VS 7% control group) according to a pooled analysis of
seven RCTs [45]. The odds ratio for treatment response was clinically significant at day seven
with Ketamine in patients with unipolar depression [45]. The effects of Ketamine dissipated at
day 14, with only 10.9% responding to Ketamine compared to the control group [45].
Due to the chronic course of depression, the role of Ketamine was evaluated in the context
of multiple infusions of Ketamine. In this review, 13 studies administered multiple infusions of
IV Ketamine with improvement in six studies. Out of the remaining seven studies with no
significant benefits for Ketamine group, participants received ECT in addition to IV Ketamine
in five studies. The results posit discussion of two clinical questions: Ketamine as a potent
antidepressant option in combination with ECT and efficacy of multiple infusions of IV
Ketamine.
The lack of effectiveness of Ketamine in combination with ECT can be attributed to ECT
treatment ceiling and blunting of response [37]. A meta-analysis reported that Ketamine
augmentation of ECT resulted in a significantly greater reduction in depressive symptoms at
first treatment, but this efficacy did not last throughout the complete course of ECT treatments
[45]. This was in contrast to another meta-analysis reporting no additional benefit of Ketamine
when used along with ECT [46]. Considering small to moderate benefits with this combination
[47], Kellner and Iosifescu opined that this small -to-moderate effect size in patients with
chronic and TRD is worth investigating [48]. The existing scientific evidence lacks definitive,
adequately designed and well-powered studies in pursuit of this clinical question [48].
In this review article, Ghasemi and colleagues reported that multiple infusions of low dose
Ketamine (0.5 mg/kg, three times on three test days) were equally effective in improving
depression scores compared to ECT [32]. This effect was observable at 72 h and one week
after the last/third injection of Ketamine on BDI and HDRS scales. It is exciting to note the
Psychiatric Quarterly
Ketamine resulted in faster and more rapid improvement in earlier stages of treatment. These
results should be carefully considered in the context of smaller sample size and titration
method for ECT, possibly affecting the efficacy of ECT early in the course of treatment.
While it is a positive finding, it should be explored in studies of optimal sample size and robust
study design [32].
Ketamine was effective in five studies, when administered orally (two studies) and intra-
nasally (three studies). Oral Ketamine is suggested to exert significant antidepressant effect
with lesser risk of side effects but a slower pace of action than IV Ketamine. This conclusion
was drawn from a systematic review of two RCTs, one open-label trial, five retrospective chart
reviews, and five case reports.
Also, Esketamine, a nasal spray, was approved by the Food and Drug Administration
(FDA) for unipolar depression in March 2019 [49]. The recommended dose range for
Esketamine is 56 mg or 84 mg during induction phase (week 1 and 4) and maintenance phase
(week five to long-term) [50].
Ketamine antidepressant efficacy is determined by different variables such as route of
administration and dosage [51]. The most frequently prescribed dose of IV Ketamine is
0.5 mg/kg; however, some patients respond to dose range of 0.1 mg/kg to 0.75 mg/kg [51].
Higher doses are associated with greater risk of undesirable effects. The effectiveness of
Ketamine is also evident by oral, sublingual, transmucosal, intranasal, intravenous, intramus-
cular, and subcutaneous routes [51].
The most common side effects associated with ketamine are nausea, vomiting, dizziness,
diplopia, drowsiness, dysphoria, hallucinations, and confusion. Ketamine can cause delirium
in about 6% to 12% of patients [51]. Patients receiving Ketamine require close monitoring of
vital signs and cardiac status. It is also essential to monitor for neuropsychiatric functioning in
the clinic for return to baseline function [52].
This systematic review has several strengths. An electronic search of academic databases
combined with manual searching for references provides an exhaustive search for relevant
evidence. It provides an overview of RCTs of Ketamine in comparison to other treatment
options. However, this review also has several limitations. Due to heterogeneity, a meta-
analysis could not be performed. It is also important to consider the higher risk of bias in 10
studies while interpreting the results of these studies.
Conclusion
Ketamine is an effective treatment option for patients with MDD with considerable adverse
when administered via IV, IN and oral routes. The dose range for IV Ketamine ranges between
0.5 mg/kg to 1 mg/kg, 2884 mg for IN dose, and oral dose ranging from 50 mg daily to three
times a day. It is noteworthy that Ketamine was equally effective compared to ECT with a
faster response among patients who received Ketamine. Ketamine argumentation of ECT
needs to be explored further in well-designed studies of adequate sample size. The short-lived
antidepressant effect of Ketamine is a potential limitation, needing further studies administer-
ing multiple infusions.
Compliance with Ethical Standards
Disclosure of Potential Conflicts of Interest None to report.
Psychiatric Quarterly
Research Involving Human Participants and/or Animals Not applicable since it is a review article.
Informed Consent Not applicable since it is a review article.
References
1. Daly EJ, Singh JB, Fedgchin M, Cooper K, Lim P, Shelton RC, et al. Efficacy and safety of intranasal
esketamine adjunctive to oral antidepressant therapy in treatment-resistant depression: a randomized clinical
trial. JAMA Psychiat. 2018;75(2):13948. https://doi.org/10.1001/jamapsychiatry.2017.3739.
2. Zhong X, He H, Zhang C, Wang Z, Jiang M, Li Q, et al. Mood and neuropsychological effects of different
doses of ketamine in electroconvulsive therapy for treatment-resistant depression. J Affect Disord.
2016;201:12430. https://doi.org/10.1016/j.jad.2016.05.011.
3. Liu Q, He H, Yang J, Feng X, Zhao F, Lyu J. Changes in the global burden of depression from 1990 to
2017: Findings from the Global Burden of Disease study. J Psychiatr Res. 2019;(August):17. https://doi.
org/10.1016/j.jpsychires.2019.08.002
4. Faquih AE, Memon RI, Hafeez H, Zeshan M, Naveed S. A review of novel antidepressants: a guide for
clinicians. Cureus. 2019;11(3). https://doi.org/10.7759/cureus.4185
5. Greenberg PE, Fournier AA, Sisitsky T, Pike CT, Kessler RC. The economic burden of adults with major
depressive disorder in the United States (2005 and 2010). J Clin Psychiatry. 2015;76(2):15562. https://doi.
org/10.4088/JCP.14m09298.
6. Murrough JW, Soleimani L, Dewilde KE, et al. Ketamine for rapid reduction of suicidal ideation: a
randomized controlled trial. Psychol Med. 2015;45(16):3 57180. https://doi.org/10 .1017
/S0033291715001506.
7. Williams RD. Characteristics of completed sucides. Psychiatr Times. 2016.
8. Zeshan M, Waqas A, Naveed S, Ghulam H, Manocha P. Factors predicting length of stay in an adolescent
psychiatric unit, south Bronx, NY: a short report. J Can Acad Child Adolesc Psychiatry. 2018;27(2):1427.
9. Loo CK, Gálvez V, OKeefe E, et al. Placebo-controlled pilot trial testing dose titration and intravenous,
intramuscular and subcutaneous routes for ketamine in depression. Acta Psychiatr Scand. 2016;134(1):48
56. https://doi.org/10.1111/acps.12572.
10. Grunebaum, M. F., Galfalvy, H. C., Choo, T. H., Keilp, J. G., Moitra, V. K., Parris, M. S., ... & Oquendo
MA. Ketamine for rapid reduction of suicidal thoughts in major depression: a midazoam-controlled
randomized clinical trial. Am J Psychiatry 2018;175(5):327335. doi:https://doi.org/10.1038/nm.2451.A.
11. Abdallah CG, Fasula M, Kelmendi B, Sanacora G, Ostroff R. Rapid antidepressant effect of ketamine in the
electroconvulsive therapy setting. J ECT. 2012;28(3):15761. https://doi.org/10.1097/YCT.0b013e31824
f8296.
12. Murrough JW, Iosifescu DV, Chang L, al Jurdi RK, Green C, Perez A, et al. P.2.F.028 antidepressant
efficacy of ketamine in treatment-resistant major depression: a two-site, randomised controlled trial. Eur
Neuropsychopharmacol. 2013;23(10):S4112. https://doi.org/10.1016/s0924-977x(13)70651-5.
13. Sos P, Klirova M, Novak T, Kohutova B, Horacek J, Palenice T. Relationship of ketamines antidepressant
and psychotomimetic effects in unipolar depression. Act Nerv Super Rediviva. 2013;55(12):5763.
14. Anderson IM, Blamire A, Branton T, Brigadoi S, Clark R, Downey D, et al. Randomised controlled trial of
ketamine augmentation of electroconvulsive therapy to improve neuropsychological and clinical outcomes
in depression (ketamine-ECT study). Effic Mech Eval. 2017;4(2):1112. https://doi.org/10.3310/eme04020.
15. Carspecken CW, Borisovskaya A, Lan ST, Heller K, Buchholz J, Ruskin D, et al. Ketamine anesthesia does
not improve depression scores in electroconvulsive therapy: a randomized clinical trial. J Neurosurg
Anesthesiol. 2018;30(4):30513. https://doi.org/10.1097/ANA.0000000000000511.
16. Järventausta K, Chrapek W, Kampman O, Tuohimaa K, Björkqvist M, Häkkinen H, et al. Effects of S-
ketamine as an anesthetic adjuvant to propofol on treatment response to electroconvulsive therapy in
treatment-r esistant depres sion: a randomized pilot s tudy. J ECT. 2013;29 (3):15 861.
https://doi.
org/10.1097/YCT.0b013e318283b7e9.
17. Singh JB, Fedgchin M, Daly E, Xi L, Melman C, de Bruecker G, et al. Intravenous Esketamine in adult
treatment-resistant depression: a double-blind, double-randomization, placebo-controlled study. Biol
Psychiatry. 2016;80(6):42431. https://doi.org/10.1016/j.biopsych.2015.10.018.
18. Xu Y, Li Y, Huang X, Chen D, She B, Ma D. Single bolus low-dose of ketamine does not prevent
postpartum depression: a randomized, double-blind, placebo-controlled, prospective clinical trial. Arch
Gynecol Obstet. 2017;295(5):116774. https://doi.org/10.1007/s00404-017-4334-8.
19. Burger J, Capobianco M, Lovern R, Boche B, Ross E, Darracq MA, et al. A double-blinded, randomized,
placebo-controlled sub-dissociative dose ketamine pilot study in the treatment of acute depression and
Psychiatric Quarterly
Suicidality in a military emergency department setting. Mil Med. 2016;181(10):11959. https://doi.
org/10.7205/milmed-d-15-00431.
20. Gamble JJ, Bi H, Bowen R, Weisgerber G, Sanjanwala R, Prasad R, et al. Ketamine-based anesthesia
improves electroconvulsive therapy outcomes: a randomized-controlled studyLanesthésie à base de
kétamine améliore les résultats de lélectro-convulsivothérapie : une étude randomisée contrôlée. Can J
Anesth Can danesthésie. 2018;65(6):63646. https://doi.org/10.1007/s12630-018-1088-0.
21. Hu YD, Xiang YT, Fang JX, Zu S, Sha S, Shi H, et al. Single i.v. ketamine augmentation of newly initiated
escitalopram for major depression: results from a randomized, placebo-controlled 4-week study. Psychol
Med. 2016;46(3):62335. https://doi.org/10.1017/S0033291715002159.
22. Kheirkhah F, Tayyebi G, Rabiee SM, Moghadamnia AA, Bijani A. Efficacy of different doses of ketamine
as a bolus in major depressive disorder. Casp J Intern Med. 2018;9(3):2207. https://doi.org/10.22088
/cjim.9.3.220.
23. Zarate CA, Singh JB, Carlson PJ, Brutsche NE, Ameli R, Luckenbaugh DA, et al. A randomized trial of an
N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63(8):
85664. https://doi.org/10.1001/archpsyc.63.8.856.
24. Lai R, Katalinic N, Glue P, Somogyi AA, Mitchell PB, Leyden J, et al. Pilot dose-response trial of I.V.
ketamine in treatment-resistant depression. World J Biol Psychiatry. 2014;15(7):57984. https://doi.
org/10.3109/15622975.2014.922697.
25. Salehi B, Mohammadbeigi A, Kamali AR, Taheri-Nejad MR, Moshiri I. Impact comparison of ketamine
and sodium thiopental on anesthesia during electroconvulsive therapy in major depression patients with
drug-resistant; a double-blind randomized clinical trial. Ann Card Anaesth. 2015;18(4):48690. https://doi.
org/10.4103/0971-9784.166444.
26. Xu R, Zhan Y, Chen S. Effect of intraoperative single administration of sub-anesthesia ketamine on breast
cancer patients with depression. Biomed Res. 2017;2017(Special Issue
HealthScienceandBioConvergenceTechnologyEdition-II):S552-S556.
27. Jiang M, Wang MH, Wang X. Bin, et al. effect of intraoperative application of ketamine on postoperative
depressed mood in patients undergoing elective orthopedic surgery. J Anesth. 2016;30(2):2327. https://doi.
org/10.1007/s00540-015-2096-7.
28. Kudoh A, Takahira Y, Katagai H, Takazawa T. Small-dose ketamine improves the postoperative state of
depressed patients. Anesth Analg. 2002;95(1):1148. https://doi.org/10.1097/00000539-200207000-00020.
29. Fan W, Yang HK, Sun Y, et al. Ketamine rapidly relieves acute suicidal ideation in cancer patients: A
randomized controlled clinical trial. Oncotarget. 2017;8(2):235660. https://doi.org/10.18632
/oncotarget.13743.
30. Fava M, Freeman MP, Flynn M, Judge H, Hoeppner BB, Cusin C, et al. Double-blind, placebo-controlled,
dose-ranging trial of intravenous ketamine as adjunctive therapy in treatment-resistant depression (TRD).
Mol Psychiatry. 2018;25:1592603. https://doi.org/10.1038/s41380-018-0256-5.
31. Su TP, Chen MH, Li CT, Lin WC, Hong CJ, Gueorguieva R, et al. Dose-related effects of adjunctive
ketamine in taiwanese pati ents with treatme nt-res istant dep ression. Neur opsychop harmacol ogy.
2017;42(13):248292. https://doi.org/10.1038/npp.2017.94.
32. Ghasemi M, Kazemi MH, Yoosefi A, Ghasemi A, Paragomi P, Amini H, et al. Rapid antidepressant effects
of repeated doses of ketamine compared with electroconvulsive therapy in hospitalized patients with major
depressive disorder. Psychiatry Res. 2014;215(2):35561. https://doi.org/10.1016/j.psychres.2013.12.008.
33. Alizadeh NS, Maroufi A, Nasseri K, et al. Antidepressant effect of combined ketamine and electroconvul-
sive therapy on patients with major depressive disorder: A randomized trial. Iran J Psychiatry Behav Sci.
2015;9(3). https://doi.org/10.17795/ijpbs-1578
34. Chen Q, Min S, Hao X, Peng L, Meng H, Luo Q, et al. Effect of low dose of ketamine on learning memory
function in patients undergoing electroconvulsive therapy-a randomized, double-blind, controlled clinical
study. J ECT. 2017;33(2):8995. https://doi.org/10.1097/YCT.0000000000000365.
35. Fernie G, Currie J, Perrin JS, Stewart CA, Anderson V, Bennett DM, et al. Ketamine as the anaesthetic for
electroconvulsive therapy: the KANECT randomised controlled trial. Br J Psychiatry. 2017;210(6):4228.
https://doi.org/10.1192/bjp.bp.116.189134.
36. Wang X, Chen Y, Zhou X, Liu F, Zhang T, Zhang C. Effects of propofol and ketamine as combined
anesthesia for electroconvulsive therapy in patients with depressive disorder. J ECT. 2012;28(2):12832.
https://doi.org/10.1097/YCT.0b013e31824d1d02.
37. Yoosefi A, Sepehri AS, Kargar M, Akhondzadeh S, Sadeghi M, Rafei A, et al. Comparing effects of
ketamine and thiopental administration during electroconvulsive therapy in patients with major depressive
disorder: a randomized, double-blind study. J ECT. 2014;30(1):1521. https://doi.org/10.1097/YCT.0b013
e3182a4b4c6.
Psychiatric Quarterly
38. Lenze EJ, Farber NB, Kharasch E, Schweiger J, Yingling M, Olney J, et al. Ninety-six hour ketamine
infusion with co-administered clonidine for treatment-resistant depression: a pilot randomised controlled
trial. World J Biol Psychiatry. 2016;17(3):2308. https://doi.org/10.3109/15622975.2016.1142607.
39. Lapidus KAB, Levitch CF, Perez AM, et al. NIH Public Access. 2015;76(12):9706. https://doi.
org/10.1016/j.biopsych.2014.03.026.A.
40. Canuso CM, Singh JB, Fedgchin M, Alphs L, Lane R, Lim P, et al. Efficacy and safety of intranasal
esketamine for the rapid reduction of symptoms of depression and suicidality in patients at imminent risk for
suicide: results of a double-blind, randomized, placebo-controlled study. Am J Psychiatry. 2018;175(7):
62030. https://doi.org/10.1176/appi.ajp.2018.17060720.
41. Arabzadeh S, Hakkikazazi E, Shahmansouri N, Tafakhori A, Ghajar A, Jafarinia M, et al. Does oral
administration of ketamine accelerate response to treatment in major depressive disorder? Results of a
double-blind controlled trial. J Affect Disord. 2018;235(February):23641. https://doi.org/10.1016/j.
jad.2018.02.056.
42. Jafarinia M, Afarideh M, Tafakhori A, Arbabi M, Ghajar A, Noorbala AA, et al. Efficacy and safety of oral
ketamine versus diclofenac to alleviate mild to moderate depression in chronic pain patients: a double-blind,
randomized, controlled trial. J Affect Disord. 2016;204:18. https://doi.org/10.1016/j.jad.2016.05.076.
43. Haile CN, Murrough JW, Iosifescu DV, Chang LC, al Jurdi RK, Foulkes A, et al. Plasma brain derived
neurotrophic factor (BDNF) and response to ketamine in treatment-resistant depression. Int J
Neuropsychopharmacol. 2014;17(2):3316. https://doi.org/10.1017/S1461145713001119.
44. McGirr A, Berlim MT, Bond DJ, Fleck MP, Yatham LN, Lam RW. A systematic review and meta-analysis
of randomized, double-blind, placebo-controlled trials of ketamine in the rapid treatment of major depres-
sive episodes. Psychol Med. 2015;45(4):693704. https://doi.org/10.1017/S0033291714001603.
45. Newport DJ, Carpenter LL, McDonald WM, Potash JB, Tohen M, Nemeroff CB. Ketamine and other
NMDA antagonists: early clinical trials and possible mechanisms in depression. Am J Psychiatry.
2015;172(10):95066. https://doi.org/10.1176/appi.ajp.2015.15040465.
46. McGirr A, Berlim MT, Bond DJ, Neufeld NH, Chan PY, Yatham LN, et al. A systematic review and meta-
analysis of randomized controlled trials of adjunctive ketamine in electroconvulsive therapy: efficacy and
tolerability. J Psychiatr Res. 2015;62:2330. https://doi.org/10.1016/j.jpsychires.2015.01.003.
47. Anderson IM, Blamire A, Branton T, Clark R, Downey D, Dunn G, et al. Ketamine augmentation of
electroconvulsive therapy to improve neuropsychological and clinical outcomes in depression (ketamine-
ECT): a multicentre, double-blind, randomised, parallel-group, superiority trial. Lancet Psychiatry.
2017;4(5):36577. https://doi.org/10.1016/S2215-0366(17)30077-9.
48. Kellner CH, Iosifescu DV. Ketamine and ECT: better alone than together? Lancet Psychiatry. 2017;4(5):
3489. https://doi.org/10.1016/S2215-0366(17)30099-8.
49. FDA. FDA approves new nasal spray medication for treatment-resistant depression. FDA News Release.
2019.
50. Companies JP. Spravato: Package Insert. 2020.
51. Andrade C. Andrade, C. (2017). Ketamine for depression, 4: in what dose, at what rate, by what route, for
how long, and at what frequency?. The Journal of clinical psychiatry, 78(7), e852-e857. 2017;81(April).
52. Rosenbaum, S.B., Gupta, V,. & Palacios JL. Ketamine. Statpearls Publishing; 2018.
PublishersNote Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional affiliations.
Psychiatric Quarterly
Raheel Imtiaz Memon, M.D., is a second-year psychiatry resident at Henry Ford Allegiance Health, Jackson,
Michigan. He did his medical school from Liaquat University of Medical & Health Sciences, Jamshoro, Pakistan.
He is interested in pursuing a career in child and adolescent psychiatry with a particular focus on
neurodevelopmental disorders, specifically autism spectrum disorder, and the impact of substances and trauma
during childhood. He will be applying for child and adolescent psychiatry fellowship after completion of general
psychiatry residency and planning to stay in academia later on.
Sadiq Naveed, M.D., is an assistant professor in the Department of Psychiatry and Behavioral Sciences at the
University of Kansas Medical Center. He is boardcertified in child and adolescent psychiatry, as well as adult
psychiatry. He also completed Master of Public Health degree from Benedictine University in Illinois. He earned
his medical degree from Nishtar Medical College in Multan, Pakistan, and received his training in adult
psychiatry at Griffin Memorial Hospital in Norman, Oklahoma. He also completed his training in child and
adolescent psychiatry at the University of Kansas Medical Center. Dr. Sadiq Naveed is Honorary Research
Fellow at Human Development Research Foundation of Pakistan. He is also completing his postgraduate course
in University of Massachusetts, Boston.
Amber Ehsan Faquih, M.D., graduated from Dow University of Health Sciences, Karachi, Pakistan. Presently,
she is working as research assistant and working on different projects. She is aspiring residency applicant and
keenly interested in pursuing her career in psychiatry.
Ania Fida graduated from King Edward Medical University, started Psychiatry Residency in Pakistan then
moved to the US. Since then, she has engaged in clinical rotation and also have worked as a research assistant.
She will be starting her Psychiatry Residency Training at Medical College of Wisconsin Central Wausau in July
2020.
Noureen Abbas, MD, graduated from Fatima Memorial Hospital College of Medicine & Dentistry. She is
currently working as a research assistant. She will be starting a residency physician at Cahaba- University of
Alabama at Birmingham in July 2020.
Amna Mohyud Din Chaudhary, M.D., is a graduate of Nishtar Medical College and Hospital, Multan,
Pakistan. She is primarily interested in psychiatry and a general psychiatry residency aspirant. Her interests
include mood disorder, and schizophrenia.
Zheala Qayyum, MD. MMSc is the Assistant Clinical Professor of Psychiatry at Yale School of Medicine. She
is board certified in general psychiatry, child and adolescent psychiatry and consultation liaison psychiatry. She
also completed her Master of Medical Science in Medical Education from Harvard Medical School. She is
currently the Program Director for the Child and Adolescent psychiatry fellowship at Boston Childrens Hospital
and faculty at Harvard Medical School. Her interests included early onset psychosis, supporting mental health of
LGBTQ youth, psychoncology and palliative care in children and adolescents. Additionally, her research
interests lie in the supervision of trainees in the event of patient death by suicide.
Psychiatric Quarterly
Affiliations
Raheel Imtiaz Memon
1
& Sadiq Naveed
2
& Amber Ehsan Faquih
3
& Ania Fida
4
& Noureen
Abbas
5
& Amna Mohyud Din Chaudhary
6
& Zheala Qayyum
7
Raheel Imtiaz Memon
RMEMON2@HFHS.ORG
Amber Ehsan Faquih
amber_jawed17@live.com
Ania Fida
an.fida@hotmail.com
Noureen Abbas
noureenabbasmd@gmail.com
Amna Mohyud Din Chaudhary
dramna2014@gmail.com
Zheala Qayyum
zheala.qayyum@childrens.harvard.edu
1
Resident Physician, Henry Ford Allegiance Health, Jackson, MI, USA
2
University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, USA
3
Dow University of Health Sciences, Karachi, Pakistan
4
King Edward Medical University, Lahore, Pakistan
5
FMH College of Medicine & Dentistry, Lahore, Pakistan
6
Nishtar Medical University, Multan, Pakistan
7
Boston Childrens Hospital, Boston, MA, USA
Psychiatric Quarterly