6
DISEASE MANAGEMENT Consider augmentation or switching strategies when first-line antidepressants are unsuccessful in major depressive disorder When treating major depressive disorder that has not responded to an adequate trial of a first-line antidepressant, the current evidence most strongly supports switching to another first-line antidepressant or augmentation with the atypical antipsychotics aripiprazole or quetiapine. Lithium and liothyronine (triiodothyronine) may be used to augment tricyclic antidepressants, but further studies of their use to augment newer antidepressants are needed. If initial treatment does not succeed ... Major depressive disorder (MDD) is a common disorder that reduces health-related quality of life, results in poorer overall health, impairs the ability to work and is associated with considerable economic costs to society. [1,2] Although there have been numerous advances in the pharmacological treatment of MDD, initial treatment with first-line anti- depressants (e.g. selective serotonin reuptake inhibitors [SSRIs], serotonin and norepinephrine [noradrenlaine] reup- take inhibitors [SNRIs], bupropion and mirtazapine) does not provide an adequate clinical response in many patients. [1] After an initially unsuccessful treatment of MDD, it is vital that the next agent is chosen with care, as a good choice can improve outcomes and resolve illness, whereas a poorer choice may lead to persistent depression with a more chronic and morbid disease course. [3,4] This article provides a summary of a recent review by Connolly and Thase [1] of the current evidence regarding second-line treatment of adults with MDD who have not had an adequate clinical response to their first antidepressant medication treatment. y try, try again with another approach Many guidelines offer recommendations for the initial management of MDD; however, guidance as to which option is best for patients with treatment-resistant MDD is more limited. [1] After the failure of an initial anti- depressant trial that has been optimized in dose and dura- tion, current evidence-based clinical guidelines [5-8] recom- mend improving efficacy with the following main pharma- cological strategies: switching to another antidepressant; [5-8] augmentation or adjunctive therapy with another agent (which is not thought to be an antidepressant itself) to the antidepressant regimen. [5-8] Agents that are recom- mended to augment antidepressant therapy include atypical antipsychotics, [6,8] lithium [5-8] and liothyronine (triiodothyronine). [5-8] Although other pharmacological strategies, such as combination therapy with two antidepressants and aug- mentation with stimulants or buspirone, are commonly used in clinical practice, there is a lack of evidence to support their use. [1,5-8] Nonpharmacological therapies, such as the addition of psychotherapy to pharmacological treatment and the use of neurostimulation therapies (electroconvulsive therapy or transcranial magnetic stimulation) may have beneficial effects in treatment-resistant MDD; [1] however, a discussion of their use is beyond the scope of this review. Switch to a different antidepressant In patients who have not responded to an adequate trial of an antidepressant, the antidepressant agent used may be switched to another agent, [1,2,5-8] which produces a response in 18–60% of patients. [1] Strategies include changing to another class of newer-generation antidepressant (generally considered to be the favoured option), or changing to an- other agent in the same class or an older-generation anti- depressant. Unfortunately, the few high-quality studies that have in- vestigated this issue have generally not shown consistent and/or clinically significant differences between various switching strategies (table I). [1] Therefore, guidance on the class of agent that offers a unique clinical advantage after treatment failure with an SSRI or another first-line anti- depressant agent cannot yet be provided. Further studies are clearly needed to determine the best approach with regard to this important and common treatment step. [1] Augment with atypical antipsychotics Of the augmentation strategies, adjunctive treatment with atypical antipsychotics has been the most systematically and rigorously studied, with current data supporting their use in patients with treatment-resistant MDD. [1] In a meta-analysis of data from 16 randomized controlled trials (RCTs) of atypical antipsychotic versus placebo augmentation in 3480 patients with treatment-resistant MDD, the addition of an atypical antipsychotic was significantly (p < 0.0001) more 1172-0360/11/0012-0007/$19.95 ª 2011 Adis Data Information BV. All rights reserved. 7

Consider augmentation or switching strategies when first-line antidepressants are unsuccessful in major depressive disorder

Embed Size (px)

Citation preview

Page 1: Consider augmentation or switching strategies when first-line antidepressants are unsuccessful in major depressive disorder

DISEASE MANAGEMENT

Consider augmentation or switching strategies when first-lineantidepressants are unsuccessful in major depressive disorder

When treating major depressive disorder that has not

responded to an adequate trial of a first-line antidepressant,

the current evidence most strongly supports switching to

another first-line antidepressant or augmentation with the

atypical antipsychotics aripiprazole or quetiapine. Lithium

and liothyronine (triiodothyronine) may be used to augment

tricyclic antidepressants, but further studies of their use to

augment newer antidepressants are needed.

If initial treatment does not succeed ...

Major depressive disorder (MDD) is a common disorder

that reduces health-related quality of life, results in poorer

overall health, impairs the ability to work and is associated

with considerable economic costs to society.[1,2] Although

there have been numerous advances in the pharmacological

treatment of MDD, initial treatment with first-line anti-

depressants (e.g. selective serotonin reuptake inhibitors

[SSRIs], serotonin and norepinephrine [noradrenlaine] reup-

take inhibitors [SNRIs], bupropion and mirtazapine) does not

provide an adequate clinical response in many patients.[1]

After an initially unsuccessful treatment of MDD, it is

vital that the next agent is chosen with care, as a good choice

can improve outcomes and resolve illness, whereas a poorer

choice may lead to persistent depression with a more

chronic and morbid disease course.[3,4] This article provides

a summary of a recent review by Connolly and Thase[1] of

the current evidence regarding second-line treatment of

adults with MDD who have not had an adequate clinical

response to their first antidepressant medication treatment.

y try, try again with another approach

Many guidelines offer recommendations for the initial

management of MDD; however, guidance as to which

option is best for patients with treatment-resistant MDD

is more limited.[1] After the failure of an initial anti-

depressant trial that has been optimized in dose and dura-

tion, current evidence-based clinical guidelines[5-8] recom-

mend improving efficacy with the following main pharma-

cological strategies:� switching to another antidepressant;[5-8]

� augmentation or adjunctive therapy with another agent

(which is not thought to be an antidepressant itself) to the

antidepressant regimen.[5-8] Agents that are recom-

mended to augment antidepressant therapy include

atypical antipsychotics,[6,8] lithium[5-8] and liothyronine

(triiodothyronine).[5-8]

Although other pharmacological strategies, such as

combination therapy with two antidepressants and aug-

mentation with stimulants or buspirone, are commonly used

in clinical practice, there is a lack of evidence to support

their use.[1,5-8] Nonpharmacological therapies, such as the

addition of psychotherapy to pharmacological treatment and

the use of neurostimulation therapies (electroconvulsive

therapy or transcranial magnetic stimulation) may have

beneficial effects in treatment-resistant MDD;[1] however, a

discussion of their use is beyond the scope of this review.

Switch to a different antidepressant

In patients who have not responded to an adequate trial of

an antidepressant, the antidepressant agent used may be

switched to another agent,[1,2,5-8] which produces a response

in 18–60% of patients.[1] Strategies include changing to

another class of newer-generation antidepressant (generally

considered to be the favoured option), or changing to an-

other agent in the same class or an older-generation anti-

depressant.

Unfortunately, the few high-quality studies that have in-

vestigated this issue have generally not shown consistent

and/or clinically significant differences between various

switching strategies (table I).[1] Therefore, guidance on the

class of agent that offers a unique clinical advantage after

treatment failure with an SSRI or another first-line anti-

depressant agent cannot yet be provided. Further studies are

clearly needed to determine the best approach with regard to

this important and common treatment step.[1]

Augment with atypical antipsychotics

Of the augmentation strategies, adjunctive treatment with

atypical antipsychotics has been the most systematically and

rigorously studied, with current data supporting their use in

patients with treatment-resistant MDD.[1] In a meta-analysis

of data from 16 randomized controlled trials (RCTs) of

atypical antipsychotic versus placebo augmentation in 3480

patients with treatment-resistant MDD, the addition of an

atypical antipsychotic was significantly (p < 0.0001) more

1172-0360/11/0012-0007/$19.95 ª 2011 Adis Data Information BV. All rights reserved.

7

Page 2: Consider augmentation or switching strategies when first-line antidepressants are unsuccessful in major depressive disorder

effective than that of placebo with regard to rates of response

(odds ratio [OR] 1.69; 95% CI 1.46, 1.95) and remission

(OR 2.00; 95% CI 1.69, 2.37).[17] However, it remains to be

seen how the clinical risks and benefits of these agents

translate into real-world outcomes. The optimal duration of

augmentation therapy and the cost effectiveness of this

strategy relative to other strategies remain unknown.[1]

The rationale for the use of these agents to augment

treatment with SSRIs or SNRIs is largely based on the broad

receptor binding profiles of this medication class.[18] Atyp-

ical antipsychotics bind strongly to serotonin 5-HT2A re-

ceptors, aripiprazole and ziprasidone are also 5-HT1A partial

agonists, and ziprasidone and quetiapine (via its N-desalkyl

metabolite) inhibit norepinephrine reuptake. Different atyp-

ical antipsychotics may be beneficial in different patient

groups or may be more useful when used to augment treat-

ment with different antidepressants, but further study into

this area is required.[1]

Evidence supports the use of aripiprazole orquetiapine

The strongest available clinical data supports the use of

aripiprazole or quetiapine to augment antidepressant treat-

ment.[1] In 6-week RCTs in patients with a history of non-

response to adequate trials of antidepressant monotherapy,

aripiprazole[19-21] and quetiapine[23,24] (particularly at a do-

sage of 300 mg/day) appeared to be effective augmentation

strategies (table II). It is unclear if augmentation with que-

tiapine is of use in patients who are resistant to treatment

with more than one prior course of antidepressant therapy.[1]

Table I. Summary of the evidence for switching antidepressant medications in patients (pts) with majordepressive disorder (MDD) who do not respond to first-line antidepressant therapy, as reviewed byConnolly and Thase[1]

Switching to venlafaxine

In four RCTs in SSRI nonresponders,[3,9-11] a significant benefit with regard to response[11] and/or remission[9,11] was shown in pts switched to

venlafaxine vs a second SSRI in two trials

Overall, venlafaxine appears to be an effective and useful agent after the failure of an SSRI, but whether it is markedly more effective than other

options seems doubtful

Switching to a newer SNRI

Studies are needed to determine the role of newer SNRIs in treatment-resistant MDD, as no available RCTs have investigated switching to newer

SNRIs (e.g. duloxetine, desvenlafaxine and milnacipran) in treatment-resistant pts

Switching to bupropion

Although switching from an SSRI to bupropion is a commonly used strategy in the US, well designed trials of this strategy are lacking

A STAR*D study did not support any preference for switching to bupropion over either SSRIs or venlafaxine in pts with SSRI-resistant MDD[3]

Switching to mirtazapine

Although more studies are needed, the limited available evidence suggests that this agent may hold promise for effective and perhaps more rapid

remission after an SSRI failure, but does not indicate that it is more or less effective than other second-line options

In a large RCT in SSRI nonresponders,[12] remission rates in pts switched to mirtazapine were not significantly different than that in pts switched

to a second SSRI (sertraline) [38% vs 28%]; however, mirtazapine was associated with significantly faster times to response and remission

In the ARGOS comparison of mirtazapine with a second SSRI or venlafaxine as a second-line agent after an SSRI failure,[11] response and

remission rates with mirtazapine were numerically similar to those for SSRIs and lower than those with venlafaxine (statistical analysis not

performed)

In the STAR*D study in pts who had not responded to two consecutive antidepressants,[13] remission rates were similar when switched to

mirtazapine or nortriptyline (12.3% vs 19.8%)

Switching to a tricyclic or related antidepressant

Very little data support the use of a tricyclic or related antidepressant instead of another second-line treatment strategy

In an RCT in fluoxetine nonresponders, no significant difference was seen between the addition of mianserin (a tetracyclic antidepressant)

to ongoing fluoxetine treatment vs fluoxetine alone[14]

In pts who had failed to respond to either imipramine or sertraline, treatment with the opposite medication resulted in significantly improved

remission and response rates; although these results are of little value in indicating a preferred strategy, they do suggest that switching

antidepressants in general can improve outcomes, and that imipramine may be a promising second-line treatment after SSRI failure[15]

Switching to an MAOI

Because of their dietary restrictions and potentially dangerous medication interactions, MAOIs are typically reserved for more severely refractory

MDD

No RCTs have studied the use of MAOIs vs other available options for the treatment of MDD resistant to SSRIs or other first-line antidepressants

In a STAR*D study in pts resistant to three antidepressants, no clear advantage in remission rates was seen in pts receiving tranylcypromine over

those receiving mirtazapine plus venlafaxine (6.9% and 13.7%), and the MAOI was tolerated less well[16]

MAOI = monoamine oxidase inhibitor; RCT = randomized, controlled trial; SNRI= serotonin and norepinephrine (noradrenaline) reuptake

inhibitors; SSRI = selective serotonin reuptake inhibitors.

8

Drugs Ther Perspect 2011; Vol. 27, No. 12

Page 3: Consider augmentation or switching strategies when first-line antidepressants are unsuccessful in major depressive disorder

More data needed for other agents

In 4-week RCTs,[25,26] the addition of risperidone was

beneficial in improving outcomes in patients with treatment-

resistant MDD, but there was no significant advantage with

risperidone over placebo with regard to the proportion of

patients remaining relapse-free at the end of a 24-week

trial[27] (table II). The lack of sustained benefit in this

trial[27] casts doubt on the utility of this adjunctive strategy;

risperidone is not currently recommended as a first-line

augmentation option.[1,2]

To date, olanzapine has only been studied in combination

with fluoxetine.[1] Patients who did not respond to two trials

of SSRIs, including fluoxetine as the second agent, bene-

fited from the addition of olanzapine in an 8-week RCT[22]

(table II). Whether olanzapine/fluoxetine offers comparable

benefits in patients who do not respond to other treatment

sequences has not been evaluated.[1]

No large RCTs have evaluated the use of ziprasidone or

other recently approved antipsychotics (e.g. paliperidone,

iloperidone, asenapine) to augment antidepressant medica-

tions in patients with treatment-resistant MDD.[1,2] Although

ziprasidone has a favourable pharmacodynamic profile and

perhaps the best tolerability profile for minimizing weight

gain and related metabolic effects among the atypical anti-

psychotics, there is currently insufficient empirical support

to recommend the addition of ziprasidone to antidepressant

therapy.[1,2]

Must weigh risks versus benefits

Augmentation with an atypical antipsychotic should be

initiated only after careful appraisal of its potential benefits

and risks of adverse effects.[1,2,28] These agents are asso-

ciated with increased risks of metabolic adverse effects and

complications, including weight gain, lipid abnormalities

and impaired glucose tolerance, as well as tardive dyskinesia

and acute extrapyramidal syndromes.[28] In the meta-analysis

of placebo-controlled trials, atypical antipsychotics were

associated with an increased risk of discontinuation due to

Table II. Summary of the key efficacy results of augmentation with atypical antipsychotics in randomizedcontrolled trials in patients (pts) with major depressive disorder resistant to treatment with first-lineantidepressants. Pts continued to receive treatment with a first-line antidepressant with the addition of anatypical antipsychotic or placebo. Only results from trial arms relevant to augmentation therapy are reportedAugmentation regimensa (no. of pts) Durationb Results (antipsychotic vs comparator)

(wk) response ratec (% of pts) NNTd

Aripiprazole

Aripiprazole (182) vs placebo (176)[19] 6 33.7* vs 23.8 10

Aripiprazole (191) vs placebo (190)[20] 6 32.4*** vs 17.4 6.7

Aripiprazole (177) vs placebo (172)[21] 6 46.3*** vs 26.7 5

Olanzapine

Olanzapine (198) vs control (203)[22]e 8 40.4** vs 29.6 9.3

Quetiapine

Quetiapine 150 mg (166) or quetiapine 300 mg (166) vs placebo (160)[23] 6 55.4 and 57.8* vs 46.3 8.7f

Quetiapine 150 mg (143) or quetiapine 300 mg (146) vs placebo (143)[24] 6 51.7 and 58.9* vs 46.2 7.8f

Risperidone

Risperidone (106) vs placebo (112)[25] 4 46.2** vs 29.5g 8.3

Risperidone (64) vs placebo (30)[26] 4 54.7* vs 33.3 4.6

Risperidone (122) vs placebo (119)[27] 24 46.7 vs 45.4h No significant

advantage

a Unless otherwise indicated, dosages were titrated based on response and tolerability.

b Length of double-blind augmentation phase.

c Defined as 50% reduction in Montgomery-Asberg depression rating scale score, unless otherwise indicated.

d As reported in Connolly and Thase.[1]

e Preplanned analysis of two identically designed studies, in which previous nonresponders received fluoxetine for 8 wk; pts who did not respond

to fluoxetine were randomized to 8 additional wk of olanzapine plus fluoxetine or fluoxetine alone.

f Quetiapine 300 mg vs placebo. No significant advantage for quetiapine 150 mg vs placebo.

g Response defined as 50% reduction in Hamilton Depression Rating Scale score.

h Response defined as remaining free of depressive relapse at 24 wk.

NNT = number needed to treat for one clinical response; * p < 0.05, ** p < 0.01, *** p < 0.001 vs comparator.

9

1172-0360/11/0012-0009/$19.95 ª 2011 Adis Data Information BV. All rights reserved.

Page 4: Consider augmentation or switching strategies when first-line antidepressants are unsuccessful in major depressive disorder

adverse events relative to placebo (OR 3.91; 95% CI 2.68,

5.72).[17] As the risk of various adverse effects vary be-

tween these agents, treatment should be tailored to the in-

dividual.[28] For example, of the two recommended first-line

augmentation options, the risk of metabolic adverse effects

appears to be greater with quetiapine than with aripiprazole.

Longer-term studies are needed to fully evaluate the benefit-

risk profiles of these agents when used as augmentation

strategies in treatment-resistant MDD.[1]

No response to a tricyclicantidepressant? Try adding lithium ...

Lithium, which has shown efficacy as an augment to

antidepressant therapy, is postulated to act by increasing

serotonergic neurotransmission.[1] In a meta-analysis of ten

RCTs of lithium augmentation of various antidepressants

(including tricyclic antidepressants [TCAs], SSRIs and other

antidepressants) in 269 patients with depressive disorder, the

overall OR for response with lithium relative to placebo

was 3.1 (95% CI 1.8, 5.4), which corresponds to a number

needed to treat (NNT) to achieve one clinical response of

5.[29] The mean response rate was significantly higher with

lithium augmentation than with placebo (41.2% vs 14.4% of

patients; p < 0.001).The evidence for the efficacy of lithium augmentation is

strongest for patients whose MDD has been inadequately

treated by an initial trial of a TCA.[1] However, there are

little data for lithium augmentation of SSRIs or other newer

antidepressants. Lithium, which has comparatively weaker

effects on serotonergic neurotransmission, may have little po-

tential to benefit patients who have not responded adequately

to antidepressants that affect serotonergic neurotransmission,

as responses to enhancing serotonergic neurotransmission

would likely have already been seen.[1] In an open-label,

randomized STAR*D (Sequenced Treatment Alternatives to

Relieve Depression) trial in patients who did not respond to

two sequential trials with newer antidepressants, 15.9% of 69

patients who received lithium augmentation achieved the pri-

mary endpoint of remission.[30]

The relevance of lithium augmentation in contemporary

clinical practice is less clear than might be assumed from the

recommendations found in current guidelines.[5-8] The effi-

cacy of adding lithium to TCAs is well established; how-

ever, the use of TCAs is typically reserved for patients with

more advanced levels of treatment resistance. In contrast,

lithium should be used to augment SSRIs and other newer

antidepressants with caution; however, these agents are the

most commonly recommended first-line antidepressants.[1]

Lithium is associated with a relatively low therapeutic index

and its use is associated with long-term risks of thyroid and

renal compromise.[1]

y or liothyronine

For augmentation of antidepressant therapy, the preferred

form of thyroid hormone is liothyronine.[31] It is recom-

mended instead of levothyroxine, which is used to treat

hypothyroidism, because liothyronine is thought to have

activity within the CNS and a rapid onset of action.[31]

Liothyronine augmentation has shown efficacy in pa-

tients who have not responded to TCAs.[1] In a meta-anal-

ysis of eight controlled trials in 292 TCA nonresponders,

patients who received liothyronine augmentation were twice

as likely to respond than those who received controls

(relative response 2.09; 95% CI 1.31, 3.32; p = 0.002). This

corresponds to an absolute increase in response rate of

23.2% (95% CI 4.5, 41.9; p = 0.02), and an NNT to achieve

one clinical response of 4.3.[32]

Questions remain as to whether the findings with TCAs

can be generalized to treatment with newer antidepressants.

In the STAR*D trial, 24.7% of 73 patients receiving lio-

thyronine augmentation of newer antidepressants achieved

remission (no significant difference relative to lithium aug-

mentation).[30] Moreover, an RCT in 36 patients nonresponsive

to SSRI therapy failed to find that liothyronine augmentation

had a significant benefit over that with placebo.[33]

Although liothyronine may appear to be a better option for

use in combination with SSRIs and other newer antidepressants

than lithium because of its generally better tolerability profile,

the remission rate observed in STAR*D was not significantly

better than with lithium, prolonged use of adjunctive thyroid

hormone was associated with adverse effects such as osteope-

nia, and the only placebo-controlled study of augmentation of

newer antidepressants with liothyronine did not shown a sig-

nificant benefit.[1] Further RCTs, therefore, are needed before

liothyronine can be recommended for euthyroid patients who

have not responded to first-line antidepressant treatment.

More evidence needed for augmentationwith psychostimulants

The use of traditional psychostimulants would seem to

be a rational strategy to improve depressive symptoms, as

these agents promote wakefulness, reduce fatigue and im-

prove mood by increasing levels of synaptic dopamine,

norepinephrine and serotonin to varying degrees.[1] Indeed,

methylphenidate and amfetamines are prescribed by some

psychiatrists to treat depressive symptoms, based on clinical

experience, case series and open-label trials. However, only

a few well designed studies of stimulant augmentation of

10

Drugs Ther Perspect 2011; Vol. 27, No. 12

Page 5: Consider augmentation or switching strategies when first-line antidepressants are unsuccessful in major depressive disorder

antidepressants have been published and a systematic review

failed to establish the efficacy of this approach.[34]

Modafinil, a wakefulness-promoting agent, is thought to

act primarily on dopamine and norepinephrine neuro-

transmission.[1] Pooled data from the two RCTs in patients

with partial benefit with SSRIs indicated that modafinil

augmentation modestly improved symptoms of depression,

sleepiness and fatigue relative to placebo.[35] Of note, as

these studies were in ‘enriched’ samples (i.e. all patients had

fatigue and sleepiness at baseline),[35] evidence is lacking

that modafinil augmentation would be of benefit in anti-

depressant nonresponders without fatigue or sleepiness.[1]

The potential for tolerance of therapeutic effects and abuse,

as well as the current lack of strong efficacy data, should be

taken into account when the off-label use of psychostimu-

lants or modafinil is being considered to augment anti-

depressant therapy.[1]

Lack of benefits with adjunctivebuspirone ...

Buspirone, an anxiolytic, is a partial agonist at 5-HT1A

receptors.[1] A potential role for buspirone augmentation in

treatment-resistant MDD was suggested by its effects on

serotonin neurotransmission, which may enhance the effects

of SSRIs and SNRIs. Although a number of open-label trials

suggested that buspirone augmentation was beneficial, there

is no convincing evidence from RCTs that buspirone has a

place in the treatment of patients with MDD, regardless of

whether or not they have high levels of baseline anxiety.[1]

y or pindolol in treatment-resistant disease

Pindolol, which was originally developed as a b-adrenergicreceptor antagonist, is a relatively potent 5-HT1A receptor

antagonist.[1] Interestingly, evidence from recent meta-anal-

yses[36,37] suggests that pindolol may accelerate and enhance

response to antidepressant therapy during the first 2–4 weeks

of therapy (particularly in patients with non-resistant MDD,

who are SSRI-naive or who are at the beginning of their

illness[36]). However, it appears to have little or no signif-

icant effect on longer-term (6 week) treatment outcomes.[36,37]

High-quality data suggesting that augmentation with this

agent can improve outcomes in patients with treatment-

resistant MDD are currently lacking.[1]

Cannot recommend combiningantidepressants

Although treatment with a combination of two different

antidepressant medications is frequent in patients with re-

sistant MDD in clinical practice,[1] current guidelines do not

recommend this strategy.[5-8]

High-quality evidence to support adding another anti-

depressant agent to the treatment of patients who are not re-

sponding to antidepressant monotherapy is lacking.[1] A recent

single-blind RCT in 665 patients with chronic or recurrent

MDD concluded that combination treatment (sustained-

release bupropion plus escitalopram, or extended-release

venlafaxine plus mirtazapine) did not offer clinical advantages

over escitalopram monotherapy, with the extended-release

venlafaxine plus mirtazapine combination possibly increasing

the risk of adverse events.[38] No significant differences in

remission and response rates and most secondary outcomes

were shown between treatment groups during the acute

(12 week) or long-term (7 month) treatment phases.[38]

Small RCTs suggest that mirtazapine[39] and mianserin,[14]

which have similar receptor profiles, may prove useful when

added to treatment with other first-line antidepressants in

patients who had not responded to an adequate trial of an

antidepressant. Further study on the use of antidepressant

combinations in patients with treatment-resistant MDD are

needed before this strategy can be recommended.[1]

Disclosure

This review was adapted from Drugs 2011; 71 (1): 43-64[1] by Adis

editors and medical writers. The preparation of these articles was not

supported by any external funding.

References1. Connolly KR, Thase ME. If at first you don’t succeed: a review of the

evidence for antidepressant augmentation, combination and switching

strategies. Drugs 2011; 71 (1): 43-64

2. Philip NS, Carpenter LL, Tyrka AR, et al. Pharmacologic approaches

to treatment resistant depression: a re-examination for the modern era.

Expert Opin Pharmacother 2010 Apr; 11 (5): 709-22

3. Rush AJ, Trivedi MH, Wisniewski SR, et al. Bupropion-SR, sertra-

line, or venlafaxine-XR after failure of SSRIs for depression. N Engl

J Med 2006; 354 (12): 1231-42

4. Tranter R, O’Donovan C, Chandarana P, et al. Prevalence and out-

come of partial remission in depression. J Psychiatry Neurosci 2002;

27 (4): 241-7

5. American Psychiatric Association practice guideline for treatment of

patients with major depressive disorder, third edition. Arlington (VA);

American Psychiatric Association, 2010 Oct

6. Institute for Clinical Systems Improvement (ICSI) healthcare guide-

line: major depression in adults in primary care, thirteenth edition.

Bloomington (MN); ICSI, 2010 May

7. Malhi GS, Adams D, Porter R, et al. Clinical practice recommenda-

tions for depression. Acta Psychiatr Scand Suppl 2009; 119 (439): 8-26

8. Lam RW, Kennedy SH, Grigoriadis S, et al. Canadian Network for

Mood and Anxiety Treatments (CANMAT) clinical guidelines for the

management of major depressive disorder in adults. III: pharma-

cotherapy. J Affect Disord 2009; 117 Suppl. 1: S26-43

11

1172-0360/11/0012-0011/$19.95 ª 2011 Adis Data Information BV. All rights reserved.

Page 6: Consider augmentation or switching strategies when first-line antidepressants are unsuccessful in major depressive disorder

9. Poirier MF, Boyer P. Venlafaxine and paroxetine in treatment-resistant

depression: double-blind, randomised comparison. Br J Psychiatry

1999 Jul; 175: 12-6

10. Lenox-Smith AJ, Jiang Q. Venlafaxine extended release versus citalo-

pram in patients with depression unresponsive to a selective serotonin

reuptake inhibitor. Int Clin Psychopharmacol 2008; 23 (3): 113-9

11. Baldomero EB, Ubago JG, Cercos CL, et al. Venlafaxine extended

release versus conventional antidepressants in the remission of de-

pressive disorders after previous antidepressant failure: ARGOS

study. Depress Anxiety 2005; 22 (2): 68-76

12. Thase ME, Kremer C, Rodrigues H. Mirtazapine versus sertraline

after SSRI non-response [abstract]. The Annual Meeting of the New

Clinical Drug Evaluation Unit (NCDEU) of the National Institute of

Mental Health; 2001 May 28-31; Phoenix (AZ)

13. Fava M, Rush AJ, Wisniewski SR, et al. A comparison of mirtazapine

and nortriptyline following two consecutive failed medication treat-

ments for depressed outpatients: a STAR*D report. Am J Psychiatry

2006; 163 (7): 1161-72

14. Ferreri M, Lavergne F, Berlin I, et al. Benefits from mianserin aug-

mentation of fluoxetine in patients with major depression non-re-

sponders to fluoxetine alone. Acta Psychiatr Scand 2001; 103 (1): 66-72

15. Thase ME, Rush AJ, Howland RH, et al. Double-blind switch study of

imipramine or sertraline treatment of antidepressant-resistant chronic

depression. Arch Gen Psychiatry 2002; 59 (3): 233-9

16. McGrath PJ, Stewart JW, Fava M, et al. Tranylcypromine versus

venlafaxine plus mirtazapine following three failed antidepressant

medication trials for depression: a STAR*D report. Am J Psychiatry

2006 Sep; 163 (9): 1531-41

17. Nelson JC, Papakostas GI. Atypical antipsychotic augmentation in

major depressive disorder: a meta-analysis of placebo-controlled

randomized trials. Am J Psychiatry 2009 Sep; 166 (9): 980-91

18. Papakostas GI, Shelton RC. Use of atypical antipsychotics for treat-

ment-resistant major depressive disorder. Curr Psychiatry Rep 2008;

10 (6): 481-6

19. Berman RM, Marcus RN, Swanink R, et al. The efficacy and safety of

aripiprazole as adjunctive therapy in major depressive disorder: a

multicenter, randomized, double-blind, placebo-controlled study. J

Clin Psychiatry 2007; 68 (6): 843-53

20. Marcus RN, McQuade RD, Carson WH, et al. The efficacy and safety

of aripiprazole as adjunctive therapy in major depressive disorder: a

second multicenter, randomized, double-blind, placebo-controlled

study. J Clin Psychopharmacol 2008; 28 (2): 156-65

21. Berman RM, Fava M, Thase ME, et al. Aripiprazole augmentation in

major depressive disorder: a double-blind, placebo-controlled study in

patients with inadequate response to antidepressants. CNS Spectr

2009; 14 (4): 197-206

22. Thase ME, Corya SA, Osuntokun O, et al. A randomized, double-

blind comparison of olanzapine/fluoxetine combination, olanzapine,

and fluoxetine in treatment-resistant major depressive disorder. J Clin

Psychiatry 2007; 68 (2): 224-36

23. Bauer M, Pretorius HW, Constant EL, et al. Extended-release que-

tiapine as adjunct to an antidepressant in patients with major de-

pressive disorder: results of a randomized, placebo-controlled, double-

blind study. J Clin Psychiatry 2009; 70 (4): 540-9

24. El-Khalili N, Joyce M, Atkinson S, et al. Extended-release quetiapine

fumarate (quetiapine XR) as adjunctive therapy in major depressive

disorder (MDD) in patients with an inadequate response to ongoing

antidepressant treatment: a multicentre, randomized, double-blind,

placebo-controlled study. Int J Neuropsychopharmacol 2010; 13 (7):

917-32

25. Mahmoud RA, Pandina GJ, Turkoz I, et al. Risperidone for treatment-

refractory major depressive disorder: a randomized trial. Ann Intern

Med 2007; 147 (9): 593-602

26. Keitner GI, Garlow SJ, Ryan CE, et al. A randomized, placebo-con-

trolled trial of risperidone augmentation for patients with difficult-to-

treat unipolar, non-psychotic major depression. J Psychiatr Res 2009;

43 (3): 205-14

27. Rapaport MH, Gharabawi GM, Canuso CM, et al. Effects of risper-

idone augmentation in patients with treatment-resistant depression:

results of open-label treatment followed by double-blind continuation.

Neuropsychopharmacology 2006; 31 (11): 2505-13

28. Goodwin G, Fleischhacker W, Arango C, et al. Advantages and dis-

advantages of combination treatment with antipsychotics ECNP

consensus meeting, March 2008, Nice. Eur Neuropsychopharmacol

2009; 19 (7): 520-32

29. Crossley NA, Bauer M. Acceleration and augmentation of anti-

depressants with lithium for depressive disorders: two meta-analyses

of randomized, placebo-controlled trials. J Clin Psychiatry 2007; 68

(6): 935-40

30. Nierenberg AA, Fava M, Trivedi MH, et al. A comparison of lithium

and T3 augmentation following two failed medication treatments

for depression: a STAR*D report. Am J Psychiatry 2006; 163 (9):

1519-30

31. Altshuler LL, Bauer M, Frye MA, et al. Does thyroid supplementation

accelerate tricyclic antidepressant response? A review and meta-anal-

ysis of the literature. Am J Psychiatry 2001; 158 (10): 1617-22

32. Aronson R, Offman HJ, Joffe RT, et al. Triiodothyronine augmenta-

tion in the treatment of refractory depression: a meta-analysis. Arch

Gen Psychiatry 1996; 53 (9): 842-8

33. Joffe RT, Sokolov ST, Levitt AJ. Lithium and triiodothyronine

augmentation of antidepressants. Can J Psychiatry 2006; 51 (12):

791-3

34. Candy M, Jones L, Williams R, et al. Psychostimulants for depression.

Cochrane Database Syst Rev 2008; (2): CD006722

35. Fava M, Thase ME, DeBattista C, et al. Modafinil augmentation of

selective serotonin reuptake inhibitor therapy in MDD partial re-

sponders with persistent fatigue and sleepiness. Ann Clin Psychiatry

2007; 19 (3): 153-9

36. Portella MJ, de Diego-Adelino J, Ballesteros J, et al. Can we really

accelerate and enhance the selective serotonin reuptake inhibitor anti-

depressant effect? A randomized clinical trial and a meta-analysis of

pindolol in nonresistant depression. J Clin Psychiatry 2011; 72 (7): 962-9

37. Whale R, Terao T, Cowen P, et al. Pindolol augmentation of serotonin

reuptake inhibitors for the treatment of depressive disorder: a sys-

tematic review. J Psychopharmacol 2010 Apr; 24 (4): 513-20

38. Rush AJ, Trivedi MH, Stewart JW, et al. Combining medications to

enhance depression outcomes (CO-MED): acute and long-term out-

comes of a single-blind randomized study. Am J Psychiatry 2011;

168 (7): 689-701

39. Carpenter LL, Yasmin S, Price LH. A double-blind, placebo-con-

trolled study of antidepressant augmentation with mirtazapine. Biol

Psychiatry 2002; 51 (2): 183-8

12

Drugs Ther Perspect 2011; Vol. 27, No. 12