You are on page 1of 18

Postgraduate Medicine

ISSN: (Print) (Online) Journal homepage: www.tandfonline.com/journals/ipgm20

Considerations when selecting an antidepressant:


a narrative review for primary care providers
treating adults with depression

C. Brendan Montano, W. Clay Jackson, Denise Vanacore & Richard Weisler

To cite this article: C. Brendan Montano, W. Clay Jackson, Denise Vanacore & Richard Weisler
(2023) Considerations when selecting an antidepressant: a narrative review for primary
care providers treating adults with depression, Postgraduate Medicine, 135:5, 449-465, DOI:
10.1080/00325481.2023.2189868

To link to this article: https://doi.org/10.1080/00325481.2023.2189868

© 2023 The Author(s). Published by Informa


UK Limited, trading as Taylor & Francis
Group.

Published online: 05 Apr 2023.

Submit your article to this journal

Article views: 7100

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ipgm20
POSTGRADUATE MEDICINE
2023, VOL. 135, NO. 5, 449–465
https://doi.org/10.1080/00325481.2023.2189868

REVIEW

Considerations when selecting an antidepressant: a narrative review for primary


care providers treating adults with depression
C. Brendan Montanoa, W. Clay Jacksonb, Denise Vanacorec and Richard Weislerd,e
a
Montano Wellness LLC, CT Clinical Research, University of Connecticut Medical School, Farmington, CT, USA; bWest Cancer Center, Department of
Family Medicine and Department of Psychiatry, University of Tennessee College of Medicine, Memphis, TN, USA; cSchool of Nursing, Eastern
University, St. Davids, PA, USA; dP.A. & Associates; Department of Psychiatry, Duke University, Durham, NC, USA; eDepartment of Psychiatry,
University of North Carolina at Chapel Hill, Chapel Hill, NC, USA

ABSTRACT ARTICLE HISTORY


Major depressive disorder (MDD) is a debilitating mental disorder that can be treated with Received 16 December 2022
a number of different antidepressant therapies, each with its own unique prescribing considera­ Accepted 8 March 2023
tions. Complicating the selection of an appropriate antidepressant for adults with MDD is the
KEYWORDS
heterogeneity of clinical profiles and depression subtypes. Additionally, patient comorbidities,
MDD; primary care;
preferences, and likelihood of adhering to treatment must all be considered when selecting an treatment selection;
appropriate therapy. With the majority of prescriptions being written by primary care practi­ antidepressant; partial
tioners, it is appropriate to review the unique characteristics of all available antidepressants, response; comorbidities;
including safety considerations. Prior to initiating antidepressant treatment and when patients tolerability; adherence;
do not respond adequately to initial therapy and/or exhibit any hypomanic or manic symptoms, guidelines; measurement-
bipolar disorder must be ruled out, and evaluation for psychiatric comorbidities must be con­ guided care
sidered as well. Patients with an inadequate response may then require a treatment switch to
another drug with a different mechanism of action, combination, or augmentation strategy. In this
narrative review, we propose that careful selection of the most appropriate antidepressant for
adult patients with MDD based on their clinical profile and comorbidities is vital for initial
treatment selection.
Strategies must be considered for addressing partial and inadequate responses as well to help
patients achieve full remission and sustained functional recovery. This review also highlights data for
MDD clinical outcomes for which gaps in the literature have been identified, including the effects of
antidepressants on functional outcomes, sleep disturbances, emotional and cognitive blunting, anxiety,
and residual symptoms of depression.

PLAIN LANGUAGE SUMMARY


Major depressive disorder (MDD) is a leading cause of disability worldwide and can affect each patient
differently. Antidepressants play a critical role in treatment; however, with multiple antidepressant options
available, it is important that providers select the best fit for each patient. Rather than use a “one size fits all”
approach, it is important to consider each patient’s symptoms, medical and psychiatric comorbidities, as
well as their treatment preferences. A clear summary of each antidepressant’s distinctive characteristics is
essential for providers to select antidepressants to best match each patient’s needs.
This narrative review aims to discuss the latest information on available antidepressants, including
their risks and benefits and how they impact symptoms of MDD such as sleep disturbances, anxiety,
emotional blunting, and changes in cognition, as well as different treatment goals, such as the ability to
function in everyday life. This information can guide clinical practice recommendations and further
enable shared decision-making between the provider and patient, incorporating individual treatment
needs and preferences.
In addition, many patients do not reach their treatment goals with the first antidepressant or may
continue to have symptoms of depression after treatment. This review discusses strategies to increase
the likelihood of symptom improvement and creates awareness of patient-specific considerations.
Overall, careful, personalized selection of antidepressant treatment is critical for finding the right
balance of maximized antidepressant effect with minimized side effects, leading to the best possibility
for patients to tolerate the medication and ultimately helping patients reach their treatment goals.

CONTACT C. Brendan Montano docmontano@aol.com Montano Wellness LLC, 160 West St., Ste. 1A, Cromwell, CT 06416, USA
© 2023 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (http://creativecommons.org/licenses/by-nc-nd/4.0/),
which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited, and is not altered, transformed, or built upon in any way.
The terms on which this article has been published allow the posting of the Accepted Manuscript in a repository by the author(s) or with their consent.
450 C. B. MONTANO ET AL.

1. Introduction especially for moderate to severe MDD [8]. This multidisciplin­


ary approach may include pharmacotherapy, psychotherapy,
Depression, or major depressive disorder (MDD), is recognized as
a combination of medications and psychotherapy, or other
an important mental health concern in the United States (US)
somatic therapies such as electroconvulsive therapy, transcra­
and global populations. The devastating impact depression has
nial magnetic stimulation or light therapy, and alternative
on individuals and society is well documented [1,2]. The 2020
therapies [3,5,8]. Pharmacotherapy for depression has been
National Survey on Drug Use and Health reported that an esti­
available for more than 50 years, since the first antidepressants
mated 14.8 million adults in the US had a major depressive
targeting multiple monoaminergic pathways became available
episode with severe impairment in the past year. Globally, the
in the 1950s, and remains the mainstay of treatment [9].
12-month prevalence of MDD is 6%, and the risk of MDD over
Nevertheless, a healthy diet and exercise are also important
a lifetime is approximately 11.1%–14.6% [2,3]. With the emer­
factors in the global treatment of MDD [10,11]. An increase in
gence of the SARS-CoV-2 virus and the COVID-19 pandemic in
our understanding of the neurobiology of depression has
2020, the COVID-19 Mental Disorders Collaborators estimate that
provided a more complex picture beyond monoaminergic
throughout 2020, the pandemic led to a clinically significant
transmission, with MDD now thought to involve a complex
27.6% increase in cases of depression. The exacerbation of
interplay of various genes, neural networks, structural neuro­
MDD as a global burden highlights an urgent need to strengthen
biological changes, and neurotransmitters including serotonin,
mental healthcare systems, which includes prioritizing promo­
norepinephrine, dopamine, glutamate, GABA-ergic systems,
tion of well-being and development of depression mitigation
and histamine at certain brain regions [12–15]. This complexity
strategies [4]. Optimizing the management of depression in
is further reflected by the different subtypes of depression
primary care settings represents a critical strategy for addressing
that can occur, such as anxious depression. Furthermore, psy­
the burden of depression, as approximately 60% of mental
chiatric and medical comorbidities including sleep disorders,
healthcare is provided in primary care settings and almost 80%
anxiety disorders, attention deficit/hyperactivity disorder
of antidepressant prescriptions are written by providers who are
(ADHD), obesity and eating disorders, as well as pain and
not mental health care specialists [5]. This narrative literature
cardiovascular disease are now regarded as important risk
review aims to differentiate available antidepressants by present­
factors for the development of depression that can also influ­
ing a comprehensive examination of their risks and benefits in
ence treatment [3,8,13,16]. As our understanding of the neu­
order to support personalized treatment selection and help
robiology of depression has advanced, so too has the number
patients with MDD reach their treatment goals.
of treatments available, with more than 35 US Food and Drug
Administration (FDA)-approved antidepressant medications
[3,17]. This understanding provides both an opportunity and
2. Treatment options for adults with MDD
a challenge – an opportunity to tailor treatment to the
Depression is highly heterogeneous in its clinical presentation patient’s specific clinical and pharmacogenomic profile,
and is associated with a constellation of symptoms across comorbidities, and preferences, and a challenge, especially in
a wide range of different emotional, physical, functional, and primary care, because clinicians who do not specialize in
cognitive domains depending on each individual patient pre­ psychiatry have limited exposure to the nuances of the differ­
sentation [5–7]. For this reason, a multidisciplinary approach ent compounds and their risk-benefit profiles [12,18].
to the management of adult patients with MDD is increasingly The abundance of pharmacotherapeutic options for MDD
considered to provide them with the best chance of achieving has coincided with a consistent increase in the number of
remission, often defined as the absence of clinical levels of patients being prescribed antidepressants (Figure 1) [19]. As
depressive symptoms, and sustained functional recovery, many as 1 in 10 people in the US are currently prescribed an
POSTGRADUATE MEDICINE 451

Figure 1. Growth in number of patients prescribed antidepressants for MDD: 1996–2015 [19].
Reproduced from Luo Y, Kataoka Y, Ostinelli EG, Cipriani A and Furukawa TA. National prescription patterns of antidepressants in the treatment of adults with major depression in the US
between 1996 and 2015: a population representative survey based analysis. Front Psychiatry. 2020;11:35. doi: 10.3389/fpsyt.2020.00035; under the terms of the Creative Commons
Attribution License (CCBY). https://creativecommons.org/licenses/by/4.0/. The SE of number of adults with MDD is shown by error bars.aPatients with multiple antidepressants: referring to
patients who were prescribed >1 antidepressant during that year, i.e. both patients with combination therapy and patients who changed previous monotherapy into a new drug in
that year. MDD: major depressive disorder.

antidepressant, reflecting possible overtreatment of patients algorithms to follow for treatment initiation or switch. In this
with subsyndromal MDD or adjustment disorders; however, article, we review some of the latest data for antidepressants
only half of patients diagnosed with MDD receive an adequate to treat adults with MDD and revisit the latest clinical practice
prescription, suggesting that overprescribing and inadequate recommendations, highlighting the importance of tailoring
prescribing must be addressed [12]. It is possible that the array treatment to the clinical profile and comorbidities of the
of antidepressants is not well differentiated and understood patient. Additionally, when emotional blunting is secondary
by some primary care providers, who frequently contend with to antidepressant selection, guidelines for diagnosis and stra­
time constraints and large caseloads, not to mention the dis­ tegies for treatment modification must be developed [21,22].
ruption to healthcare delivery in primary care settings caused We propose that careful selection of the most appropriate
by the COVID-19 pandemic. As a result, physicians in primary antidepressant for adult patients with MDD is the best way
care may default to 1 or 2 different antidepressants for all to counter both inadequate prescribing of antidepressants
adult patients, thus applying a “one size fits all” approach to and address partial and inadequate responses to antidepres­
a disease with a highly individualized clinical profile. This is sant therapy. This article also highlights data for MDD clinical
consistent with the observation that inadequate responses to outcomes for which gaps in the literature have been identified
initial antidepressants are common, and clear guidance is that may influence response to treatment, including the
lacking for primary care providers concerning how partial effects of antidepressants on functional outcomes, sleep dis­
responses may be addressed [10]. Just as with initial antide­ turbances, anxiety, emotional and cognitive blunting, and any
pressant selection, the choice of medication in cases of partial residual symptoms of depression.
response should be guided by the patient’s clinical profile or
depression subtype and comorbid conditions. Additionally,
4. Overview of antidepressant therapies by class
some antidepressants are associated with emotional and cog­
nitive blunting, which may add to the burden of illness for Acute-phase pharmacotherapies for MDD include selective sero­
patients so affected [20]. tonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reup­
take inhibitors (SNRIs), tricyclic antidepressants (TCAs),
monoamine oxidase inhibitors (MAOIs), N-methyl-D-aspartate
3. Addressing literature gaps
(NMDA) receptor antagonists, atypicals, and serotonin modulators
There are few review articles aimed at educating a primary that have more complex mechanisms of action and cannot be
care audience about the factors to consider when choosing an easily categorized (Table 1) [8,12,13,23–26]. The efficacy of these
antidepressant, as well as about general principle-guided treatments compared with placebo has been established in
452 C. B. MONTANO ET AL.

Table 1. Antidepressant profiles by class.


Mechanism of antidepressant Symptoms/Clinical
Class effect Examples profilea AEs Additional considerations
SSRIs ● Targeted and selective ● Fluoxetine Some are also ● Headaches, GI distress, ● Avoid many of the
serotonin reuptake ● Paroxetine specifically approved insomnia, fatigue, anxiety, anticholinergic and
inhibition [32] ● Fluvoxamine to treat [32]: sexual dysfunction, weight cardiac AEs of the TCAs
● Panic disorder
● Sertraline gain, falls/fractures, [32]
● Social anxiety
● Citalopram coagulopathy [32,34–36] ● Do not require dietary
● Escitalopram [32] disorder ● Prolongation of QT interval and drug-related restric­
● OCD
with citalopram and escita­ tions [32]
● PTSD
lopram [32,35] ● Often prescribed first-line
● Bulimia nervosa
because of a more tar­
● Premenstrual dys­
geted MOA and safer AE
phoric disorder profile. Subtle AE differ­
ences must be weighed
by the prescriber
● May not be suited for
patients with sleep dis­
turbances [8,12,32]
● Risk of GI bleeding may
be doubled with conco­
mitant NSAID use [32,36]
● May be associated with
increased risk of falls and
fractures with long-term
use (may be less suited
to elderly patients)
[8,34,107,108]
● Associated with emo­
tional blunting [20,22]
● Associated with disconti­
nuation syndrome [12]
SNRIs ● Dual inhibition of serotonin ● Venlafaxine ● Venlafaxine may be ● Nausea, insomnia, dry ● Side effects are similar
and norepinephrine ● Desvenlafaxine suitable for mouth, headache, to SSRIs, but may reflect
reuptake ● Duloxetine comorbid anxiety increased blood pressure, noradrenergic activity
● May have an effect on ● Levomilnacipran ● Duloxetine may be sexual dysfunction, weight [8]
other neurotransmitters [37] suited to treat gain [12,33,37] ● May not be suited for
[12,37] depression with pain patients with sleep dis­
[33,37] turbances [33]
Serotonin ● Multimodal SSRI and ● Vilazodone ● Nausea, diarrhea, ● Better side-effect profile
modulator a partial 5-HT1A receptor headache, insomnia [48,53] than most
agonist [52,53] antidepressants with
● Partial agonist activity at 5- lower risk of sexual
HT1A receptors [48,109] dysfunction or weight
gain [12,52,53]
● Multimodal; thought to be ● Vortioxetine ● Older patients (aged ● Nausea, diarrhea, dizziness ● Better side-effect profile
mediated through antago­ 64–88 years) [52,55] than most
nist effect on the 5-HT3, 5- ● TESD antidepressants with
HT7, and 5-HT1D receptors; ● Deficits in cognitive lower risk of sexual
processing speed
partial agonist activity on dysfunction or weight
● Comorbid anxiety
the 5-HT1B receptor; ago­ gain [55]
nist effect on the 5-HT1A [106] ● Excellent for avoiding
receptor; and inhibition of emotional blunting and
the 5-HT transporter [51] cognitive blunting in
● Also enhances the extra­ most patients. Enhances
cellular concentration of speed of cognitive pro­
dopamine, histamine, nore­ cessing [54,55]
pinephrine, and acetylcho­
line [109]

(Continued )
POSTGRADUATE MEDICINE 453

Table 1. (Continued).
Mechanism of antidepressant Symptoms/Clinical
Class effect Examples profilea AEs Additional considerations
Atypical ● Norepinephrine-dopamine ● Bupropion ● Fatigue ● Headache, agitation, ● Increased seizure risk in
reuptake inhibitor [8,12] ● Anxious distress [8] insomnia, loss of appetite, patients with epilepsy
● No direct effects on sero­ weight loss, sweating, ● No sexual dysfunction or
tonin, histamine, acetyl­ hypertension [8] weight gain
choline, or adrenaline ● Contraindicated in eating
receptors [50] disorder due to weight
loss
● May also help patients
quit smoking [8]
● Commonly used for aug­
mentation [50]
● Targets both melatonin and ● Agomelatineb ● Comorbid insomnia ● Toxic hepatitis (rare, ● The first melatonergic
serotonin receptors [48] sporadic) [48] antidepressant designed
[43,110] to restore perturbed
biological rhythms
[47,110]
● Requires monitoring of
liver function [48]
● Possibly lower risk of
sexual side effects [110]
● Inhibits norepinephrine ● Mirtazapine ● Comorbid insomnia, ● Sedation, increased ● Reduced sexual
alpha-2 autoreceptors, GAD, OCD, PTSD, appetite, weight gain [8] dysfunction compared
allowing more norepi­ social anxiety ● Potential increase in cho­ with SSRIs/SNRIs [42]
nephrine to be released disorder, headaches/ lesterol [8,43] ● Some risk of reduced
from presynaptic nerve migraine [42] WBC count [42]
terminals [12] ● May have fast onset of
● Potent agonist of histami­ action compared with
nergic H1 receptors [43] SSRIs [41]
● Also blocks serotonergic 5-
HT2A/2C and 5-HT3 receptors
[42,43]
● Serotonin antagonist and ● Trazodone ● Comorbid insomnia ● Sedation, nausea, priapism ● Reduced risk of sexual
reuptake inhibitor; com­ ● Nefazodone or GAD [44,45] (rare), orthostatic dysfunction, but may
bined effect on serotonin hypotension [8] cause priapism [44]
reuptake pumps and 5- ● Often used to induce
HT2A/2C receptors via both sleep as a positive effect;
receptor agonist and slow-release formulation
antagonist activities [44] may be less sedating
[8,46]
● Concerns with hepato­
toxicity and liver function
monitoring limit the use
of nefazodone [12,44]
MAOIs ● Inhibit the activity of the ● Isocarboxazid ● MDD with atypical ● Weight gain, fatigue, ● Generally, not
enzyme monoamine ● Phenelzine features, or patients orthostatic hypotension, prescribed first-line due
oxidase, preventing the ● Tranylcypromine with poor response sexual side effects [8] to food and drug-drug
breakdown of monoamine ● Selegiline to other interactions that may
neurotransmitters [8,27,28] ● Moclobemide antidepressants cause serotonin
[8,12,28] [8,28,29] syndrome and
hypertensive crises
[8,12,28,29]
● Selegiline and moclobe­
mide may be considered
safer and more suitable
for use earlier in treat­
ment
TCAs ● Elevate extracellular ● Imipramine ● Amitriptyline for ● Weight gain, sedation, dry ● Generally, not
serotonin and ● Amitriptyline pain, melancholia mouth, nausea, blurred prescribed first-line due
norepinephrine levels via ● Doxepin (psychomotor vision, constipation, to increased
uptake inhibition [12] ● Desipramine slowing, diurnal tachycardia, orthostatic anticholinergic and
● Antagonize muscarinic ● Nortriptyline mood variation) hypotension [8,19] cardiotoxic side effects,
acetylcholine receptors ● Amoxapine ● Comorbid insomnia risk of falls [8]
● Clomipramine ● Pain
● Maprotiline ● Social anxiety
● Trimipramine disorder
● Protriptyline ● Severely depressed
[3,8,12] with melancholic
features [8,19]

(Continued )
454 C. B. MONTANO ET AL.

Table 1. (Continued).
Mechanism of antidepressant Symptoms/Clinical
Class effect Examples profilea AEs Additional considerations
NMDA receptor ● Ketamine is ● Ketamine ● Ketamine and ● Ketamine and esketamine: ●Ketamine and esketamine:
Have rapid-onset efficacy
antagonists a noncompetitive NMDA ● Esketamine esketamine: TRD Treatment-emergent
in TRD [23]
and glutamate receptor ● Dextromethorphan/ with ≥2 failed adverse events generally
antagonist, predicted to bupropion antidepressants include, but are not limited ● Intranasal esketamine
mediate its effect by treatment, or for to, psychiatric and intravenous keta­
a secondary increase in MDD patients with (psychotomimetic effects mine are effective routes
structural synaptic acute suicidality and dissociation), of administration [23]
connectivity [111] [23,57] neurologic/cognitive ● To be administered only
● Esketamine, like ketamine, (dizziness, drowsiness and in settings with multidis­
is postulated to inhibit light-headedness), ciplinary trained person­
NMDA receptor on gamma- hemodynamic, nel [23]
aminobutyric acid inter­ genitourinary, and abuse ● Esketamine administra­
neurons to restore synaptic liability [23] tion may require REMS in
function [112] ● Dextromethorphan/ bupro­ some countries (e.g. the
● Dextromethorphan/bupro­ pion: Dizziness, nausea, United States) [23]
pion increases norepi­ headache, diarrhea, som­
nephrine availability by nolence, dry mouth [24] ● Some treatment-
inhibiting reuptake and emergent adverse events
acting as alpha-4-beta-2 with ketamine are dose
nicotinic antagonists [56] dependent
● In cases of discontinua­
Dextromethorphan is an tion, a transitions-of-care
uncompetitive antagonist plan is necessary for
of the NMDA receptor and ongoing surveillance of
a sigma-1 receptor agonist. depressive symptoms
Bupropion increases [23]
plasma levels of dextro­
methorphan by inhibiting Dextromethorphan/
CYP2D6, and may be bupropion:
related to noradrenergic ● Rapid efficacy compared
and/or dopaminergic to antidepressant mono­
mechanisms [56,59] therapy [56]
● May increase seizure risk
in patients taking higher
doses, and with certain
other medicines and
medical conditions [59]
● To be avoided for
patients who have or had
seizure disorder or eating
disorder, and if a MAOI is
used in the last 14 days
[59]
● May increase risk of
hypertension [59]
a
Not all are on-label. Consult the prescribing guide for the specific antidepressant.
b
Not currently available for use in the United States.
5-HT: serotonin, AE: adverse event, GAD: generalized anxiety disorder, GI: gastrointestinal, CYP2D6: cytochrome P450 2D6, MAOI: monoamine oxidase inhibitor,
MDD: major depressive disorder, MOA: mechanism of action, NMDA receptor: N-methyl-D-aspartate receptor, NSAID: nonsteroidal anti-inflammatory drug, OCD:
obsessive-compulsive disorder, PTSD: posttraumatic stress disorder, REMS: Risk Evaluation and Mitigation Strategy, SNRI: serotonin-norepinephrine reuptake
inhibitor, SSRI: selective serotonin reuptake inhibitor, TCA: tricyclic antidepressant, TESD: treatment-emergent sexual dysfunction, TRD: treatment-resistant
depression, WBC: white blood cell

randomized controlled trials (RCTs) [12,23–26]. Below we discuss sedation, resulting in relatively high discontinuation rates.
some of the key safety and prescribing considerations for each. They can also block cardiac sodium channels, which in the
case of overdose can lead to sudden cardiac death [12]. Once
SSRIs and SNRIs became available, there was generally less use
4.1. TCAs and MAOIs of TCAs, but they continue to be of use for patients with
TCAs and MAOIs were the first antidepressants to be introduced comorbid insomnia, pain, and social anxiety disorder, and in
following the discovery of their antidepressant properties in the severe or resistant MDD with melancholia [8,19].
1950s [27,28]. TCAs act primarily by elevating extracellular ser­ The broad mechanism of action (MOA) of MAOIs means
otonin and norepinephrine levels via uptake inhibition [12]. that they have significant adverse events, such as weight gain,
TCAs are prone to anticholinergic side effects (e.g. dry mouth, fatigue, and sexual side effects, making them now used almost
blurry vision, constipation, urinary retention), which often limit exclusively in patients who have not responded to other anti­
their utility. TCAs can cause significant weight gain and depressant classes or those with MDD with atypical features,
POSTGRADUATE MEDICINE 455

such as reactive moods or sensitivity to rejection [8,28,29]. particular, may cause sexual dysfunction [33,37]. Other side
Early, nonselective MAOIs had potentially lethal interactions effects include fatigue, insomnia, and headache [33].
with food, particularly foods rich in tyramine, and with other Antidepressants, and in particular SSRIs and SNRIs, are also
medications, which can precipitate fatal serotonin syndrome associated with emotional blunting or indifference [20,22].
or hypertensive crises [12]. Because of this, MAOIs should not This may be underrecognized by both patients and clinicians
be used in antidepressant augmentation with SSRIs because of and poses a challenge for effective treatment because it is
a potentially lethal increase in serotonin in the central nervous frequently a cause for treatment discontinuation [20,22]. Later
system (CNS) [29,30]. There is some evidence that the risk and in this review, we have a dedicated section on treatment
severity of adverse dietary reactions with MAOIs may be less strategies for emergent emotional blunting in MDD.
severe than previously thought, especially when proper mon­ Another issue associated with the use of SSRIs and SNRIs is
itoring by clinicians and adherence by patients are followed the emergence of withdrawal symptoms of varying severity
[29,31]. following discontinuation or interruption of treatment [33,39].
Symptoms of this so-called “discontinuation syndrome” can
include tremors, sweating, tachycardia, agitation, neuralgia,
sleep disturbances, vivid dreams, GI symptoms, worsening
4.2. SSRIs and SNRIs
anxiety, and mood instability [33,39]. Symptoms can be unpre­
SSRIs are the most widely used antidepressants in contempor­ dictable and may affect everyone differently, emerging within
ary treatment for MDD and include fluoxetine, sertraline, par­ a few days to weeks and potentially persisting for months or
oxetine, fluvoxamine, citalopram, and escitalopram [8,12,32]. even years [33,39]. In particular, sertraline and paroxetine of
A large body of literature supports the efficacy of SSRIs com­ the SSRIs, and venlafaxine and its active metabolite, desvenla­
pared with placebo for the treatment of MDD, and their faxine, of the SNRIs, have been associated with significant
relative safety has driven a sharp increase in the prescribing withdrawal symptoms, which may be due to their shorter half-
of pharmacotherapy for MDD [8,12,32]. Within the class, SSRIs lives [12,33,37,39,40]. Tapering of the treatment over weeks to
have noteworthy yet nuanced differences in their pharmaco­ months may help to mitigate symptoms related to disconti­
logic profiles that may impact their clinical application, of nuation of SSRIs and SNRIs [33].
which prescribers should be aware [12]. A slower emergence
of the therapeutic effects of fluoxetine compared with other
4.3. Atypicals
SSRIs is one example. Further, some SSRIs are also specifically
approved to treat panic disorder, social anxiety disorder, 4.3.1 Mirtazapine: A pooled analysis showed fast onset of
obsessive-compulsive disorder (OCD), posttraumatic stress dis­ antidepressant effect with mirtazapine, and a meta-analysis
order (PTSD), bulimia nervosa, and/or premenstrual dysphoric has confirmed accelerated onset of efficacy of mirtazapine
disorder. Sertraline and paroxetine have several FDA approvals but equal efficacy by week 8 compared with SSRIs (fluoxetine,
and indications to treat a myriad of psychiatric disorders [12]. citalopram, paroxetine, and sertraline) [41]. Mirtazapine has
All SSRIs may cause nausea, headaches, gastrointestinal (GI) significant sedation properties, tends to promote weight gain
issues, insomnia, fatigue, weight gain, anxiety, dizziness, dry and causes dry mouth, but has a low rate of sexual dysfunc­
mouth, and sexual side effects and have been associated with tion [8,12]. The sedative, antiemetic, anxiolytic, and appetite-
increased risk of falls and fractures [8,27,33,34]. Paroxetine stimulating effects of mirtazapine suggest that it can also be
eventually may cause more weight gain, and paroxetine and prescribed off-label for insomnia, panic disorder, PTSD, OCD,
citalopram may be the most sedating. Sertraline may have generalized anxiety disorder (GAD), social anxiety disorder,
more adverse GI effects, and paroxetine and fluoxetine have headaches, and migraines [42,43]. Mirtazapine will generally
more drug-drug interactions via the cytochrome P450 (CYP) be prescribed for MDD after an SSRI, or in depressed adults
2D6 hepatic enzyme inhibition [12]. SSRIs, particularly citalo­ with comorbid insomnia or those who are underweight [42].
pram, have the potential to prolong the QT interval, which can 4.3.2 Trazodone: Trazodone was an early second-generation
lead to torsade de pointes, a rare but potentially fatal arryth­ antidepressant whose low side-effect and toxicity profile made
mia [32,35]. SSRIs have also been associated with coagulopa­ it a popular alternative to MAOIs and TCAs in the early 1980s;
thy, and use in combination with pain relievers such as aspirin, it is still relatively widely used [8,12]. It is considered a sedating
naproxen, and blood thinners may increase the risk of bleed­ antidepressant, making it suited to patients with comorbid
ing [32,36]. In addition, SSRIs may cause the syndrome of insomnia or GAD [44,45]. A slow-release preparation is avail­
inappropriate antidiuretic hormone secretion, with the great­ able that lowers plasma levels and tends to minimize the
est risk among elderly patients. sedating side effects relative to the standard formulation
SNRIs include venlafaxine, desvenlafaxine (the active meta­ [46]. Nefazodone has a structure analogous to trazodone but
bolite of venlafaxine), duloxetine, milnacipran, and levomilna­ has different pharmacologic properties. It has largely fallen out
cipran [37]. Each of these medications is efficacious (superior of use because of concerns about hepatotoxicity and the need
to placebo in controlled studies and meta-analyses), and ven­ for periodic liver function monitoring [12,44].
lafaxine (75–150 mg/day) and duloxetine (60 mg/day) showed 4.3.3 Agomelatine: Agomelatine was first marketed in
comparable efficacy in a pair of similarly designed RCTs Europe in 2009 and has been available in Australia since
directly comparing these 2 antidepressants [8,38]. Side effects 2010 but is not currently available in the United States [47].
most common to the class of SNRIs include nausea and vomit­ Agomelatine has a unique MOA, targeting both melatonergic
ing, dizziness, and sweating. Duloxetine and venlafaxine, in and serotonergic receptors, with demonstrated superior
456 C. B. MONTANO ET AL.

effects on subjective or objective sleep measures, in addition for MDD with suicidality [23]. However, both ketamine
to being an effective antidepressant [48]. In countries where it and esketamine carry potential for clinically significant
is available, it is often recommended for individuals with MDD adverse effects, abuse, and misuse, and have limited [23]
and sleep disturbances [48]. While agomelatine has been evidence of long-term efficacy. Patient selection for these
demonstrated to be safe and effective, there is potential for agents should involve thorough evaluation of treatment
drug interactions mediated via CYP1A2, and it is also asso­ history and other physical and psychological factors [58],
ciated with liver damage with longer-term use and is contra­ and it is recommended that a psychiatrist or other health­
indicated in patients with impaired liver function [47]. care provider with expertise in the treatment of patients
4.3.4 Bupropion: Bupropion was initially approved in the with mood disorders conduct a psychiatric assessment
United States in the 1980s as an antidepressant but sub­ before and after treatment [23]. In addition, clinicians
sequently removed from the market because of induction must exercise caution when prescribing, factoring in rele­
of seizures in patients with MDD. It was remarketed after vant regulatory requirements, such as the Risk Evaluation
lower doses were noted to be safer [12,49]. A meta-analysis and Mitigation Strategy drug safety program [23].
of 7 RCTs demonstrated comparable efficacy between 4.5.2 Dextromethorphan-bupropion: Similar to ketamine,
bupropion and the SSRIs fluoxetine, sertraline, and parox­ dextromethorphan is an NMDA receptor antagonist with
etine [41]. The side-effect profile of bupropion reflects its multimodal activity [56]; however, its clinical use has been
unique MOA, having no sexual dysfunction or weight gain limited by the difficulty in achieving therapeutic plasma
tendencies [12], yet it is also more activating with regard levels due to its rapid metabolism via cytochrome P450
to insomnia and anxiety, causing agitation, mild cognitive 2D6 (CYP2D6) [56]. An oral tablet combining dextromethor­
dysfunction, and GI upset, and should be avoided in indi­ phan with bupropion, a known antidepressant and inhibitor
viduals with comorbid eating disorders because of the of CYP2D6 that increases the bioavailability and half-life of
increased risk of seizures [8]. It is frequently prescribed as dextromethorphan and leads to sustained therapeutic con­
a combination antidepressant, often added to an initially centrations [24,56], has shown promising results in a 6-week
partially effective SSRI [50]. randomized, double-blind, placebo-controlled, phase 3
study [24]; significant improvements in depressive symp­
toms compared to placebo were demonstrated as early as
4.4. Serotonin modulators 1 week after treatment initiation [24]. Notably, dextro­
methorphan-bupropion was not associated with psychomi­
4.4.1 Vilazodone and vortioxetine: Vilazodone and vortioxe­
metic effects. The FDA granted approval for its use in MDD
tine are more recent entries to the antidepressant space,
in August 2022 [59]; however, additional data investigating
and each may be considered a “serotonin modulator and
long-term efficacy and safety will help define its role in the
stimulator” or multimodal antidepressant [12,51]. Vilazodone
MDD treatment landscape [60].
appears to have a lower risk of weight gain and sexual side
effects than the SSRIs or SNRIs based on noncomparative
trials [12,52,53]. Vilazodone should be taken with food to
ensure adequate absorption, and a titrated dosing schedule 5. Assessment of suicide risk in patients starting
is recommended to minimize GI effects that are frequently antidepressant therapy
observed [48,53].
When initiating patients on antidepressant therapy, suicide
Vortioxetine has demonstrated suitability for patients with
risk must be monitored, as some antidepressants have been
MDD aged 18 to 88 years, including those experiencing treat­
associated with an increased risk of suicidal thoughts and
ment-related sexual dysfunction with other antidepressants,
behaviors, particularly in children, adolescents, and young
and those with deficits in cognitive processing speed [54,55].
adults [48,61,62]. In 2004, the FDA directed manufacturers
GI symptoms, which were dose dependent, were the most
of all antidepressants to revise their labeling as the result of
common adverse events in clinical trials. Vortioxetine can be
an increase in suicidality among children and adolescents
taken with or without food [52,55].
being treated with antidepressants. This warning was based
on a combined analysis of 24 short-term (up to 4 months),
placebo-controlled trials of 9 antidepressant drugs among
4.5. NMDA receptor antagonists
more than 4400 children and adolescents with MDD or
4.5.1 Ketamine and esketamine: The delayed onset of effect other psychiatric disorders. A greater risk of suicide during
seen with most traditional antidepressants has led to great the first few months of treatment was observed in patients
interest in the use of agents with a rapid onset of effect receiving antidepressants (4% vs. 2% in patients receiving
[56]. NMDA receptor antagonists [23], such as ketamine placebo) [62]. In 2007, the FDA revised the warning, chan­
and esketamine, have demonstrated a rapid and robust ging the target period from childhood and adolescence to
antidepressant effect in adults with treatment-resistant young adulthood (aged 18–24 years) during initial treat­
depression (TRD), defined as lack of response to at least ment [62]. However, clinicians should continue to monitor
2 adequate antidepressant treatments from different phar­ for suicidality in all patients being treated for depression
macological classes [57], with esketamine also approved with or without antidepressant medication.
POSTGRADUATE MEDICINE 457

6. Selecting pharmacotherapy for the treatment of For acceptability (defined as treatment discontinuations due to
MDD: what do the guidelines say? Factors to any cause), agomelatine, citalopram, escitalopram, fluoxetine,
consider when initiating antidepressant treatment in sertraline, and vortioxetine were more tolerable than other anti­
primary care settings depressants, whereas amitriptyline, clomipramine, duloxetine,
fluvoxamine, reboxetine, trazodone, and venlafaxine had the
6.1. Efficacy
highest dropout rates [64]. The authors noted that the results
Given that depression is a heterogeneous illness with multiple should inform guideline and policy developers on the relative
clinical presentations, antidepressant selection should be merits of the different antidepressants available [64]. Until there
based on patient profile and tolerability, especially considering is consensus regarding the superiority of one class of antide­
any comorbidities (for example, ADHD) [12,63]. Clinical prac­ pressant over another, initial selection of an antidepressant will
tice guidelines for the treatment of MDD generally emphasize be based largely on cost, patient preference and previous
the importance of tailoring the selected therapy to the patient responses, anticipated side effects, nuances of patient’s clinical
based on their clinical profile and individual preferences profile including comorbidities, and their specific and most
(Figures 2 and 3) [3,8,48]. For example, the 2020 Royal severe/dominant symptoms (Figure 2) [3,8]. Additionally, phar­
Australian and New Zealand College of Psychiatrists (RANZP) macologic properties of the medication (e.g. half-life, actions on
clinical practice guidelines for major depression note that CYP enzymes, other drug interactions) must be considered [49].
there is a modest hierarchy with respect to efficacy of anti­
depressants. These guidelines state that, generally, the medi­
cations with a broader spectrum of action, such as TCAs,
7. Patient clinical profile and comorbidities
appear to be more clinically efficacious than those that are
more selective [3]. Although TCAs and MAOIs may exert effec­ It is logical that an initial antidepressant is selected based on
tiveness in treating depression, their safety profiles are of the most prominent/severe depressive symptoms experienced
significant concern [12]. Hence, more selective may mean by the patient (e.g. suicide risk, sleep disturbances, cognitive
less effective in some patients. However, the 2010 American disturbances, anxiety, somatic symptoms, or atypical features
Psychiatric Association (APA) Practice Guideline for the such as oversleeping and overeating) [28,48]. Clinical practice
Treatment of Patients With Major Depressive Disorder states guidelines address the different depression subtypes that can
that the effectiveness of antidepressants is generally compar­ be an important modifier of antidepressant efficacy, such as
able between and within classes of medications [8]. The depression with melancholic features (psychomotor slowing,
Canadian Network for Mood and Anxiety Treatments diurnal mood variation), depression with anxious distress, or
(CANMAT) 2016 Clinical Guidelines for the Management of depression with psychotic or catatonic features [3,48]. The
Adults With Major Depressive Disorder provide evidence of Korean Medication Algorithm Project for Depressive Disorder
superiority of some antidepressants over others. However, provides first- and second-line recommendations for nonpsy­
they also emphasize the importance of tailoring pharmacolo­ chotic depression, psychotic depression, and specific depres­
gic treatment to the patient’s symptoms and preferences, sion subtypes [62]. Other patient-related factors, such as cost,
similarly stressing that there are no absolutes [48]. age, level of sexual activity, comorbid conditions, preferences,
A comprehensive and relatively recent network meta-analysis use of concomitant medications, response to prior antidepres­
by Cipriani et al. consisting of 522 clinical trials and 116,477 sants, and pregnancy status, all become necessary considera­
participants with acute MDD reported that in head-to-head tions as well [8,48]. In addition, pharmacogenetic testing can
studies, agomelatine, amitriptyline, escitalopram, mirtazapine, sometimes guide providers in choosing potential pharma­
paroxetine, venlafaxine, and vortioxetine were more effective cotherapy, and patients may qualify for enrollment in clinical
than other antidepressants, whereas fluoxetine, fluvoxamine, trials, as multiple neurotransmitter and receptor targets are
reboxetine, and trazodone were the least efficacious drugs [64]. currently under investigation and may help guide and

Figure 2. Treating MDD in primary care: factors influencing initial antidepressant selection [3,8,84].
458 C. B. MONTANO ET AL.

Figure 3. Summary algorithm for selecting an antidepressant [3,8,48,88].

improve patient outcomes, including time to response and antidepressant that suits their preferences while also being
complete remission of depression [65,66]. best suited to their clinical profile [3]. The concept of the
“therapeutic alliance” between the patient and provider is
important in primary care settings, with the aim being to
8. Tolerability and adherence
establish an effective partnership, taking into consideration
The 2010 APA guideline states that ”the optimal regimen is the patient’s preferences and ensuring that they adhere to
the one that the patient prefers and will adhere to [8].” treatment and do not discontinue due to issues with toler­
Tolerability and patient preference therefore become critical ability without first speaking with their provider. This gives
because they relate directly to medication adherence, which patients the greatest chance of achieving response, remission,
has implications for response and remission, and antidepres­ and sustained functional recovery while on therapy [8].
sants differ in their potential to cause certain side effects (as Measurement-guided care, which uses validated screening
described in the previous section and Table 1) [8,67]. Indeed, and monitoring tools, is encouraged for use by clinicians to
an important part of patient satisfaction with MDD treatment quantify the presence and severity of depressive symptoms
comes from adequately addressing concerns about side and track improvements in functional outcomes and adverse
effects such as weight gain and sexual dysfunction. In events following initiation of an antidepressant. The incor­
a study of 180 patients receiving treatment for MDD, key poration of measurement-guided care increases the likelihood
concerns expressed by patients included weight gain, with­ of remission, treatment adherence, and sustained functional
drawal effects, and sexual dysfunction [68]. However, other recovery [8,69].
side effects commonly associated with antidepressants, of
which patients may be less aware, include hypertensive crises,
10. Guideline recommendations for antidepressants
serotonin syndrome, and cardiovascular and neurological
effects, all of which are important for the prescriber to con­ There is considerable variability among clinical practice guide­
sider [8]. Other factors that can influence adherence include lines. The 2010 APA guideline describes the different classes of
the patient’s clinical profile, sociocultural background, finan­ antidepressants but does not make prescriptive recommenda­
cial factors, and beliefs regarding the effectiveness of the tions concerning their selection [8]. The 2016 CANMAT guide­
treatment. This is where physician expertise and a strong lines list SSRIs, SNRIs, agomelatine, bupropion, mirtazapine,
doctor-patient relationship become vital [67]. and vortioxetine as first-line recommendations [48]. Second-
line recommendations include TCAs, quetiapine, and trazo­
done (owing to higher side-effect burden); moclobemide and
9. Patient preference and shared decision-making
selegiline (potential serious drug interactions); levomilnacipran
From the perspective of the primary care provider, physician (lack of comparative and relapse-prevention data); and vilazo­
expertise can help guide the patient to select the done (lack of comparative and relapse-prevention data and
POSTGRADUATE MEDICINE 459

the need to titrate and take with food) [48]. Third-line recom­ 11.2. Assessing treatment adherence and reevaluating
mendations include MAOIs (owing to higher side-effect bur­ diagnosis: bipolar disorder (BPD) and comorbid ADHD as
den and potential serious drug and dietary interactions) and examples
reboxetine (lower efficacy) [48]. The 2020 RANZP guidelines
Unipolar depression is a diagnosis of exclusion, as bipolar
for major depression list 7 molecules with different MOAs that
depression, or BPD, must first be ruled out. Often the first
they recommend as “Choice” antidepressants, which is
presentation of BPD is a major depressive episode (MDE),
a unique approach to treatment recommendations (escitalo­
which may be indistinguishable from that of unipolar depres­
pram, vortioxetine, agomelatine, venlafaxine, mirtazapine,
sion [70]. Up to 25% of patients presenting with an MDE may
bupropion, amitriptyline) [3]. The RANZP guidelines also pro­
in fact have BPD [71]. Screening tools are available for eva­
vide guidance on selecting these molecules according to the
luation of BPD in primary care, such as the Rapid Mood
patient’s clinical profile and to minimize side effects [3]. While
Screener, a validated scale, and the Mood Disorder
many if not all recommendations provided by the guidelines
Questionnaire; both are time-sensitive and extremely helpful
discussed may continue to be applicable, updates are needed,
for ruling out BPD [70]. It is also important to rule out BPD
particularly for inclusion of newer agents such as NMDA recep­
because there is evidence suggesting that antidepressant
tor antagonists.
monotherapy in individuals with BPD can induce states of
hypomania, mood destabilization, and potential increase in
suicidality [72,73]. The consensus is that individuals diag­
11. Partial or inadequate response to initial nosed with BPD should not be treated with antidepressant
antidepressant monotherapy as a first-line strategy, as mood stabilizers
and second-generation antipsychotics are considered to be
Regardless of the intervention used, a substantial proportion
more appropriate and are less likely to cause mood cycling
of patients do not adequately respond or achieve remission
[72,74]. Once BPD is excluded, the APA guideline suggests
after initial treatment. For example, about 40% of patients
that if at least moderate improvement in symptoms of uni­
treated with second-generation antidepressants do not
polar depression is not observed within 4–8 weeks of treat­
respond, and approximately 70% do not achieve remission
ment initiation, the diagnosis should be reassessed.
[10]. Partial or inadequate response is important to address
Antidepressant side effects should be taken into considera­
because a lack of early improvement in symptoms (defined in
tion, complicating co-occurring conditions and psychosocial
RCTs as a 20%–30% reduction from baseline in a depression
factors reviewed, and the treatment plan adjusted accord­
rating scale after 2–4 weeks) is a predictor of later antidepres­
ingly [8,75,76]. Other factors to consider include pharmaco­
sant nonresponse/nonremission. However, there is only low-
kinetic or pharmacodynamic influences on efficacy, as blood
quality evidence to support early switching at 2 or 4 weeks for
levels of antidepressants can help determine if dosage
nonresponders to an initial antidepressant [48]. Inadequate
adjustments are needed [8]. Treatment adherence should
responses to antidepressants can also prompt nonadherence,
also be evaluated [8], as there is evidence that up to 20%
which can be an important driver of poor response to an
of patients may have poor adherence [48]. This should be
antidepressant therapy [3,67]. In primary care settings where
done before moving on to a different antidepressant.
defining partial or inadequate response may be ambiguous,
ADHD is frequently comorbid with both BPD and MDD, and
measurement-guided care using time-efficient patient- and
a mean prevalence of 7.8% of comorbid ADHD in individuals with
clinician-completed rating scales may help with patient mon­
depression has been reported, complicating identification and
itoring following initiation of an antidepressant (Figure 3). This
management of each disorder [16,63,77]. Depressive symptoms
can assist both the patient and provider in tracking and clin­
in adults with ADHD may manifest as a result of coping with
ical management of symptoms and functioning over time,
lower hedonic tone, and in one study, up to 28% of individuals
identifying factors influencing inadequate response, and
referred to a tertiary clinic for mood and anxiety disorders had
understanding if treatment adjustments are needed [8,69].
undetected ADHD [16,78]. Symptoms of ADHD may also be
masked by substance use disorder, as individuals with ADHD
have double the risk of substance abuse and dependence com­
11.1. Increasing the dose/duration of antidepressants pared with the general population [79]. Symptoms of ADHD
Almost all approved antidepressants have an initial dose that respond best to catecholaminergic agents such as psychostimu­
can be incrementally increased depending on the patient’s lants or norepinephrine-dopamine reuptake inhibitors such as
response and tolerability [8]. Partial responses to antidepres­ bupropion, which has shown potential off-label use for treat­
sants can be addressed by first increasing the dosage at 2 to ment of ADHD [80]. In addition, viloxazine, an SNRI previously
4 weeks if tolerated [3,48]. In addition, the 2010 APA guideline used for treatment of MDD in Europe, was recently approved by
suggests doing this after 4–8 weeks of treatment and provides the FDA for treatment of ADHD and has shown promise as
guidance for the starting dose of the different classes of anti­ a nonstimulant option [27,81,82]. Serotonergic agents alone do
depressants and the usual daily dosage, which should be not improve symptoms of ADHD; thus, initiation of treatment
increased incrementally [8]. Prescribing information for each with an SSRI alone for MDD in an individual with comorbid ADHD
antidepressant can also be consulted for the most up-to-date is likely to yield unsatisfactory response to treatment for either
dosing information. MDD or ADHD [16].
460 C. B. MONTANO ET AL.

In primary care settings where depressive symptoms may recovery, as well as of the impact of comorbid symptoms such as
be more frequently reported by adults and therefore more distractibility, sleep disturbance/apnea, emotional blunting, and
easily diagnosed, clinicians may benefit from asking adult anxiety. This section aims to highlight recently published data on
patients presenting with depressive symptoms questions antidepressant effectiveness for these specific issues where lit­
about previous diagnosis or family history of ADHD, or erature gaps have been identified. We conducted a search of
whether they had difficulty in school. This may help to identify articles in English from 2013 to the present pertaining to use of
comorbid ADHD in their patients [77]. antidepressants for functional recovery, and comorbid symp­
toms such as sleep disturbances, emotional blunting, and anxi­
ety. Data continue to emerge on a range of antidepressants that
11.3. Combination, switching, and augmentation
may be prescribed to address these specific challenges in indivi­
strategies
duals with MDD, and longer-term and real-world studies are
In clinical practice guidelines, there is no consistent evidence providing insights into which antidepressants may be best suited
supporting switching within or between antidepressant classes. for patients experiencing specific symptoms. Of note is that
Switching within a class has been noted as acceptable if the distinguishing symptoms such as sleep disturbances or emo­
antidepressant was not taken as prescribed because of poor tional blunting with antidepressant therapy from residual symp­
adherence or side effects; however, there is limited guidance on toms of MDD remain a challenge.
how to switch to a different antidepressant in the same class
[3,48]. There are few RCTs comparing a switch strategy to continu­
12.1. Antidepressant therapy, cognitive dysfunction, and
ing the same antidepressant, and thus the value of switching
functional outcomes
between or within classes remains controversial [48]. One theory
regarding why patients often respond suboptimally to their initial In 2015, CANMAT published recommendations for assessment
antidepressant is that receptor tolerance to drug action precipi­ of functional outcomes in individuals with MDD, which high­
tates antidepressant nonresponse, so treatment switching to lighted the critical impact of depressive symptoms on social,
a different class could be considered a reasonable strategy [83]. occupational, and physical functioning [48,86]. Functional sta­
There is limited information about which treatments may be best tus relates to an individual’s ability to “perform the tasks of
suited as second-line therapies in patients who have not achieved daily life and to engage in mutual relationships with other
an adequate response with their initial antidepressant, but the people in ways that are gratifying and that meet the needs of
selection of the subsequent therapy should account for the class/ the individual and the community in which they live” [86]. The
MOA and response to initial antidepressant [84]. CANMAT guidelines emphasized that recovery from depres­
Combination and augmentation strategies are additional sion must involve both relief of symptoms and improvement
approaches to consider if the patient continues to have of functioning, but systematic reviews show that functional
a partial or inadequate response at the maximally tolerated outcomes are only modestly correlated with symptom out­
dose of initial therapy after an adequate duration of treatment comes, and functional improvement may lag behind symptom
[3,62,76]. The combination of 2 antidepressants with different improvement, which is prioritized as an outcome in the major­
MOAs may have synergistic effects that enhance response in ity of RCTs [48,86,87].
inadequate responders, and there is some evidence support­ Symptoms of cognitive impairment may have a particularly
ing the addition of bupropion to an SSRI [8,76]. Further, pre­ adverse effect on functional outcomes and have been linked to
scribers must be aware of the risk for serotonin syndrome psychosocial dysfunction, patient employment status, and ability
when combining antidepressants. to functionally recover [88–91]. Further, antidepressants that are
Augmentation typically involves the addition of a non- best suited to treat cognitive symptoms may lead to improve­
antidepressant to a current antidepressant. As we have men­ ments in functional outcomes for patients, but there are few
tioned, multiple comorbidities can drive the patient to experi­ studies that address this subject. PERFORM-J, a 6-month, non­
ence symptoms of depression, which must be kept in mind interventional, prospective, longitudinal study, investigated the
when approaching augmentation and combination strategies. relationship among cognitive disturbances, severity of depres­
Augmentation with lithium and atypical antipsychotics (e.g. sive symptoms, and social functioning in patients with MDD in
aripiprazole, cariprazine [85], brexpiprazole, risperidone, que­ Japan. Notably, improvement of depressive symptoms in the
tiapine, or olanzapine) may be appropriate strategies to con­ early stages of antidepressant treatment was associated with
sider for non- or partial responders who may have mixed a reduced risk of residual cognitive symptoms after 6 months
features, TRD, or BPD, for example [3,8,48]. For adults with in patients with MDD, highlighting the importance of early
MDD who have comorbid ADHD, most antidepressants can be improvements in depressive symptoms for long-term remission
combined (with monitoring) with long- or short-acting stimu­ and the need for early and aggressive monitoring and treatment
lants to treat ADHD symptoms [16,63,77,79]. Long-acting pre­ modifications with measurement-guided care [91,92]. Also, resi­
parations are preferred when using psychostimulants. dual cognitive disturbances have been demonstrated in indivi­
duals with MDD even after achieving remission from mood
symptoms, which can have an impact on long-term outcomes
12. Antidepressant therapy for MDD: filling the
including remission, relapse, and social functioning [92]. In
literature gaps
a 3-year follow-up study, approximately 44% of individuals with
There is heightened awareness of the ability of antidepressants MDD who reported partial/full remission with treatment contin­
to aid patients with MDD struggling to achieve full functional ued to experience impairments in cognitive function despite the
POSTGRADUATE MEDICINE 461

resolution of their depressed mood [90]. The precise mechanisms 12.2. Antidepressant therapy and sleep disturbances
for the lasting cognitive impairment, even in remitters from
Sleep disturbance is a prominent symptom in patients with
mood symptoms, are unclear, but it is possible that neurobiolo­
MDD, and depressed patients with sleep disturbance are more
gical insults due to recurring depressive episodes may be respon­
likely to present with more severe symptoms and difficulties
sible (some patients with MDD show atrophy of the
with treatment [94,95]. Insomnia is a predisposing factor for
hippocampus, which may be associated with lingering cognitive
the development of new or recurring depression in young and
symptoms) [92].
older adults [94]. Persistent insomnia is the most common
Baune and colleagues conducted a systematic review of 35
residual symptom in depressed patients, is considered a vital
studies evaluating pharmacologic and nonpharmacologic
predictor of depression relapse, and may contribute to
treatments of cognitive impairment primarily in the domains
adverse clinical outcomes [94]. There are several different
of memory, attention, processing speed, and executive func­
proposed mechanisms involved in sleep disturbances and
tion in clinical depression [89]. The results showed that various
MDD; most notably for patients with MDD, there may be
classes of antidepressants exert improving effects on cognitive
a disruption to circadian rhythm, specifically rapid eye move­
function across several cognitive domains. Specifically, SSRIs,
ment (REM) sleep disturbances and dysfunction of the mono­
the selective serotonin reuptake enhancer tianeptine, the SNRI
aminergic and cholinergic systems [94,95]. Improving sleep
duloxetine, vortioxetine, and other antidepressants such as
outcomes in patients with MDD should be prioritized in pri­
bupropion and moclobemide may exert specific positive
mary care; however, some antidepressants can worsen sleep
effects on cognitive function in depression, such as learning,
quality due to their role in modulating levels of neurotrans­
memory, and executive function [89]. In addition to their
mitters involved in the sleep-wake cycle. These include the
positive effects on cognitive function, sertraline, citalopram,
SNRIs, MAOIs, SNRIs, SSRIs, and TCAs [94,96].
and nortriptyline or paroxetine improved overall psychosocial
Polysomnography studies have reported that SSRIs, SNRIs,
functioning, and duloxetine improved overall social function­
and activating TCAs increase REM sleep latency, suppress
ing and enjoyment of life, which may be due in part to its
REM sleep, and impair sleep continuity. Sedating antidepres­
pain-alleviating effect [89]. Further, in a separate analysis, Pan
sants, such as mirtazapine and trazodone, decrease sleep
and colleagues note that while most antidepressants exert
latency, ameliorate sleep efficiency, and increase slow-wave
procognitive effects, vortioxetine and duloxetine are the only
sleep, with little effect on REM sleep. In this regard, sedating
antidepressants that have established these effects when
antidepressants are favorable for patients with comorbid
a rigorous methodology is used [90].
depression and sleep disturbance [94]. In a meta-analysis of
A recent publication reported on the real-world effective­
276 RCTs exploring insomnia and somnolence associated
ness of vortioxetine for functional outcomes in patients with
with second-generation antidepressants, 10 of 14 antidepres­
MDD. In this 24-week observational study of outpatients initi­
sants showed significantly higher rates of insomnia than pla­
ating MDD treatment with vortioxetine, the Sheehan Disability
cebo, with the following increasing order of incidence:
Scale (SDS) was used as the main outcome measure to evalu­
escitalopram, duloxetine, venlafaxine, paroxetine, reboxetine,
ate patient functioning. Significant improvements in function­
fluoxetine, fluvoxamine, sertraline, desvenlafaxine, and bupro­
ing were observed, and the proportion of patients with severe
pion. Agomelatine was the only antidepressant that had
functional impairment decreased substantially [93]. Sustained
a lower likelihood of inducing insomnia than placebo.
improvements in functioning were seen over the 24 weeks of
Mirtazapine, milnacipran, and citalopram did not differ from
treatment irrespective of vortioxetine treatment line [93].
placebo in terms of insomnia rates [96].
In one of the few studies directly assessing antidepres­
sant effects on functional outcomes, a meta-analysis of 17
RCTs exploring occupational (workplace) impairment in
12.3. Emotional blunting
MDD reported a small positive effect versus placebo of
duloxetine, desvenlafaxine, venlafaxine, paroxetine, escitalo­ Limited evidence-based information exists on how emotional
pram, levomilnacipran, and vortioxetine on occupational blunting, or “numbness” of both positive and negative emo­
impairment in the short term as measured by the SDS- tions caused by antidepressant therapy, may be addressed in
work subscale. However, the authors questioned the overall individuals with MDD [22]. Patients with MDD who suffer from
clinical significance of the findings due to a small effect size emotional blunting are subject to a reduction in a broader
[87]. While the ability of antidepressants to exert positive range of emotions, including love, affection, fear, and anger,
effects on cognition is relatively well studied, more large- which is distinct from anhedonia and apathy, and may lead to
scale, longer-term studies that directly assess their impact reduced quality of life or reduced social responsibilities [22].
on functional outcomes are needed to be able to provide Emotional blunting is frequently reported by patients receiv­
more robust treatment recommendations for one antide­ ing antidepressant treatment for MDD, particularly SSRIs, and
pressant over another [48,87,89]. From a primary care per­ is considered to be one of the most prominent side effects
spective, sustained functional recovery in patients with MDD leading to treatment discontinuation [21,22,97].
should be considered an important treatment goal [91], and Approximately 40%–60% of patients who suffered from MDD
providers should maintain a partnership with their patients and were treated with either SSRIs, SNRIs, and TCAs experi­
to monitor functional outcomes with antidepressant therapy enced some degree of emotional blunting [97,98]. Some stu­
over time. dies suggest that emotional blunting is not simply a side effect
462 C. B. MONTANO ET AL.

of antidepressants, but also a residual symptom of depression one week, with dose reductions permitted based on individual
due to incomplete treatment, adding further complexity to tolerability [106].
understanding how it may be mitigated by appropriate treat­
ment selection [22,98].
Studies aimed at understanding the etiology of antidepres­
Conclusions
sant-induced emotional blunting have generally focused on
SSRIs and a possible modulating effect on frontal lobe activity The multitude of different types of antidepressants for MDD
via serotonergic signaling [22]. Other studies hypothesize represents both an opportunity and a challenge for primary care
a mutually opposed relationship between serotonergic and providers who must tailor the antidepressant to their patient.
dopaminergic systems, which may have inhibitory effects on Practitioners prescribing antidepressants in primary care can
rewarding and aversive stimuli [22,99]. Emotional blunting achieve better outcomes for their patients and help prevent
with antidepressants appears to be dose related, with several them from cycling through a range of different antidepressants
case studies noting a reduction in emotional blunting with an with inadequate response through careful selection of an initial
incremental tapering of the dose of SSRI. Thus, from a clinical antidepressant therapy. An initial therapy that fits the patient’s
perspective, dose reduction of SSRIs or other antidepressant is clinical profile and comorbidities is most likely to be successful and
a promising strategy if clinically feasible [22]. If there is any tolerated. Providers need to not only focus on relieving the symp­
question as to whether emotional blunting is still of concern, toms related to MDD, but also focus on improving functional
the Oxford Depression Questionnaire should be administered outcomes for their patients. Prioritizing the management of resi­
[100]. Another strategy is to switch to a different class of dual symptoms that can adversely impact response, remission,
antidepressant [22]. In more recent studies, emotional blunt­ and precipitate reoccurrence will help provide the patient with
ing has been found to be less frequent in patients taking the best chance of symptomatic and functional remission.
agomelatine than in those taking escitalopram (an SSRI), and
less severe when medication was switched from SSRIs/SNRIs
to vortioxetine [101,102]. Augmentation may also be consid­
List of Abbreviations
ered; however, the evidence for this as an effective strategy to
address emotional blunting is less robust [22]. 5-HT serotonin
ADHD attention deficit/hyperactivity disorder
AE adverse event
APA American Psychiatric Association
BPD bipolar disorder
12.4. MDD and comorbid anxiety CANMAT Canadian Network for Mood and Anxiety Treatments
CNS central nervous system
Few dedicated trials have been conducted in patients with CYP2D6 cytochrome P450 2D6
anxious depression, as MDD with comorbid GAD has not yet FDA US Food and Drug Administration
been recognized as a distinct disorder. Consequently, specific GAD generalized anxiety disorder
treatment guidelines and recommendations are lacking [103]. GI gastrointestinal
MAOI monoamine oxidase inhibitor
Fortunately, there is considerable therapeutic overlap between MDD major depressive disorder
agents used to treat both anxiety and depression, with many MDE major depressive episode
pharmacotherapies demonstrating efficacy for both disorders MOA mechanism of action
NMDA N-methyl-D-aspartate
[103]. Treatment selection for patients with MDD and comorbid
NSAID nonsteroidal anti-inflammatory drug
anxiety should therefore focus on antidepressants that also OCD obsessive-compulsive disorder
demonstrate efficacy for anxiety, or vice versa, such as the PTSD posttraumatic stress disorder
SSRIs, SNRIs, and serotonin modulators (e.g. vortioxetine and RANZP Royal Australian and New Zealand College of Psychiatrists
RCT randomized controlled trial
vilazodone). Goodwin provides recommendations for treatment
REM rapid eye movement
options for MDD and GAD that include off-label use of TCAs, REMS Risk Evaluation and Mitigation Strategy
agomelatine, vortioxetine, and bupropion [103]. SDS Sheehan Disability Scale
In a meta-analysis of 3 RCTs, vilazodone was found to be SIADH syndrome of inappropriate antidiuretic hormone secretion
SNRI serotonin-norepinephrine reuptake inhibitor
superior to placebo in the short-term treatment of GAD [104].
SSRI selective serotonin reuptake inhibitor
However, a later meta-analysis demonstrated poor tolerability, TCA tricyclic antidepressant
highlighting the need for additional studies to confirm overall TESD treatment-emergent sexual dysfunction
benefit [105]. Recently published data from an 8-week, open- TRD treatment-resistant depression
US United States
label study of vortioxetine in 100 adult patients with severe
WBC white blood cell
MDD and comorbid severe GAD demonstrated clinically mean­
ingful and statistically significant improvements in patient-
and clinician-assessed symptoms of depression and anxiety.
In the trial, vortioxetine was administered as a first-line ther­ Acknowledgments
apy or in patients switching to vortioxetine due to inadequate
Under the direction of the authors, medical writing assistance was provided by
response to another antidepressant [106]. Significant improve­
Katherine Price, PhD, and Leandra Dang, PharmD, on behalf of Syneos Health
ments in overall functioning and health-related quality of life Medical Communications, LLC. Takeda Pharmaceuticals U.S.A., Inc., and
were also noted [106]. In the trial, treatment with vortioxetine H. Lundbeck A/S provided funding to Syneos Health for support in writing
was initiated at 10 mg/day and titrated up to 20 mg/day after this manuscript.
POSTGRADUATE MEDICINE 463

Funding 10. Gartlehner G, Gaynes BN, Amick HR, et al. Comparative benefits
and harms of antidepressant, psychological, complementary, and
This review was funded by Takeda Pharmaceuticals U.S.A., Inc., and exercise treatments for major depression: an evidence report for
H. Lundbeck A/S. a clinical practice guideline from the American College of
Physicians. Ann Intern Med. 2016;164(5):331–341.
11. Ljungberg T, Bondza E, Lethin C. Evidence of the importance of
Declaration of interest dietary habits regarding depressive symptoms and depression.
Int J Environ Res Public Health. 2020;17(5):1616.
C. Brendan Montano has received consulting fees from AbbVie, Acadia, 12. Santarsieri D, Schwartz TL. Antidepressant efficacy and side-effect
Alkermes, Arbor, Eisai, Neos, Novo Nordisk, Otsuka, Sunovion, Supernus, burden: a quick guide for clinicians. Drugs Context. 2015;4:212290.
and Takeda, and has also received payment/honoraria from AbbVie, Arbor, 13. Kupfer DJ, Frank E, Phillips ML. Major depressive disorder: new
Eisai, Otsuka, and Takeda. W. Clay Jackson has received consulting fees and clinical, neurobiological, and treatment perspectives. Lancet.
payment/honoraria from AbbVie, Alkermes, Genentech, Otsuka, and 2012;379(9820):1045–1055.
Sunovion; has received honoraria from AbbVie, Alkermes, Genentech, 14. aan het Rot M, Mathew SJ, Charney DS. Neurobiological mechan­
Otsuka, and Sunovion for attending meetings and/or travel; and has partici­ isms in major depressive disorder. CMAJ. 2009;180(3):305–313.
pated on a data safety monitoring board or advisory board for AbbVie, 15. Yamada Y, Yoshikawa T, Naganuma F, et al. Chronic brain histamine
Alkermes, Genentech, Otsuka, and Sunovion. Denise Vanacore has received depletion in adult mice induced depression-like behaviours and
payment/honoraria from AbbVie. Richard Weisler has received grants or impaired sleep-wake cycle. Neuropharmacology. 2020;175:108179.
contracts from Allergan, Alkermes, Astellas, Axsome Therapeutics, 16. Katzman MA, Bilkey TS, Chokka PR, et al. Adult ADHD and comor­
Ironshore, Janssen, Lundbeck, Major League Baseball, Neos Therapeutics, bid disorders: clinical implications of a dimensional approach. BMC
Otsuka America Pharmaceuticals, Sirtsei Pharmaceuticals, Shire, Supernus Psychiatry. 2017;17(1):302.
Pharmaceuticals, Takeda, and Validus. The authors have no other relevant
17. FDA. In: Depression medicines [Internet]. September 2019. Silver
affiliations or financial involvement with any organization or entity with
Spring (MD): Food and Drug Administration. Available fromhttps://
a financial interest in or financial conflict with the subject matter or materials
www.fda.gov/media/132665/download
discussed in the manuscript apart from those disclosed.
18. Papakostas GI, Jackson WC, Rafeyan R, et al. Overcoming chal­
A reviewer on this manuscript has disclosed receiving manuscript or
lenges to treat inadequate response in major depressive disorder.
speaker’s fees from Astellas, Eisai, Eli Lilly, Elsevier Japan, Janssen
J Clin Psychiatry. 2020;81:3.
Pharmaceuticals, Kyowa Yakuhin, Lundbeck Japan, Meiji Seika Pharma,
19. Luo Y, Kataoka Y, Ostinelli EG, et al. National prescription patterns
Mitsubishi Tanabe Pharma, MSD, Nihon Medi-Physics, Novartis, Otsuka
of antidepressants in the treatment of adults with major depres­
Pharmaceutical, Shionogi, Shire, Sumitomo Pharma, Takeda Pharmaceutical,
sion in the US between 1996 and 2015: a population representative
Tsumura, Viatris, Wiley Japan, and Yoshitomi Yakuhin; and research grants
survey based analysis. Front Psychiatry. 2020;11:35.
from Eisai, Mochida Pharmaceutical, Meiji Seika Pharma, Shionogi and
20. Sansone RA, Sansone LA. SSRI-induced indifference. Psychiatry
Sumitomo Pharma. Peer reviewers on this manuscript have no other relevant
(Edgmont). 2010;7(10):14–18.
financial relationships or otherwise to disclose.
21. Rosenblat JD, Simon GE, Sachs GS, et al. Treatment effectiveness and
tolerability outcomes that are most important to individuals with
bipolar and unipolar depression. J Affect Disord. 2019;243:116–120.
References 22. Ma H, Cai M, Wang H. Emotional blunting in patients with major
depressive disorder: a brief non-systematic review of current
1. Villarroel MA, Terlizzi EP. Symptoms of depression among adults:
research. Front Psychiatry. 2021;12:792960.
united States. NCHS Data Brief. 2019;2020(379):1–8.
23. McIntyre RS, Rosenblat JD, Nemeroff CB, et al. Synthesizing the
2. Delphin-Rittmon ME. In: SAMHSA: key substance use and mental
evidence for ketamine and esketamine in treatment-resistant
health indicators in the United States: results from the 2020
depression: an international expert opinion on the available evi­
national survey on drug use and health [Internet]. Jul 2020.
dence and implementation. Am J Psychiatry. 2021;178(5):385–399.
Rockville (MD): Substance Abuse and Mental Health Services
Administration. Available fromhttps://www.samhsa.gov/data/sites/ 24. Iosifescu DV, Jones A, O’Gorman C, et al. Efficacy and safety of
default/files/reports/slides-2020-nsduh AXS-05 (dextromethorphan-bupropion) in patients with major
/2020NSDUHNationalSlides072522.pdf depressive disorder: a phase 3 randomized clinical trial (GEMINI).
3. Malhi GS, Bell E, Singh AB, et al. The 2020 Royal Australian and New J Clin Psychiatry. 2022;83(4):21m14345.
Zealand College of Psychiatrists clinical practice guidelines for 25. Kryst J, Kawalec P, Mitoraj AM, et al. Efficacy of single and repeated
mood disorders: major depression summary. Bipolar Disord. administration of ketamine in unipolar and bipolar depression: a
2020;22(8):788–804. meta-analysis of randomized clinical trials. Pharmacol Rep. 2020;72
4. Sakai C, Tsuji T, Nakai T, et al. Change in antidepressant use after (3):543–562.
initiation of ADHD medication in Japanese adults with comorbid 26. Ng J, Rosenblat JD, Lui LMW, et al. Efficacy of ketamine and
depression: a real-world database analysis. Neuropsychiatr Dis esketamine on functional outcomes in treatment-resistant depres­
Treat. 2021;17:3097–3108. sion: a systematic review. J Affect Disord. 2021;293:285–294.
5. Park LT, Zarate CA Jr. Depression in the primary care setting. N Engl 27. Richards JB, Papaioannou A, Adachi JD, et al. Effect of selective
J Med. 2019;380(6):559–568. serotonin reuptake inhibitors on the risk of fracture. Arch Intern
6. Ferenchick EK, Ramanuj P, Pincus HA. Depression in primary care: Med. 2007;167(2):188–194.
part 1—screening and diagnosis. BMJ. 2019;365:l794. 28. Kang S, Han M, Park CI, et al. Use of serotonin reuptake inhibitors
7. American Psychiatric Association. Diagnostic and statistical manual and risk of subsequent bone loss in a nationwide population-based
of mental disorders: DSM-5. 5th. Washington (DC): American cohort study. Sci Rep. 2021;11(1):13461.
Psychiatric Publishing; 2013. 29. Faquih AE, Memon RI, Hafeez H, et al. A review of novel antide­
8. Gelenberg AJ, Freeman MP, Markowitz JC, et al. In: Practice guide­ pressants: a guide for clinicians. Cureus. 2019;11(3):e4185.
line for the treatment of patients with major depressive disorder­ 30. De Bodinat C, Guardiola-Lemaitre B, Mocaer E, et al. Agomelatine,
[Internet] 3rd. Washington (DC): American Psychiatric Association; the first melatonergic antidepressant: discovery, characterization
2010. Available fromhttp://www.psychiatryonline.com/pracGuide/ and development. Nat Rev Drug Discov. 2010;9(8):628–642.
pracGuideTopic_7.aspx 31. Rosenbaum SB, Gupta V, Patel P, et al. Ketamine. Updated 2022
9. Ban TA. The role of serendipity in drug discovery. Dialogues Clin Nov 24. Available from: https://www.ncbi.nlm.nih.gov/books/
Neurosci. 2006;8(3):335–344. NBK470357/
464 C. B. MONTANO ET AL.

32. Salahudeen M, Wright CM, Peterson GM. Esketamine: new hope for 53. Staiger B. What is agomelatine and why isn’t it available in the
the treatment of treatment-resistant depression? A narrative United States? [Internet]. Updated 2019 Oct 28. Available from.
review. Ther Adv Drug Saf. 2020;11:1–23. https://walrus.com/articles/what-is-agomelatine-and-why-isn-t-it-
33. Cosci F, Chouinard G. The monoamine hypothesis of depression available-in-the-united-states
revisited: could it mechanistically novel antidepressant strategies? 54. Kennedy SH, Lam RW, McIntyre RS, et al. Canadian Network for Mood
In: Quevedo J, Carvalho AF, Zarate CA, eds. Neurobiology of and Anxiety Treatments (CANMAT) 2016 clinical guidelines for the
Depression: Road to Novel Therapeutics. London: Academic Press. management of adults with major depressive disorder: section 3.
2019. 63–73. Pharmacological treatments. Can J Psychiatry. 2016;61(9):540–560.
34. Sub Laban T, Saadabadi A. Monoamine oxidase inhibitors (MAOI). 55. Jefferson JW, Pradko JF, Muir KT. Bupropion for major depressive
Treasure Island (FL): StatPearls Publishing; 2022. disorder: pharmacokinetic and formulation considerations. Clin
35. Suchting R, Tirumalajaru V, Gareeb R, et al. Revisiting monoamine Ther. 2005;27(11):1685–1695.
oxidase inhibitors for the treatment of depressive disorders: 56. Patel K, Allen S, Haque MN, et al. Bupropion: a systematic review
a systematic review and network meta-analysis. J Affect Disord. and meta-analysis of effectiveness as an antidepressant. Ther Adv
2021;282:1153–1160. Psychopharmacol. 2016;6(2):99–144.
36. Bodkin JA, Dunlop BW. Moving on with monoamine oxidase 57. Sanchez C, Asin KE, Artigas F. Vortioxetine, a novel antidepressant
inhibitors. Focus (Am Psychiatr Publ). 2021;19(1):50–52. with multimodal activity: review of preclinical and clinical data.
37. Grady MM, Stahl SM. Practical guide for prescribing MAOIs: Pharmacol Ther. 2015;145:43–57.
debunking myths and removing barriers. CNS Spectr. 2012;17 58. Citrome L. Vilazodone, levomilnacipran and vortioxetine for major
(1):2–10. depressive disorder: the 15-min challenge to sort these agents out.
38. Chu A, Wadhwa R. Selective serotonin reuptake inhibitors. Treasure Int J Clin Pract. 2015;69(2):151–155.
Island (FL): StatPearls Publishing; 2022. 59. Wang SM, Han C, Lee SJ, et al. Vilazodone for the treatment of
39. Carvalho AF, Sharma MS, Brunoni AR, et al. The safety, tolerability depression: an update. Chonnam Med J. 2016;52(2):91–100.
and risks associated with the use of newer generation antidepres­ 60. Trintellix [package insert]. Deerfield. IL: Takeda Pharmaceuticals
sant drugs: a critical review of the literature. Psychother America, Inc; 2021.
Psychosom. 2016;85(5):270–288. 61. Montano CB, Jackson WC, Vanacore D, et al. Practical advice for
40. Warden SJ, Fuchs RK. Do selective serotonin reuptake inhibitors primary care clinicians on the safe and effective use of vortioxetine
(SSRIs) cause fractures? Curr Osteoporos Rep. 2016;14(5):211–218. for patients with major depressive disorder (MDD). Neuropsychiatr
41. FDA. In: FDA Drug Safety Communication: revised recommenda­ Dis Treat. 2022;18:867–879.
tions for Celexa (citalopram hydrobromide) related to a potential 62. Stahl SM. Dextromethorphan/bupropion: a novel oral NMDA
risk of abnormal heart rhythms with high doses [Internet]. updated (N-methyl-d-aspartate) receptor antagonist with multimodal
2012 Mar 28. Silver Spring (MD): Food and Drug Administration. activity. CNS Spectr. 2019;24(5):461–466.
Available fromhttps://www.fda.gov/drugs/drug-safety-and- 63. Messer MM, Haller IV. Minn Med.2018;November/December:36–40.
availability/fda-drug-safety-communication-revised- 64. Sanacora G, Frye MA, McDonald W, et al. A consensus statement on
recommendations-celexa-citalopram-hydrobromide-related the use of ketamine in the treatment of mood disorders. JAMA
42. Yuet WC, Derasari D, Sivoravong J, et al. Selective serotonin reup­ Psychiatry. 2017;74(4):399–405.
take inhibitor use and risk of gastrointestinal and intracranial 65. Auvelity [package insert]. New York NY: Axsome Therapeutics, Inc;
bleeding. J Am Osteopath Assoc. 2019;119(2):102–111. 2022.
43. Sansone RA, Sansone LA. Serotonin norepinephrine reuptake inhi­ 66. Khabir Y, Hashmi MR, Ali Asghar A. Rapid-acting oral drug
bitors: a pharmacological comparison. Innov Clin Neurosci. 2014;11 (Auvelity) for major depressive disorder. Ann Med Surg (Lond).
(3–4):37–42. 2022;82:104629.
44. Perahia DG, Pritchett YL, Kajdasz DK, et al. A randomized, 67. Ng CW, How CH, Ng YP. Depression in primary care: assessing
double-blind comparison of duloxetine and venlafaxine in the suicide risk. Singapore Med J. 2017;58(2):72–77.
treatment of patients with major depressive disorder. J Psychiatr 68. Seok Seo J, Rim Song H, Bin Lee H, et al. The Korean medication
Res. 2008;42(1):22–34. algorithm for depressive disorder: second revision. J Affect Disord.
45. Fava GA, Benasi G, Lucente M, et al. Withdrawal symptoms after 2014;167:312–321.
serotonin-noradrenaline reuptake inhibitor discontinuation: sys­ 69. Bond DJ, Hadjipavlou G, Lam RW, et al. The Canadian network for
tematic review. Psychother Psychosom. 2018;87(4):195–203. mood and anxiety treatments (CANMAT) task force recommenda­
46. Cosci F, Chouinard G. Acute and persistent withdrawal syndromes tions for the management of patients with mood disorders and
following discontinuation of psychotropic medications. Psychother comorbid attention-deficit/hyperactivity disorder. Ann Clin
Psychosom. 2020;89(5):283–306. Psychiatry. 2012;24(1):23–37.
47. Thase ME, Nierenberg AA, Vrijland P, et al. Remission with mirtaza­ 70. Cipriani A, Furukawa TA, Salanti G, et al. Comparative efficacy and
pine and selective serotonin reuptake inhibitors: a meta-analysis of acceptability of 21 antidepressant drugs for the acute treatment of
individual patient data from 15 controlled trials of acute phase adults with major depressive disorder: a systematic review and
treatment of major depression. Int Clin Psychopharmacol. 2010;25 network meta-analysis. Lancet. 2018;391(10128):1357–1366.
(4):189–198. 71. Del Casale A, Pomes LM, Bonanni L, et al. Pharmacogenomics-
48. Jilani TN, Gibbons JR, Faizy RM, et al. Mirtazapine. Treasure Island guided pharmacotherapy in patients with major depressive dis­
(FL): StatPearls Publishing; 2022. order or bipolar disorder affected by treatment-resistant depres­
49. Croom KF, Perry CM, Plosker GL. Mirtazapine: a review of its use in sive episodes: a long-term follow-up study. J Pers Med. 2022;12
major depression and other psychiatric disorders. CNS Drugs. (2):316.
2009;23(5):427–452. 72. Li Z, Ruan M, Chen J, et al. Major depressive disorder: advances in
50. Shin JJ, Saadabadi A. Trazodone. Treasure Island (FL): StatPearls neuroscience research and translational applications. Neurosci Bull.
Publishing; 2022. 2021;37(6):863–880.
51. Jaffer KY, Chang T, Vanle B, et al. Trazodone for insomnia: 73. Carvajal C. Poor response to treatment: beyond medication.
a systematic review. Innov Clin Neurosci. 2017;14(7–8):24–34. Dialogues Clin Neurosci. 2004;6(1):93–103.
52. Goracci A, Forgione RN, De Giorgi R, et al. Practical guidance for 74. Cartwright C, Gibson K, Read J, et al. Long-term antidepressant use:
prescribing trazodone extended-release in major depression. patient perspectives of benefits and adverse effects. Patient Prefer
Expert Opin Pharmacother. 2016;17(3):433–441. Adherence. 2016;10:1401–1407.
POSTGRADUATE MEDICINE 465

75. McIntyre RS. Using measurement strategies to identify and monitor ability in clinical depression—a systematic review. Psychiatry Res.
residual symptoms. J Clin Psychiatry. 2013;74(Suppl 2):14–18. 2014;219(1):25–50.
76. McIntyre RS, Patel MD, Masand PS, et al. The Rapid Mood Screener 96. Pan Z, Grovu RC, Cha DS, et al. Pharmacological treatment of
(RMS): a novel and pragmatic screener for bipolar I disorder. Curr cognitive symptoms in major depressive disorder. CNS Neurol
Med Res Opin. 2021;37(1):135–144. Disord Drug Targets. 2017;16(8):891–899.
77. Ratheesh A, Davey C, Hetrick S, et al. A systematic review and 97. Haro JM, Hammer-Helmich L, Saragoussi D, et al. Patient-reported
meta-analysis of prospective transition from major depression to depression severity and cognitive symptoms as determinants of
bipolar disorder. Acta Psychiatr Scand. 2017;135(4):273–284. functioning in patients with major depressive disorder: a secondary
78. Gitlin MJ. Antidepressants in bipolar depression: an enduring analysis of the 2-year prospective PERFORM study. Neuropsychiatr
controversy. Int J Bipolar Disord. 2018;6(1):25. Dis Treat. 2019;15:2313–2323.
79. McElroy SL, Kotwal R, Kaneria R, et al. Antidepressants and suicidal 98. Sumiyoshi T, Hoshino T, Mishiro I, et al. Prediction of residual
behavior in bipolar disorder. Bipolar Disord. 2006;8(5 Pt 2):596–617. cognitive disturbances by early response of depressive symptoms
80. Kishi T, Ikuta T, Matsuda Y, et al. Mood stabilizers and/or antipsy­ to antidepressant treatments in patients with major depressive
chotics for bipolar disorder in the maintenance phase: a systematic disorder. J Affect Disord. 2022;296:95–102.
review and network meta-analysis of randomized controlled trials. 99. Mattingly GW, Ren H, Christensen MC, et al. Effectiveness of vor­
Mol Psychiatry. 2021;26(8):4146–4157. tioxetine in patients with major depressive disorder in real-world
81. Haddad PM, Talbot PS, Anderson IM, et al. Managing inadequate clinical practice: results of the RELIEVE study. Front Psychiatry.
antidepressant response in depressive illness. Br Med Bull. 2015;115 2022;13:824831.
(1):183–201. 100. Fang H, Tu S, Sheng J, et al. Depression in sleep disturbance:
82. Chan HN, Mitchell PB, Loo CK, et al. Pharmacological treatment a review on a bidirectional relationship, mechanisms and
approaches to difficult-to-treat depression. Med J Aust. 2013;199 treatment. J Cell Mol Med. 2019;23(4):2324–2332.
(Suppl 6):S44–S47. 101. Wang YQ, Li R, Zhang MQ, et al. The neurobiological mechanisms
83. McIntosh D, Kutcher S, Binder C, et al. Adult ADHD and comorbid and treatments of REM sleep disturbances in depression. Curr
depression: a consensus-derived diagnostic algorithm for ADHD. Neuropharmacol. 2015;13(4):543–553.
Neuropsychiatr Dis Treat. 2009;5:137–150. 102. Alberti S, Chiesa A, Andrisano C, et al. Insomnia and somnolence
84. Sternat T, Katzman MA. Neurobiology of hedonic tone: the relation­ associated with second-generation antidepressants during the
ship between treatment-resistant depression, attention-deficit treatment of major depression: a meta-analysis. J Clin
hyperactivity disorder, and substance abuse. Neuropsychiatr Dis Psychopharmacol. 2015;35(3):296–303.
Treat. 2016;12:2149–2164. 103. Goodwin GM, Price J, De Bodinat C, et al. Emotional blunting with
85. Canadian ADHD Resource Alliance (CADDRA). Canadian ADHD antidepressant treatments: a survey among depressed patients.
practice guidelines. 4th ed. Toronto (ON): CADDRA; 2018. J Affect Disord. 2017;221:31–35.
86. Verbeeck W, Bekkering GE, Van den Noortgate W, et al. Bupropion
104. Read J, Cartwright C, Gibson K. Adverse emotional and interperso­
for attention deficit hyperactivity disorder (ADHD) in adults.
nal effects reported by 1829 New Zealanders while taking
Cochrane Database Syst Rev. 2017;10(10):CD009504.
antidepressants. Psychiatry Res. 2014;216(1):67–73.
87. Yu C, Garcia-Olivares J, Candler S, et al. New insights into the
105. Wongpakaran N, van Reekum R, Wongpakaran T, et al. Selective
mechanism of action of viloxazine: serotonin and norepinephrine
serotonin reuptake inhibitor use associates with apathy among
modulating properties. J Exp Pharmacol. 2020;12:285–300.
depressed elderly: a case-control study. Ann Gen Psychiatry. 2007;6:7.
88. Qelbree [package insert]. Rockville. MD: Supernus Pharmaceuticals
106. Christensen MC, Fagiolini A, Florea I, et al. Validation of the Oxford
Inc; 2022.
Depression Questionnaire: sensitivity to change, minimal clinically
89. Kinrys G, Gold AK, Pisano VD, et al. Tachyphylaxis in major depres­
important difference, and response threshold for the assessment of
sive disorder: a review of the current state of research. J Affect
emotional blunting. J Affect Disord. 2021;294:924–931.
Disord. 2019;245:488–497.
107. Corruble E, de Bodinat C, Belaidi C, et al. Efficacy of agomela­
90. MacQueen G, Santaguida P, Keshavarz H, et al. Systematic review of
tine and escitalopram on depression, subjective sleep and
clinical practice guidelines for failed antidepressant treatment
emotional experiences in patients with major depressive dis­
response in major depressive disorder, dysthymia, and subthres­
order: a 24-wk randomized, controlled, double-blind trial.
hold depression in adults. Can J Psychiatry. 2017;62(1):11–23.
Int J Neuropsychopharmacol. 2013;16(10):2219–2234.
91. Durgam S, Earley W, Guo H, et al. Efficacy and safety of adjunctive
cariprazine in inadequate responders to antidepressants: 108. Fagiolini A, Florea I, Loft H, et al. Effectiveness of vortioxetine on
a randomized, double-blind, placebo-controlled study in adult patients emotional blunting in patients with major depressive disorder with
with major depressive disorder. J Clin Psychiatry. 2016;77(3):371–378. inadequate response to SSRI/SNRI treatment. J Affect Disord.
92. Lam RW, Parikh SV, Michalak EE, et al. Canadian Network for Mood 2021;283:472–479.
and Anxiety Treatments (CANMAT) consensus recommendations 109. Goodwin GM. Revisiting treatment options for depressed patients
for functional outcomes in major depressive disorder. Ann Clin with generalised anxiety disorder. Adv Ther. 2021;38(Suppl 2):61–68.
Psychiatry. 2015;27(2):142–149. 110. Zareifopoulos N, Dylja I. Efficacy and tolerability of vilazodone for
93. Evans VC, Alamian G, McLeod J, et al. The effects of newer anti­ the acute treatment of generalized anxiety disorder: a
depressants on occupational impairment in major depressive dis­ meta-analysis. Asian J Psychiatr. 2017;26:115–122.
order: a systematic review and meta-analysis of randomized 111. Slee A, Nazareth I, Bondaronek P, et al. Pharmacological treatments
controlled trials. CNS Drugs. 2016;30(5):405–417. for generalised anxiety disorder: a systematic review and network
94. McIntyre RS, Cha DS, Soczynska JK, et al. Cognitive deficits and func­ meta-analysis. Lancet. 2019;393(10173):768–777.
tional outcomes in major depressive disorder: determinants, substrates, 112. Christensen MC, Schmidt S, Grande I. Effectiveness of vortioxetine
and treatment interventions. Depress Anxiety. 2013;30(6):515–527. in patients with major depressive disorder comorbid with general­
95. Baune BT, Renger L. Pharmacological and non-pharmacological ized anxiety disorder: results of the RECONNECT study.
interventions to improve cognitive dysfunction and functional J Psychopharmacol. 2022;36(5):566–577.

You might also like