Open access peer-reviewed chapter

Resistant Depression

Written By

Jose Alfonso Ontiveros

Submitted: 31 August 2018 Reviewed: 15 November 2018 Published: 07 June 2019

DOI: 10.5772/intechopen.82568

From the Edited Volume

Antidepressants - Preclinical, Clinical and Translational Aspects

Edited by Olivier Berend

Chapter metrics overview

1,145 Chapter Downloads

View Full Metrics

Abstract

The term resistant points out a clinical phenomenon in which there is a lack of response to one or more therapeutic interventions. Resistance to major depressive disorder treatment causes distress to patients and their relatives, and increases the number of hospital admissions, outpatient consultations, use of psychoactive drugs, and treatment costs. Despite its serious medical and psychosocial medical implications, the definition of treatment resistant depression continues to be ambiguous and controversial. The lack of an agreement on definition, as well as the research on the subject being difficult, limits the practical knowledge on the best treatment options for groups of treatment resistant depression (TRD) patients. We review the concept and definitions of treatment resistant depression as well as the medical literature on different treatment methods studied and comparative studies. Finally, some relevant neurobiological data are reviewed.

Keywords

  • treatment
  • major depressive disorder
  • resistant depression
  • antidepressants
  • review

1. Introduction

The term “resistant” is widely employed in medical practice to point out a clinical phenomenon in which there is a lack of response to one or more therapeutic interventions. The presence of treatment resistance implies a specific series of clinical interventions, typically multidisciplinary, and focuses on solving or minimizing a medical problem. Resistance to major depressive disorder treatment, but also to other depressive disorders, such as dysthymia and bipolar depression, causes distress to patients and their relatives, and increases the number of hospital admissions, outpatient consultations, the use of psychoactive drugs, and treatment costs up to six times [1]. The definition of treatment resistant depression (TRD) continues to be ambiguous and controversial despite its serious medical and psychosocial implications. In medical literature, we can find more than 10 different definitions [2]. Many authors have published staging systems with their own definitions, descriptions, and characteristics on TRD [3, 4, 5, 6, 7]. The most accepted definition is a lack of response to two different pharmacological classes of antidepressants [8]. However, this definition may seem simplistic today as published treatment results on TRD patients emerge. The lack of an agreed TRD definition as well as the difficulties to do research on the subject limit our practical knowledge on the best treatment options for groups of TRD patients.

We review the concept and definitions of TRD. We also review the medical literature on different treatment methods studied as well as comparative studies. Finally, we review some relevant emerging neurobiological data.

Advertisement

2. Treatment resistance depression (TRD)

2.1 The concept of resistant depression

Although this phenomenon had already been described, many authors have introduced the concept of TRD since 1974 [9, 10]. This concept arises at a time when there were only tricyclic, tetracyclic, and monoamine-oxidase inhibitors (MAOIs) antidepressants available. In spite of its importance, the definition of treatment resistance regarding major depression continues to be a wide and inconsistent notion. A review of the literature identifies a range of definitions for TRD that go from non-response to a single antidepressant (for 4 or more weeks) to lack of response to different classes of antidepressants and electroconvulsive therapy (ECT) [2]. Treatment should be appropriate in dose and duration [2, 11, 12, 13], and patients must have full compliance to it [13] to consider a patient as resistant to treatment. There is no consensus on the number of treatments and when these are indicators of resistance.

A dichotomic denomination has been proposed for resistant depression, viz. an absolute and a relative. Absolute resistance is an inappropriate anti-depressive response toward a treatment given for an appropriate period of time at the maximum non-toxic dose. On the other hand, relative resistance to treatment is defined when this is given at a suboptimal dose or duration [5, 10, 14]. The terms “chronic,” “refractory,” and “difficult-to-treat depression” have been employed as synonyms in the absence of a nonspecific number of clinical trials for one or more antidepressants. Treatment refractory depression refers to major depression that does not respond to multiple sequential treatments. There is no clear difference between treatment resistant depression and refractory depression [5, 8, 11]. Chronicity, however, refers to a pathological clinical phenomenon that lasts for 2 or more years. There is no consensus on depressive symptom severity to consider it as resistant. It has been suggested that a score of 16 or more on the Hamilton depression 17-item scale (HAM-D) is enough to confirm the diagnosis. However, patients with persistent mild or moderate depressive symptoms may have a worse prognosis than those in remission [15]. The definition of Berlim and Turecki [2], which considers TRD as an episode of major depression that has not improved after two proper attempts with different classes of antidepressants, prevails today. The European Medicine Agency (EMA), on a TRD definition review, considers it as a clinically relevant major depression that has not benefited from at least two appropriate attempts of treatment with at least two antidepressants with a different action mechanism [16]. The definition which considers TRD as an episode of major depression that has not improved after two proper attempts with different classes of antidepressants [2] seems to be backed up by the STAR*D study, which shows that improvement chances diminish after the second treatment failure [17, 18]. Treatment resistance to pharmacologic treatment seems to move on a continuum that ranges from total response to total resistance to therapeutic intervention and not as an all-or-nothing phenomenon [5, 19, 20]. However, no definition has been investigated regarding validity and predictability [5, 21]. Inconsistencies on the definition not only give rise to difficulties at estimating its prevalence [17, 22] but can also delay research of the most efficient treatment schemes.

2.2 Prevalence of resistant depression

In spite of pharmacological treatment advances in major depression, the final objective of achieving a sustained improvement continues to be insufficient [23]. It is estimated that about only 30–40% of patients achieve remission after the first attempt of treatment with antidepressants. In 3671 ambulatory patients treated with escitalopram, only 37% achieved remission [18]. Even after an appropriate sequence of treatments, 10–20% of the patients with major depression continue with significant symptoms for 2 years or more [24, 25]. The STAR*D study (sequenced treatment alternatives to relieve depression study) showed that accumulated remission after four treatment trials with antidepressants for 14 months was 67% [26]. Patients with chronic depression seem to have less opportunity for recovery [27] and tend to be more resistant to treatment [1, 28]. TRD is also associated to longer time of treatment and increased costs [29].

2.3 Antidepressant treatment resistance assessment scales and stratification systems

2.3.1 Antidepressant treatment assessment scales

From the multiple published assessment scales, three of them stand out in literature: the Antidepressant Treatment History Form [30], the Harvard Antidepressant Treatment History [31], and the Massachusetts General Hospital Antidepressant Treatment Response Questionnaire [6]. The clinician performs the former two; the latter is performed by the patient.

The Antidepressant Treatment History Form (ATHF; 1990; revised 1999) was originally designed to assess the efficacy of antidepressant treatment before ECT [30]. The scale has five treatment levels, which go from 0 (no treatment) to 5 (high antidepressant doses plus lithium or triiodothyronine (T3) for at least 4 weeks, including antipsychotics in patients with depression and psychotic symptoms. The scale has been modified and recently digitalized [32]. This scale has the disadvantage of not including pharmacological combination strategies or preferences on switching treatment [33]. The Antidepressant Treatment History Form has been empirically validated with the monitoring of prospective treatment [8, 33, 34, 35]. The original ATHF version has a good inter-rater reliability [3], and the digitalized version has an excellent inter-rater reliability as well, with another evaluator [32].

The Harvard Antidepressant Treatment History (HATH) allows to systematically assess the dose and duration of previous antidepressant medication trials. The patient identifies all the antidepressants taken from a list of all available once, a series of systematic questions over dose, duration, and response are asked to determine treatment response or resistance [31].

The Massachusetts General Hospital Antidepressant Treatment Response Questionnaire (ATRQ) is an auto-evaluating scale that defines an appropriate antidepressant treatment as optimal dosage during 6 weeks. This questionnaire provides operational criteria for adequate dosing of each antidepressant and has been used on multiple multicentric studies in TRD. In one study, it was found that MGH and ATRQ agree with the independent evaluations of clinical researchers on remote interviews [36].

2.3.2 Treatment resistant depression stratification systems

Systems may help predict the ulterior course of depression on the long-term and its response to treatment [1]. It is important to point out that all systems based on treatment administration have limitations; they were designed when therapeutic options available at the time were more limited, and despite their clinical usefulness, they have not been properly validated.

Four stratification systems stand out in literature: the Staging Method of Thase and Rush [4], the European Staging Method [5, 37], the Massachusetts General Hospital Staging model (MGH-s) [6], and The Maudsley Staging Model (MSM) [7].

Thase and Rush proposed five levels of resistance, in which patients are categorized according to the number and antidepressant class they have failed to respond, from the most frequently used to the least usual as MAOIs or ECT [4]. Not one degree of each therapeutic trial in terms of dose and duration is recorded; it assumes that the non-response to two antidepressant agents of different classes is more difficult to treat than the non-response to agents of the same group and that the change to an antidepressant of the same group is less effective. The later may be true for tricyclic antidepressants, but it is not so at switching between different serotonin reuptake inhibitors (SSRIs) [17]. This classification considers that the most effective antidepressants on order would be MAOIs, tricyclic antidepressants, and SSRIs, which has not been validated on different meta-analyses in antidepressant trials [17, 20]. It also faces the disadvantage of not including other treatments, such as drug combinations and psychotherapy, and does not provide prognostic information [38]. The Thase and Rush scale is easy to implement and provides an accessible strategy for clinicians to treat TRD patients. However, it has been widely criticized recently, since its predictive value over the course of treatment has not been systematically evaluated [8, 33, 34].

A European Staging Method (ESM) proposes the classification between non-responders, patients with TRD, and chronic resistant depression (CRD). Non-responders are defined as patients who fail to respond to a method of treatment. Patients are considered TRD if they show poor response to two treatment options with different classes of antidepressants at a proper dose over the span of 6–8 weeks. CRD is defined as a resistant or refractory episode that lasts for more than a year despite appropriate therapeutic interventions. This scale has the advantage of including the duration of the depressive episodes and does not suggest a hierarchy of antidepressants. Non-response is clearly defined as a reduction inferior to 50% on the score of the HAMD [39] or the Montgomery Asberg Depression Rating Scale (MADRS) [40] and the TRD stages correspond to the number of failed therapeutic trials. It is assumed that patients with failure to respond to two agents of different classes would be more resistant than those who do not respond to two drugs of the same group and indirectly implies that switching to a drug from the same class is less effective [6]. It should be noted that, in this classification, the differences between TRD and CRD are arbitrary [8]. ESM does not establish that two or more attempts with failed antidepressants imply a higher level of TRD, in contrast with the publications that associate the number of changes with poor response to treatment [17, 18]. Furthermore, CRD does not consider non-pharmacological measures such as ECT or psychotherapy. To date, there are no studies that prove the predictive utility or reliability of the scale.

The Massachusetts General Hospital Staging model (MGH-s) was published for the first time by Fava [6] based on the scale by Thase and Rush. The MGH-s considers dosage optimization and separately prolonging the duration of treatment, as well as an operational criterion for minimal dose and duration of treatment. It includes measures for titration and combinations and does not rank antidepressant classes [41] or an implied preference between them or a change to the same group of drugs. The MGH-s considers the number of failed treatments and the intensity and optimization of each attempt and generates a continuous variable that reflects the degree of resistance to antidepressants. MGH-s generates a continuous score that reflects the level of resistance. However, this score is randomly given [8]. Dosage optimization and duration are considered equal to increase or combination, which does not seem to be backed up on literature [42, 43, 44]. Finally, the higher score given to ECT is not sufficiently explained [7, 45].

A study published a comparison between the MGH-s and the Thase and Rush Staging Method on a sample of 115 ambulatory patients with major depression. All results showed that both models have a high correlation, but the multivariable analysis demonstrated that the MGH-s had a better prediction for non-remission [45].

Fedaku et al. [7] published The Maudsley Staging Model (MSM) method of stratification, in which the TDR score varies from 3 to 15. TRD stages are shown in three categories: mild (scores 3–6), moderate (scores 7–10), and severe (scores 11–15). It incorporates the duration and severity of the depressive episode. MSM considers class switching between different antidepressant groups, and between the same group has the same score. The scale is easy to use and may be employed as a tridimensional model regarding duration, severity, and treatment. It has been criticized, however, that disease duration is arbitrary and does not include the number of titrating attempts. Empiric value and inter-rate reliability was proven with prospective data obtained from the notes of 88 patients on a TRD specialized unit and follow-up of 62 patients in this group for a medium of 29.5 months [7].

2.4 Resistant depression treatment general principles

Qualitative and quantitative assessment of depressive symptoms with evaluation scales on each visit, assessment tool employment, and watching over adverse effects should be a routine practice in the approach to the patient with major depressive disorder, and even more so with the TRD patient. Furthermore, it is recommended to evaluate psychosocial performance, quality of life, treatment compliance and tolerance, and provide 24-h assistance [46]. It is important to encourage patients not to abandon treatment or medical attention despite of not perceiving any results. A good relationship between the clinician and the patient is also important to guarantee treatment compliance [15]. Finally, the role of psycho-education for the patients and their relatives should not be forgotten, as this includes sign and symptom identification, prognosis, suicide risk assessment, treatment options, sleep hygiene, impulse control, and sleep restriction among others. Behind restriction the initial management of depressive patients is usually done by primary care physicians or internists, but they should be referred to a mental health specialist if there is not an appropriate response to two or more treatments, as well when suicidal or homicidal ideation, psychotic or catatonic behavior are detected.

2.4.1 Treatment strategies

For patients with major depressive disorder who do not respond to initial treatment with an antidepressant, there are diverse management strategies [5, 14, 30, 47, 48, 49, 50] which have been classified and arranged in three groups as follows: (1) optimization; (2) treatment switching (to a different antidepressant, psychotherapy, or ECT); (3) augmentation or adding other treatment to the one already in use, such as a different drug, psychotherapy, or ECT (Table 1). Regardless of the strategy, it is recommended always to use one strategy at a time to assess which is the most effective. We will review each one of them.

Table 1.

Treatment strategies for resistant depression.

2.4.1.1 Optimization

Optimization consists of improving the current treatment, while supervising good tolerance. It should be noted that, in some studies with patients diagnosed as resistant and sent to specialized clinics, it has been reported that an important number of them did not receive an appropriate dose of the medication or had been taking it behind for a brief period of time [51]. This would represent a case of pseudo-resistance. Several studies show that it is important to treat a major depressive episode for 6–12 weeks before concluding non-remittance [6, 26, 52]. A study in 1627 patients, where 67% of them received less than 4 weeks of treatment and did not show response to an antidepressant [53], did not find any difference between continuing the current treatment or changing to another antidepressant. This supports the importance of keeping the patients on an antidepressant treatment for an appropriate amount of time before changing it. On the other hand, multiple studies show that if patients have less than 25% reduction of symptoms after 4 weeks treatment, it could be indicated to switch to a different treatment strategy [54].

2.4.1.2 Treatment switching

Treatment switching is the act of suspending the current treatment and replacing it with a different pharmacologic or non-pharmacologic anti-depressive strategy. This includes using a different antidepressant, switching to psychotherapy, or ECT.

2.4.1.3 Changing antidepressants

It has been suggested that, in order to switch antidepressant medications, the current medication should be gradually discontinued while the new one is slowly introduced throughout 1–2 weeks and the dosage of the new antidepressant agent should be given at a corresponding amount to the one being discontinued. The clinician should be aware of increases of adverse effects and, in relation to SSRIs, the risk of serotonin syndrome. Some clinicians prefer to switch from an SSRI to another instantly, except when the patient has received fluoxetine, where the waiting time should be no less than 4 weeks before using another SSRI due to its long half-life.

For patients resistant to SSRIs, it has been suggested to switch to a selective noradrenaline and serotonin reuptake inhibitor (SNRI), atypical antidepressants, such as bupropion or mirtazapine, tricyclic antidepressants, or MAOIs. There are, however, few studies that compare switching to each one of these groups of drugs. In one meta-analysis that included four randomized studies with 1496 patients resistant to an SSRI, remission was evident in 24% of the patients who received another SSRI, and in 28% who were introduced to a different class of antidepressant such as bupropion, mirtazapine, or venlafaxine [55]. Regarding patients resistant to SSRI, many studies, including some meta-analyses, support changing to venlafaxine [17, 55]. A meta-analysis with 3375 patients with depression resistant to an SSRI showed that changing to another SSRI led to remission on 45% of the subjects, but 54% remitted when changing to venlafaxine [17]. Concerning atypical antidepressants, the few comparative studies available on TRD patients have not found differences respecting efficacy [18, 42]. In a group of patients resistant to paroxetine, a study that compared extended release venlafaxine (225 mg/day) and mirtazapine (45 mg/day) found remissions of 41 and 36% respectively [56]. In another study, 477 patients resistant to an SSRI treated for 14 weeks with bupropion SR (average dose 238 mg/day) or sertraline (average dose 136 mg/day) remission was achieved on 21 and 18% of the subjects, respectively [57]. An 8-week study that compared mirtazapine (45 mg/day) with paroxetine (20 mg/day) in 100 patients with TRD, remission was achieved in 36 and 47% with similar tolerance. Finally, a comparative study with mirtazapine (average dose 30 mg/day) and sertraline (average dose 120 mg/day) in 250 TRD patients, remission was similar (38 versus 28%, respectively) without statistical difference [58, 59, 60]. Nevertheless, there were more adverse effects with mirtazapine such as sedation, fatigue, weight gain, and xerostomia. In TRD patients, efficacy and tolerability of tricyclic antidepressants is comparable to that of atypical antidepressants and SSRIs [61]. Currently, tricyclic antidepressants have become the fourth or fifth line of treatment in TRD patients due to undesired adverse effects such as anticholinergic effects, cardiotoxicity, and lethal potential with overdose. The STAR*D study reported the fourth or fifth line of treatment in major depressive patients due to undesired adverse effects such as anticholinergic effects, cardiotoxicity, and lethal potential with overdose. In TRD patients, efficacy and tolerability of tricyclic antidepressants is comparable to that of atypical antidepressants and SSRIs. In the STAR*D study, an open 14-week trial in 235 patients compared nortriptyline (average dose 97 mg/day) with mirtazapine (average dose 42 mg/day) showing a comparable remission of 20 versus 12% and equal tolerance [62]. In a double-blind randomized study, 168 imipramine- or sertraline-resistant patients treated for 12 weeks were randomly assigned to the other treatment [63]. Remission was comparable (23% versus 32%) with more discontinuation with imipramine switching due to adverse effects (9 versus 0%). MAOIs are rarely used today, since they carry lethal potential by interacting with other drugs and food containing tyramine [64]. Nevertheless, changing to a MAOI may still be helpful for some TRD patients [17, 64]. A randomized trial with 46 imipramine-resistant patients who received phenelzine (45–90 mg/day) for 6 weeks and 22 phenelzine-resistant patients who were switched to imipramine (150–300 mg/day) reported a higher response on patients who received phenelzine rather than imipramine (67 versus 41%) [64]. For severely depressed patients with TRD, there is not enough evidence that indicates which kind of antidepressant is superior [65]. Tricyclic antidepressants may be preferred [66]. However, a meta-analysis of 25 randomized trials on 1377 hospitalized depressed patients who received tricyclic antidepressants or SSRIs showed that tricyclic antidepressant superiority over SSRIs was low with a higher rate of adverse effects [67].

2.4.1.4 Switching to psychotherapy

Changing from a pharmacologic approach to psychotherapy may be rejected by many TRD patients [68], but still is a reasonable approach. A 12-week trial with 122 patients who did not respond to citalopram and were randomized to cognitive behavioral therapy (CBT) or different antidepressants (bupropion, sertraline, or venlafaxine) [68] reported similar remission (25 and 28%). Another study with 140 patients who did not respond to a trial with nefazodone or CBT and then were switched to the other treatment [69] reported a comparable remission of 36 and 27%, respectively.

2.4.1.5 Electroconvulsive therapy

For patients with TRD with severe depression, ECT continues to be the therapy of choice [28, 66, 70, 71, 72]. The most important indications for ECT are persistence of suicidal ideation, suicide attempt, severe weight loss with malnutrition, dehydration, food or fluids rejection, and malignant catatonia. ECT is also indicated in cases of depression with psychotic symptoms and if there is previous history of response to this treatment. ECT has been shown superior to pharmacotherapy as shown by multiple meta-analyses and randomized studies [65]. A meta-analysis of 18 studies with 1144 patients that compared ECT with pharmacotherapy found that ECT was more effective [71]. ECT approach is recommended by many guidelines [66, 70, 72, 73]. ECT is not exempt of anesthetic risks, adverse effects, logistic problems, treatment rejection, and relapse.

2.4.1.6 Augmentation

Augmentation consists of adding other treatment (pharmacologic or non-pharmacologic) to the current one [74]. A new drug, psychotherapy, or transcranial magnetic stimulation (TME) might be added. This approach has been widely used and studied; combination therapy with antipsychotics, lithium, or triiiodothyronine (T3) are generally well tolerated [52, 58, 75, 76, 77, 78, 79], while combination therapy of MAOI with other antidepressants may cause serotonin syndrome or hypertensive crisis [80]. Previous response, safety, comorbidities, ease of use, patient’s preference, and costs are factors to consider while adding other drugs to the current treatment. TRD patients, who have had additions and do not respond in 6–12 weeks at the desired dose or do not tolerate the combination, should be switched to a second combination [58]. Some authors suggest discontinuation of the supplementary drug and addition of a new one progressively over 1–2 weeks [58].

2.4.1.7 Adding a second antidepressant

Concerning depression with partial response to monotherapy with antidepressants, a second drug is usually added. However, a meta-analysis of eight studies with 808 patients that did not respond to monotherapy and that compared antidepressant combination with monotherapy, found a similar improvement on both groups [81]. The most studied antidepressants are mirtazapine and bupropion.

Mirtazapine use as an augmentation drug on TRD patients is supported by the results of open and placebo-controlled studies [81, 82]. On the STAR*D study, mirtazapine was added to patients resistant to venlafaxine and was compared with switching to tranylcypromine (a MAOI). Both approaches had no different effects [83]. However, addition of mirtazapine to resistant patients requires additional studies to establish its efficacy.

Bupropion, a noradrenergic/dopaminergic reuptake inhibitor, was studied in TRD patients [84]. Bupropion has a good tolerability and low side effect profile, including few sexual side effects.

Buspirone, a serotonin (5-HT1A) receptor partial agonist, was studied in randomized, double-blind, placebo-controlled trials combined with an SSRI in patients with TRD [85]. Buspirone, at a dosage of 41 mg/day, was compared on the STAR*D study with Bupropion SR at 267 mg/day, with similar effectiveness [44]. As with mirtazapine, bupropion addition is a popular practice as an enhancing maneuver, but additional studies are needed to justify its use on TRD patients.

2.4.1.8 Second-generation antipsychotics

For second-generation antipsychotics in patients with TRD, the following order was suggested based on benefit and a lower rate of adverse effects: aripiprazole, quetiapine, risperidone, and less frequently ziprasidone or olanzapine [58, 77, 86, 87]. Also, the use of brexpiprazole has been suggested if aripiprazole generates akathisia [88, 89]. The analysis of 16 studies comparing the addition of aripiprazole, olanzapine, quetiapine, or risperidone with placebo on 3480 patients with non-psychotic depression who failed to at least one attempt with antidepressant monotherapy [90] showed improvement in 31% of the patients who received the additional drug (31%) versus placebo (17%). Discontinue due to adverse effects was higher with antipsychotics (9 versus 2%), 4% for aripiprazole, 12% for quetiapine, and 7% for risperidone [90]. In an open, randomized 12-week trial with 1522 patients who stayed severely depressed after treatment with an antidepressant [75], the subjects received three types of treatments: aripiprazole increase (target dose 5–15 mg/day), bupropion increase (target dose 300–400 mg/day), or switching to bupropion. Response (reduction equal or higher than 50% of depression) was greater with aripiprazole (74%) than increasing bupropion (66%) or switching to bupropion (62%). There were more adverse effects with aripiprazole, such as akathisia, somnolence, weight gain, and laboratory abnormalities, but patients experienced more anxiety with bupropion. A meta-analysis of 11 randomized trials on TRD patients (n > 3000) found that effectiveness of the augmentation of atypical antipsychotics might rise with the increase of the resistance [90]. For example, the augmentation may carry more benefit for patients that do not respond to three or four failed attempts compared with those who do not respond to one.

2.4.1.9 Lithium

Augmentation of treatment with lithium was reported for the first time by De Montigny by combining it with tricyclic antidepressants [91]. Thereafter, multiple studies have demonstrated its efficacy. A meta-analysis of 10 placebo, controlled studies showed that the addition of lithium at a dose to 600–900 mg/day (plasma levels higher than 0.4 mEq/L) was superior to placebo [78, 92]. A meta-analysis of nine trials with 237 patients comparing lithium versus placebo found a higher response with lithium [84]. Lithium was effective in the augmentation with first- and second-generation antidepressants attached to a possible benefit in reducing suicide risk. A meta-analysis of nine studies with 234 patients, where double-blind trials with lithium and placebo on TRD patients were included, showed a broad effectiveness with this approach [93]. The authors concluded that it should be given for no less than 7 days at dose of 600–800 mg/day [93]. An analysis of the literature reviewing 12 randomized studies on lithium augmentation of SSRIs or atypical antipsychotic drug therapy found no statistical difference that favors the use of one approach or the other [94].

2.4.1.10 Thyroid hormone

Thyroid hormone, in particular T3, has been used as an augmenting agent since the 1960s [95]. The usual dose of T3 in the form of liothyronine is 25–50 pg and with thyroxine (T4) is 150 pg. An initial meta-analysis with T3 showed effectiveness against placebo [79]. Subsequent studies, however, have shown limited evidence of its effectiveness. A meta-analysis with four randomized studies with 95 patients who did not respond to tricyclic antidepressants compared augmentation with T3 versus placebo or T4 showed response in 53% patients who received T3 but had not statistical difference with placebo [79]. However, T3 augmentation is not very popular in the UK nor in the USA [96].

2.4.1.11 Repetitive transcranial magnetic stimulation

Repetitive transcranial magnetic stimulation (rTMS) is a non-invasive technique in which a sequence of high-intensity magnetic pulses is used to stimulate cortical neurons to treat neuropsychiatric disorders including major depressive disorder [97]. This technique may be ambulatory, can be added to the current antidepressant treatment, and has good tolerability. Initial systematic reviews on mixed populations of depressed patients that included non-resistant patients supported their use on TRD patients [98, 99, 100, 101, 102, 103]. A meta-analysis of 24 trials that included 1092 patients who underwent rTMS and sham conditions showed rTMS was superior in clinical improvement of patients with TRD. The response and remissions were 25 and 17% and 9 and 6% for the rTMS and sham conditions, respectively [104]. Short duration rTMS (1–4 weeks) has an evident antidepressant effect on TRD patients and is well tolerated. Nevertheless, remission rates and responses with rTMS are low and it is unknown if there is a sustained effect. Not known is whether effects of TMS are sustained over time and its speed of onset [104].

2.4.1.12 Psychotherapy

Addition of CBT usually helps TRD patients, as demonstrated by multiple randomized studies [105, 106, 107]. In a randomized 1-year study with CBT (12–18 sessions), it was observed that remission occurred more on CBT group of patients (28%) than in patients who did not receive it (15%). In another study, depressive symptoms were minor on a self-report 40 months after patients received CBT [108]. A 12-week study compared citalopram plus 16-session CBT with citalopram plus additional pharmacological approaches such as bupropion or buspirone in 182 ambulatory patients resistant to citalopram [109]. The number of patients who achieved remission was similar (23 and 33%). A 12-week study that compared nefazodone treatment with 16- to 20-session CBT versus nefazodone alone on 446 ambulatory patients with chronic depression (medium of 8 years) showed remission in more patients undergoing the combination (48 versus 29%) [110]. In hospitalized patients with severe depression, combination therapy is a common practice and its effectiveness is clear. A 12-week study that compares CBT and pharmacotherapy with pharmacotherapy alone in 20 hospitalized patients with chronic depression found a similar improvement [111]. An observational 12-week study with CBT added to pharmacotherapy in 24 hospitalized patients with chronic depression [112] found a 46% reduction in depressive symptoms. In TRD patients, other types of psychotherapy such as group, family, or interpersonal therapy have not been well studied.

2.4.1.13 Other addition maneuvers

Several addition maneuvers have been employed in TRD patients, such as lamotrigine combination, stimulants like methylphenidate, modafinil, and pindolol among others. A meta-analysis which included 10 studies with 289 patients undergoing lamotrigine treatment concluded that this drug had little effect on non-bipolar TRD patients [113]. Controlled studies evaluating placebo versus methylphenidate have been negative despite its regular use [114]. Modafinil did not show a sustained effect in two controlled studies. However, a subsequent retrospective analysis suggests that modafinil may help TRD patients with fatigue and somnolence [115]. Pindolol, a non-selective beta-adrenergic antagonistic with effect in the 5-HT1A auto-receptor has shown negative results against placebo on TRD patients [116]. N-methyl-D-aspartate (NMDA) receptor acting drugs like memantine, ketamine, and riluzole have been studied. Controlled studies with memantine, an NMDA-receptor antagonist, have been negative [117]. Ketamine, an NMDA-receptor antagonist anesthetic, has shown positive antidepressant results in a controlled study against placebo in patients with TRD [118]. Riluzole, a putative glutamate release inhibitor used in the treatment of amyotrophic lateral sclerosis, did not show effectiveness on a controlled study to prevent relapse after ketamine use [119].

2.4.2 Studies comparing switching versus augmentation maneuvers

Multiple guidelines suggest how to use augmentation or switching maneuvers (The American Psychiatric Association, United Kingdom National Institute of Health and Clinical Excellence guidelines among others) [28, 66, 120, 121]. However, few randomized studies comparing the effectiveness of these maneuvers on TRD patients have been performed and most of them do not differ between those patients with little or no benefit from those who improve partially.

Various studies show that augmentation or switching maneuvers are equally efficient. Thus, multiple evaluations reviewing placebo-controlled randomized studies regarding augmentation and switching approaches show similar results [76]. The medium rate of remission with augmentation was 27% and switching was 22%, with response rates (reduction of 50% or more of the symptoms) of 38 and 40%, respectively [122]. A prospective study with citalopram on two groups of 269 patients who preferred augmentation with bupropion or buspirone to switching citalopram to bupropion, sertraline, or venlafaxine had a similar remission. A randomized study with 375 TRD patients, where several treatments were assigned, including five augmentations and two switching options [123, 124] showed similar results with 37 and 41% for each strategy. On the other hand, in a group with 1522 patients (85% men), almost 50% of them undergoing post-traumatic disorder, and who continued severely depressed after the first course of treatment with an antidepressant, almost all of them on psychotherapy, the augmentation approach with aripiprazole (5–15 mg/day) or bupropion (300–400 mg/day) was slightly superior to switching antidepressants to bupropion as monotherapy [75]. Remission was achieved in 29% of the patients with aripiprazole augmentation, 27% with bupropion augmentation, and 22% with switching to bupropion. Surveillance over 24 months of the remitted patients (n = 396) showed that approximately 25% of the patients in each group relapsed. There were more adverse effects in the aripiprazole group [75]. Other studies show that augmentation approach with aripiprazole may be more effective in women than in men [125].

With patients not tolerating the antidepressant dose, it is preferable to switch antidepressants. While there is evidence that suggests that augmentation is somehow superior to switching antidepressants, the decision should be discussed with the patient. Clinical criteria would be that patients who have had partial benefit from the initial antidepressant and have few adverse effects may prefer an augmentation approach and those with less improvement and more adverse effects might prefer switching medication. However, in patients resistant to a second treatment, there is no evidence that shows how many approaches should be done before considering change of treatment. Authors suggest 1–3 trials before switching [58, 74]. Changing medications has the advantage of achieving a better compliance to treatment than when more than one medication is used [126] adding a lower risk of adverse effects, pharmacologic interactions, and costs. A study evaluated 48 trials that included 6654 patients. A comparison was made between randomized studies, which compared 11 agents used on augmentation approaches: atypical antipsychotics (aripiprazole, olanzapine, quetiapine, and risperidone), antidepressants (bupropion, buspirone), lithium, thyroid hormone, methylphenidate, pindolol, and lamotrigine [87]. The studies analyzed were compared between them or placebo in patients with TRD and the proportion of patients who responded to treatment was defined as primary effectiveness. The analysis showed that primary effectiveness was higher with quetiapine, aripiprazole, thyroid hormone, and lithium in relation to placebo, but even higher for the former two in the sensibility analysis. There were no differences regarding discontinuation rate and adverse effects (acceptability) for these treatments [87]. Quetiapine, olanzapine, aripiprazole, and lithium were less well tolerated than placebo [87].

A randomized 8-week study on 140 TRD patients with paroxetine plus risperidone, paroxetine plus trazodone, and paroxetine plus thyroid hormone showed similar remission rates of 27, 43, and 38%, respectively [123]. A 6-week randomized open study that compared adding quetiapine (target dose 300 mg/day) with adding lithium (target plasma concentration from 0.6 to 1.2 mmol/L) in 450 resistant patients got a similar remission rate of 32 and 27%, respectively [127].

A meta-analysis with 48 randomized trials (n > 6000 depressed patients) in which efficiency of augmentation agents was evaluated using results from comparisons between drugs (on head to head trials), as well as indirect comparisons of the drugs through their relative effects with a common comparator (typically a placebo) [123]. The response (reduction equal or higher than 50%) or remission was more frequent when aripiprazole, lithium, olanzapine, quetiapine, risperidone, or thyroid hormone (T3 or T4) was added, compared to placebo; results from each one were comparable. Discontinuation due to adverse effects was higher with aripiprazole, lithium, olanzapine, and quetiapine than with placebo.

2.4.3 Promising new treatments

2.4.3.1 Acetylcholine receptor acting drugs

Medications that act on the cholinergic system seem promising in the treatment of TRD patients. Controlled studies versus placebo with intravenous scopolamine (a muscarinic antagonist) in TRD patients showed promising results [128]. Mecamylamine, a nAChR antagonist added to citalopram, was also superior to placebo [129]. Other drugs like mecamylamine, S-mecamylamine, and varenicline are currently in a preliminary stage of study in depressive patients [129, 130].

2.4.3.2 N-methyl-D-aspartate (NMDA) acting drugs

Ketamine, an NMDA-receptor antagonist, has shown antidepressant effects in TRD patients in a controlled study versus placebo [118]. However, ketamine, a dissociative anesthetic administration which complicates TRD patient treatment due to its route of administration (intravenous), requires hospitalization and consultation with an anesthesiologist. The rapid effects of ketamine usually disappear in 4–6 days. Also, it is possible that patients who improve on ketamine require a long-term course of maintenance. Ketamine has been given intranasal, or by sublingual delivery. Lapidus et al. [131] compared intranasal administration of ketamine 50 mg and placebo in 20 TRD patients. Patients improved at 24 h, but not at 72 h after administration. Recent studies are currently researching intranasal administration of esketamine, the S-enantiomer of racemic ketamine on TRD patients. Initial results are very promising [132]. If accepted, esketamine would enter the list of enhancement approaches for TRD patients.

Deep brain stimulation, vagus nerve stimulation, and neurosurgical lesions have also been evaluated in different studies as therapeutic options in highly resistant TRD patients [133].

2.5 Neurobiological aspects of resistant depression

Most antidepressants act by modulating serotonin, noradrenaline, and dopamine neurotransmission, but other neurotransmission routes seem to be involved such as cholinergic, glutamatergic, neuropeptides, and neuromodulators among others. The complex neurotransmission systems are prone to failure in the short- or long-term, blocking antidepressant action. Besides, different antidepressant treatments seem to exert action by different mechanisms modulating different cerebral regions [134]. Genetic variants may explain up to 42% of antidepressant response [135]. Genetic polymorphisms for cytochrome P-450 (CYP) enzymes may lead to a reduction on enzyme activity of the CYP2D6 or CYP3A4 variants, leading to intolerance to antidepressants on high plasmatic levels [136, 137]. Some patients are rapid metabolizers, resulting in low plasmatic levels at standard doses of antidepressants, leading to resistance. Evaluation of these genetic variants is accessible with pharmacogenetic studies of antidepressants. Other resistance genetic variants may be related to p-glycoprotein (p-gp), also known as the ABCB1 drug multiresistance gene [138]. Besides this, other polymorphic variants have been associated with response to antidepressants. In the case of serotonin 2A gene (HTR2A), both coding and noncoding polimorphisms have been associated to low SSRIs response [139, 140, 141]. Furthermore, single nucleotid polymorphisms (SNPs) of the genes for brain-derived neurotrophic factor (BDNF) [142, 143], the norepinephrine transporter [144], tryptophan hydroxylase 2 [145, 146], corticotrophin releasing hormone receptor 1 [147], the glucocorticoid receptor [148, 149], and the common promoter polymorphism of the serotonin transporter gene [150, 151, 152, 153, 154, 155] have been associated with low SSRI response.

It has been published that the activation of the immune system and neuroinflammation represent a primary event in the pathophysiology of TRD [156, 157, 158] and that the effect of NSAID may increase the effect of antidepressants [159].

Finally, the catecholaminergic hypothesis of depression [160, 161], which associates depression with low levels of neurotransmitters, was accepted to explain not only the neurobiochemistry of depression, but also the effect of antidepressant drugs. This hypothesis postulates that, in depression, the function of the dopamine, noradrenaline, and indolamine serotonin monoamines is decreased. In support of this, different studies have shown changes in plasma, urine, and cerebrospinal fluid concentrations of these neurotransmitters and their metabolites, changes in the density of neuroreceptors in platelets and neurons, flattened curves in neuroendocrine challenges and also early relapses with the blockade of restriction enzymes for neurotransmitter synthesis in patients who had achieved depression remission with antidepressant treatment [162]. The existence of subtypes of depression, where noradrenergic, serotoninergic, or dopaminergic negative balance is predominant, has been also postulated. Patients with these subtypes of depression hypothetically would respond better to antidepressant drugs with noradrenergic, serotoninergic and dopaminergic effects. Unfortunately, clinical studies on the effect of antidepressants with different mechanisms of action show contradicting results, and today there are not clear clinical or biological parameters to predict the results of different antidepressant treatments [163]. It should be noted that any theory about the cause of TRD would be simplistic including that the deficit of a single neurotransmitter, genetic, or immune system and neuroinflammation responses would be present in most TRD patients. However, knowing if there are groups of patients that may respond better to drugs with different mechanisms of action continues to be important for the treatment of patients with TRD [164].

It has been suggested that, in the selection of an antidepressant drug, the clinician must observe the overall response of the patient with major depression [165]. Also, in the selection of an antidepressant drug, the clinician must observe the possible relationship between the drug’s biochemical effect and its effects on specific symptoms but also on adverse events. This became more relevant when a drug or group of drugs have failed to improve the patient and a new one has to be supplied, when adverse events force treatment change or when augmentation or change of antidepressant treatment is considered.

Advertisement

3. Conclusions

Once resistance to treatment with two drugs with different action mechanisms has been established, the next best therapeutic decision is terra ignota because there is not enough scientific information available to validate which steps are to follow: whether change treatment, adding an antidepressant, “buster therapies” like addition of lithium, thyroid hormone or stimulants, add atypical antipsychotics, rTMS or employment of newly treatments such as ketamine, or ECT. In the evaluation stages for the treatment of a patient with TRD it is important to make an evaluation and reassessment of the case. This includes confirming the diagnosis of major depressive disorder, making the differential diagnoses of bipolar depression or other forms of resistant depression such as secondary to other medical issues, drugs, etc. Medical and psychiatric comorbidities, as well as depression severity should be assessed. Also, a detailed clinical history on antidepressant use should be performed. The application of diagnostic tools or evaluation scales is relevant.

Despite of its importance and frequency, there is no consensus over what TRD is. Advances have been made over assessment tools to evaluate resistance to treatment. However, there is no consensus over which is the best stratification system for TRD. There is a lack of research that validates which treatment approaches may be more effective and which ones should be used in the different stages in the management of a resistant patient. Unfortunately, advances over neurobiology of depression cannot be transferred yet to a clinical level to help the physician choose the best treatment for a patient with major depression and even less so for a TRD patient [163, 164]. In patients who have an inadequate response to the first line of treatment, the clinician has many options to change the treatment, but if the second approach fails, other approaches seem to be equally effective in according to what is published on the literature and there are no clear guidelines that support one or the other. This outlook is discouraging for patients and physicians who are on trial and error until they find something that helps the patient. Future research on TRD patients should be centered on neurobiological factors involved in the development of the resistance including pharmacogenetics. Without the development of techniques that help us predict which factors are related to this phenomenon, treatment of TRD patients will continue to be insufficient.

Advertisement

Acknowledgments

The author wishes to thank the Department of Psychiatry of the University Hospital UANL and the INFOSAME for its support in the preparation of this manuscript.

Advertisement

Conflict of interest

Dr. Jose Alfonso Ontiveros has received research grants from AstraZeneca, Glaxo Smith Kline, Eli Lilly, Servier, Wyeth, Lundbeck, Janssen-Cilag, Pfiser, and Sunovion.

References

  1. 1. Crown WH, Finkelstein S, Berndt ER, Ling D, Poret AW, Rush AJ, et al. The impact of treatment-resistant depression on healthcare utilization and cost. The Journal of Clinical Psychiatry. 2002;63(11):963-971
  2. 2. Berlim MT, Turecki G. What is the meaning of treatment resistant/refractory major depression (TRD)? A systematic review of current randomized trials. European Neuropsychopharmacology. 2007;17(11):696-707
  3. 3. Sackeim HA, Prudic J, Devanand DP, Decina P, Kerr B, Malitz S. The impact of medication resistance and continuation pharmacotherapy on relapse following response to electroconvulsive therapy in major depression. Journal of Clinical Psychopharmacology. 1990;10(2):96-104
  4. 4. Thase ME, Rush AJ. When at first you don't succeed: Sequential strategies for antidepressant nonresponders. The Journal of Clinical Psychiatry. 1997;58(Suppl 13):23-29
  5. 5. Souery D, Amsterdam J, de Montigny C, Lecrubier Y, Montgomery S, Lipp O, et al. Treatment resistant depression: methodological overview and operational criteria. European Neuropsychopharmacology. 1999;9(1-2):83-91
  6. 6. Fava M. Diagnosis and definition of treatment-resistant depression. Biological Psychiatry. 2003;53(8):649-659
  7. 7. Fekadu A, Wooderson S, Donaldson C, Markopoulou K, Masterson B, Poon L, et al. A multidimensional tool to quantify treatment resistance in depression: The Maudsley staging method. The Journal of Clinical Psychiatry. 2009;70(2):177-184
  8. 8. Berlim MT, Turecki G. Definition, assessment, and staging of treatment-resistant refractory major depression: A review of current concepts and methods. Canadian Journal of Psychiatry. 2007;52(1):46-54
  9. 9. Fawcett J, Kravitz HM. Treatment refractory depression. In: Schatzberg AF, editor. Common Treatment Problems in Depression. Washington DC: American Psychiatric Press; 1985. pp. 2-27
  10. 10. Lehmann HE. Therapy-resistant depressions—A clinical classification. Pharmakopsychiatrie, Neuro-Psychopharmakologie. 1974;7(3):156-163
  11. 11. Fava M, Davidson KG. Definition and epidemiology of treatment-resistant depression. The Psychiatric Clinics of North America. 1996;19:179-200
  12. 12. Trivedi MH, Rush AJ, Wisniewski SR, Nierenberg AA, Warden D, Ritz L, et al. Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: Implications for clinical practice. The American Journal of Psychiatry. 2006;163(1):28-40
  13. 13. Vergouwen AC, Bakker A, Katon WJ, Verheij TJ, Koerselman F. Improving adherence to antidepressants: A systematic review of interventions. The Journal of Clinical Psychiatry. 2003;64(12):1415-1420
  14. 14. O’Reardon JP, Amsterdam JD. Treatment-resistant depression: Progress and limitations. Psychiatric Annals. 1998;28:633-640
  15. 15. Rush AJ, Thase ME, Dube S. Research issues in the study of difficult-to-treat depression. Biological Psychiatry. 2003;53:743-53.17
  16. 16. Committee for Medicinal Product for Human Use (CHMP). Concept Paper on the Need for Revision of Note for Guidance on Clinical Investigation of Medicinal Products in the Treatment of Depression with regard to Treatment Resistant Depression; European Medicines Agency. 2009. pp. 1-4. Available from: https://www.ema.europa.eu/en/documents/scientific-guideline/concept-paper-need-revision-note-guidance-clinical-investigation-medicinal-products-treatment_en-0.pdf
  17. 17. Ruhe HG, Huyser J, Swinkels JA, Schene AH. Switching antidepressants after a first selective serotonin reuptake inhibitor in major depressive disorder: A systematic review. The Journal of Clinical Psychiatry. 2006;67(12):1836-1855
  18. 18. Rush AJ, Trivedi MH, Wisniewski SR, Nierenberg AA, Stewart JW, Warden D, et al. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: A STAR*D report. The American Journal of Psychiatry. 2006;163(11):1905-1917
  19. 19. O’Reardon JP, Amsterdam JD. Overview of treatment-resistant depression and its management. In: Amsterdam JD, Hornig M, Nierenberg AA, editors. Treatment-Resistant mood disorders. New York (NY): Cambridge University Press; 2001. pp. 30-45
  20. 20. O’Reardon JP, Brunswick DJ, Amsterdam JD. Treatment-resistant depression in the age of serotonin: Evolving strategies. Current Opinion in Psychiatry. 2000;13:93-98
  21. 21. Henricus G, Ruhé HG, Van Rooijen G, Spijker J, Peeters F, Schene A. Staging methods for treatment resistant depression. A systematic review. Journal of Affective Disorders. 2012;137:35-45
  22. 22. Nemeroff CB. Prevalence and management of treatment-resistant depression. The Journal of Clinical Psychiatry. 2007;68(Suppl 8):17-25
  23. 23. Trivedi MH, Kleiber BA. Using treatment algorithms for the affective management of treatment-resistant depression. Journal of Clinical Psychiatry. 2001;62(Suppl 18):25-29
  24. 24. Keitner GI, Ryan CE, Solomon DA. Realistic expectations and a disease management model for depressed patients with persistent symptoms. The Journal of Clinical Psychiatry. 2006;67(9):1412-1421
  25. 25. Keller MB, Shapiro RW, Lavori PW, Wolfe N. Recovery in major depressive disorder: Analysis with the life table and regression models. Archives of General Psychiatry. 1982;39(8):905-910
  26. 26. Rush AJ. STAR*D: What have we learned? The American Journal of Psychiatry. 2007;164:201
  27. 27. Mueller TI, Keller MB, Leon AC, Solomon DA, Shea MT, Coryell W, et al. Recovery after 5 years of unremitting major depressive disorder. Archives of General Psychiatry. 1996;53(9):794-799
  28. 28. Malhi GS, Adams D, Porter R, et al. Clinical practice recommendations for depression. Acta Psychiatrica Scandinavica. Supplementum. 2009;(439):8-26
  29. 29. Ustün TB, Kessler RC. Global burden of depressive disorders: The issue of duration. The British Journal of Psychiatry. 2002;181:181-183
  30. 30. Sackeim HA. The definition and meaning of treatment-resistant depression. The Journal of Clinical Psychiatry. 2001;62(Suppl 16):10-17
  31. 31. Nierenberg AA, Keck P, Samson J, Rothschild AJ, Schatzberg AF. Methodological considerations for the study of treatment-resistant depression. In: Amsterdam JD, editor. Refractory Depression. New York (NY): Raven Press; 1991. pp. 1-12
  32. 32. Oquendo MA, Baca-Garcia E, Kartachov A, Khait V, Campbell CE, Richards M, et al. A computer algorithm for calculating the adequacy of antidepressant treatment in unipolar and bipolar depression. The Journal of Clinical Psychiatry. 2003;64(7):825-833
  33. 33. Ruhé HG, van Rooijen G, Spijker J, Peeters FPML, Schene AH. Staging methods for treatment resistant depression. A systematic review. Journal of Affective Disorders. 2012;137(1-3):35-45
  34. 34. Bird D, Haddad PM, Dursun SM. An overview of the definition and management of treatment-resistant depression. Klinik Psikofarmakoloji Bülteni. 2002;12:92-101
  35. 35. Fagiolini A, Kupfer DJ. Is treatment-resistant depression a unique subtype of depression? Biological Psychiatry. 2003;53:640-648
  36. 36. Chandler GM, Iosifescu D, Pollack M, Targum S, Fava M. Validation of the Massachusetts General Hospital Antidepressant Treatment History Questionnaire (ATRQ). CNS Neuroscience and Therapeutics. 2010;16:322-325. DOI: 10.1111/j.1755-5949.2009.00102.x
  37. 37. Souery D, Lipp O, Massat I, Mendlewicz J. The characterization and definition of treatment-resistant mood disorders. In: Amsterdam JD, Hornig M, Nierenberg AA, editors. Treatment-Resistant Mood Disorders. New York (NY): Cambridge University Press; 2001. pp. 30-45
  38. 38. Souery D, Van der Auwera K. The multiple facets of treatment-resistant depression. CNS Spectrums. 2004;9(11):803-807
  39. 39. Hamilton M. A rating scale for depression. Journal of Neurology, Neurosurgery, and Psychiatry. 1960;23:56-61
  40. 40. Montgomery SA, Asberg M. A new depression scale designed to be sensitive to change. The British Journal of Psychiatry. 1979;1979:134, 382-389
  41. 41. Souery D, Papakostas GI, Trivedi MH. Treatment-resistant depression. The Journal of Clinical Psychiatry. 2006;67(Suppl 6):16-22
  42. 42. Ruhe HG, Huyser J, Swinkels JA, Schene AH. Dose escalation for insufficient response to standard-dose selective serotonin reuptake inhibitors in major depressive disorder: Systematic review. The British Journal of Psychiatry. 2006;189:309-316
  43. 43. Ruhe HG, Booij J, Weert HCV, Reitsma JB, Franssen EJF, Michel MC, et al. Evidence why dose-escalation of paroxetine in major depressive disorder is not effective: A 6-week, randomized-controlled trial with assessment of serotonin transporter occupancy. Neuropsychopharmacology. 2009;34:999-1010
  44. 44. Trivedi MH, Fava M, Wisniewski SR, Thase ME, Quitkin F, Warden D, et al. Medication augmentation after the failure of SSRIs for depression. The New England Journal of Medicine. 2006;354(12):1243-1252
  45. 45. Petersen T, Papakostas GI, Posternak MA, Kant A, Guyker WM, Iosifescu DV, et al. Empirical testing of two models for staging antidepressant treatment resistance. Journal of Clinical Psychopharmacology. 2005;25(4):336-341
  46. 46. Keller MB. Issues in treatment-resistant depression. The Journal of Clinical Psychiatry. 2005;66:5-12
  47. 47. Berman RM, Narasimhan M, Charney DS. Treatment-refractory depression: Definitions and characteristics. Depression and Anxiety. 1997;5:154-164
  48. 48. Sharan P, Saxena S. Treatment-resistant depression: Clinical significance, concept and management. National Medical Journal of India. 1998;11:69-79
  49. 49. Shelton RC et al. Therapeutic options for treatment-resistant depression. CNS Drugs. 2010;24(2):131-161
  50. 50. Kornstein SG, Schneider RK. Clinical features of treatment-resistant depression. The Journal of Clinical Psychiatry. 2001;62(Suppl 16):18-25
  51. 51. Hirschfeld RM, Keller MB, Panico S, et al. The National Depressive and Manic-Depressive Association consensus statement on the under-treatment of depression. JAMA. 1997;277:333-340
  52. 52. Papakostas GI. Managing partial response or nonresponse: Switching, augmentation, and combination strategies for major depressive disorder. The Journal of Clinical Psychiatry. 2009;70(Suppl 6):16
  53. 53. Bschor T, Kern H, Henssler J, Baethge C. Switching the antidepressant after nonresponse in adults with major depression: A systematic literature search and meta-analysis. The Journal of Clinical Psychiatry. 2018;79(1):16r10749. DOI: 10.4088/JCP.16r10749
  54. 54. Kudlow PA, McIntyre RS, Lam RW. Early switching strategies in antidepressant non-responders: Current evidence and future research directions. CNS Drugs. 2014;28:601
  55. 55. Papakostas GI, Fava M, Thase ME. Treatment of SSRI-resistant depression: A meta-analysis comparing within-versus across-class switches. Biological Psychiatry. 2008;63:699
  56. 56. Gaynes BN, Dusetzina SB, Ellis AR, et al. Treating depression after initial treatment failure: Directly comparing switch and augmenting strategies in STAR*D. Journal of Clinical Psychopharmacology. 2012;32:114
  57. 57. Rush AJ, Trivedi MH, Wisniewski SR, et al. Bupropion-SR, sertraline, or venlafaxine-XR after failure of SSRIs for depression. The New England Journal of Medicine. 2006;354:1231
  58. 58. 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;71:43
  59. 59. Thase ME, Kremer C, Rodrigues H. Mirtazapine versus sertraline after SSRI non-response. European Neuropsychopharmacology. 2001;11(3):S342
  60. 60. Thase ME. Mirtazapine vs. sertraline in SSRI TRD; Symposium abstracts: The International Society for Affective Disorders (ISAD) - 2nd Biennial Conference - Cancun, Mexico 5th - 10th March 2004. Journal of Affective Disorders. 2004;78(Suppl):S49
  61. 61. Hirschfeld RM, Montgomery SA, Aguglia E, et al. Partial response and nonresponse to antidepressant therapy: Current approaches and treatment options. The Journal of Clinical Psychiatry. 2002;63:826
  62. 62. Fava M, Rush AJ, Wisniewski SR, et al. A comparison of mirtazapine and nortriptyline following two consecutive failed medication treatments for depressed outpatients: A STAR*D report. The American Journal of Psychiatry. 2006;163:1161
  63. 63. Thase ME, Rush AJ, Howland RH, et al. Double-blind switch study of imipramine or sertraline treatment of antidepressant-resistant chronic depression. Archives of General Psychiatry. 2002;59:233
  64. 64. Thase ME. The role of monoamine oxidase inhibitors in depression treatment guidelines. The Journal of Clinical Psychiatry. 2012;73(Suppl 1):10
  65. 65. Fekadu A, Rane LJ, Wooderson SC, et al. Prediction of longer-term outcome of treatment-resistant depression in tertiary care. The British Journal of Psychiatry. 2012;201:369
  66. 66. Anderson IM, Ferrier IN, Baldwin RC, et al. Evidence-based guidelines for treating depressive disorders with antidepressants: A revision of the 2000 British Association for Psychopharmacology guidelines. Journal of Psychopharmacology. 2008;22:343
  67. 67. Anderson IM. SSRIS versus tricyclic antidepressants in depressed inpatients: A meta-analysis of efficacy and tolerability. Depression and Anxiety. 1998;7(Suppl 1):11
  68. 68. Wisniewski SR, Fava M, Trivedi MH, et al. Acceptability of second-step treatments to depressed outpatients: A STAR*D report. The American Journal of Psychiatry. 2007;164:753
  69. 69. Schatzberg AF, Rush AJ, Arnow BA, et al. Chronic depression: Medication (nefazodone) or psychotherapy (CBASP) is effective when the other is not. Archives of General Psychiatry. 2005;62:513
  70. 70. Kennedy SH, Milev R, Giacobbe P, et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) Clinical guidelines for the management of major depressive disorder in adults. IV. Neurostimulation therapies. Journal of Affective Disorders. 2009;117(Suppl 1):S44
  71. 71. UK ECT Review Group. Efficacy and safety of electroconvulsive therapy in depressive disorders: A systematic review and meta-analysis. Lancet. 2003;361:799
  72. 72. American Psychiatric Association. Practice guideline for the treatment of patients with major depressive disorder. 3rd ed. American Journal of Psychiatry. 2010;167(Supplement):1. Available from: http://psychiatryonline.org/guidelines.aspx [Accessed: April 17, 2012]
  73. 73. Trangle M, Dieperink B, Gabert T, et al. Institute for Clinical Systems Improvement. Major Depression in Adults in Primary Care. Updated May 2012. Available from: http://www.icsi.org/depression_5/depression__major__in_adults_in_primary_care_3.html [Accessed: December 12, 2012]
  74. 74. Thase ME. Augmentation strategies for depression: History and concepts. CNS Spectrums. 2007;12:3
  75. 75. Mohamed S, Johnson GR, Chen P, et al. Effect of antidepressant switching vs augmentation on remission among patients with major depressive disorder unresponsive to antidepressant treatment: The VAST-D randomized clinical trial. JAMA. 2017;318:132
  76. 76. Santaguida P, MacQueen G, Keshavarz H, et al. Treatment for Depression After Unsatisfactory Response to SSRIs. Comparative Effectiveness Review No. 62 (Prepared by McMaster University Evidence-based Practice Center under Contract No. HHSA 290 2007 10060 I). AHRQ Publication No. 12-EHC050-EF. Rockville, MD: Agency for Healthcare Research and Quality; 2012. Available from: www.ahrq.gov/clinic/epcix.htm
  77. 77. Nelson JC, Papakostas GI. Atypical antipsychotic augmentation in major depressive disorder: A meta-analysis of placebo-controlled randomized trials. The American Journal of Psychiatry. 2009;166:980
  78. 78. Crossley NA, Bauer M. Acceleration and augmentation of antidepressants with lithium for depressive disorders: Two meta-analyses of randomized, placebo-controlled trials. The Journal of Clinical Psychiatry. 2007;68:935
  79. 79. Aronson R, Offman HJ, Joffe RT, Naylor CD. Triiodothyronine augmentation in the treatment of refractory depression. A meta-analysis. Archives of General Psychiatry. 1996;53:842
  80. 80. Preskorn SH. Treatment options for the patient who does not respond well to initial antidepressant therapy. Journal of Psychiatric Practice. 2009;15:202
  81. 81. Henssler J, Bschor T, Baethge C. Combining antidepressants in acute treatment of depression: A meta-analysis of 38 studies including 4511 patients. Canadian Journal of Psychiatry. 2016;61:29
  82. 82. Carpenter LL, Yasmin S, Price LH. A double-blind, placebo-controlled study of antidepressant augmentation with mirtazapine. Biological Psychiatry. 2002;51(2):183-188
  83. 83. McGrath PJ, Stewart JW, Fava M, Trivedi MH, Wisniewski SR, Nierenberg AA, et al. Tranylcypromine versus venlafaxine plus mirtazapine following three failed antidepressant medication trials for depression: A STAR*D report. The American Journal of Psychiatry. 2006;163(9):1531-1541. Quiz 666.31
  84. 84. Bodkin JA, Lasser RA, Wines JD Jr, et al. Combining serotonin reuptake inhibitors and bupropion in partial responders to antidepressant monotherapy. The Journal of Clinical Psychiatry. 1997;58(4):137-145
  85. 85. Landen M, Bjorling G, Agren H, Fahlen T. A randomized, double-blind, placebo-controlled trial of buspirone in combination with an SSRI in patients with treatment-refractory depression. The Journal of Clinical Psychiatry. 1988;59(12):664-668
  86. 86. Philip NS, Carpenter LL, Tyrka AR, Price LH. Depression: A re-examination for the modern era. Expert Opinion on Pharmacotherapy. 2010;11(5):709-722
  87. 87. Zhou X, Ravindran AV, Qin B, et al. Comparative efficacy, acceptability, and tolerability of augmentation agents in treatment-resistant depression: Systematic review and network meta-analysis. The Journal of Clinical Psychiatry. 2015;76(4):487-498
  88. 88. Thase ME, Youakim JM, Skuban A, et al. Adjunctive brexpiprazole 1 and 3 mg for patients with major depressive disorder following inadequate response to antidepressants: A phase 3, randomized, double-blind study. The Journal of Clinical Psychiatry. 2015;76:1232
  89. 89. Thase ME, Youakim JM, Skuban A, et al. Efficacy and safety of adjunctive brexpiprazole 2 mg in major depressive disorder: A phase 3, randomized, placebo-controlled study in patients with inadequate response to antidepressants. The Journal of Clinical Psychiatry. 2015;76:1224
  90. 90. Wang HR, Woo YS, Ahn HS, et al. Can atypical antipsychotic augmentation reduce subsequent treatment failure more effectively among depressed patients with a higher degree of treatment resistance? A meta-analysis of randomized controlled trials. The International Journal of Neuropsychopharmacology. 2015;18:pyv023. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4571632/
  91. 91. De Montigny C, Grunberg F, Mayer A, Deschenes JP. Lithium induces rapid relief of depression in tricyclic antidepressant drug non-responders. The British Journal of Psychiatry. 1981;138:252-256
  92. 92. Nelson JC, Baumann P, Delucchi K, et al. A systematic review and meta-analysis of lithium augmentation of tricyclic and second generation antidepressants in major depression. Journal of Affective Disorders. 2014;168:269
  93. 93. Bauer M, Döpfmer S. Lithium augmentation in treatment-resistant depression: Meta-analysis of placebo-controlled studies. Journal of Clinical Psychopharmacology. 1999;19(5):427-434
  94. 94. Edwards SJ, Hamilton V, Nherera L, Trevor N. Lithium or an atypical antipsychotic drug in the management of treatment-resistant depression: A systematic review and economic evaluation. Health Technology Assessment. 2013;17(54):1-190
  95. 95. Prange AJ Jr, Wilson IC, Rabon AM, Lipton MA. Enhancement of imipramine antidepressant activity by thyroid hormone. The American Journal of Psychiatry. 1969;126:457
  96. 96. Dorling CM. Antidepressant augmentation and combinations. Psychiatric Clinics of North America. 2000;23(4):743-755
  97. 97. Eitan R, Lerer B. Nonpharmacological, somatic treatments of depression: Electroconvulsive therapy and novel brain stimulation modalities. Dialogues in Clinical Neuroscience. 2006;8(2):241-258
  98. 98. McNamara B, Ray JL, Arthurs OJ, et al. Transcranial magnetic stimulation for depression and other psychiatric disorders. Psychological Medicine. 2001;31(7):1141-1146
  99. 99. Holtzheimer PE, Russo J, Avery DH. A meta-analysis of repetitive transcranial magnetic stimulation in the treatment of depression. Psychopharmacology Bulletin. 2001;35(4):149-169
  100. 100. Burt T, Lisanby SH, Sackeim HA. Neuropsychiatric applications of transcranial magnetic stimulation: A meta analysis. The International Journal of Neuropsychopharmacology. 2002;5(1):73-103
  101. 101. Kozel FA, George MS. Meta-analysis of left prefrontal repetitive transcranial magnetic stimulation (rTMS) to treat depression. Journal of Psychiatric Practice. 2002;8(5):270-275
  102. 102. Martin JL, Barbanoj MJ, Schlaepfer TE, et al. Repetitive transcranial magnetic stimulation for the treatment of depression. Systematic review and meta-analysis. The British Journal of Psychiatry. 2003;182:480-491
  103. 103. Couturier JL. Efficacy of rapid-rate repetitive transcranial magnetic stimulation in the treatment of depression: A systematic review and meta-analysis. Journal of Psychiatry & Neuroscience. 2005;30(2):83-90
  104. 104. Lam RW, Chan P, Wilkins-Ho M, Yatham LN. Repetitive transcranial magnetic stimulation for treatment-resistant depression: A systematic review and metaanalysis. Canadian Journal of Psychiatry. 2008;53(9):621-631
  105. 105. Trivedi RB, Nieuwsma JA, Williams JW Jr. Examination of the utility of psychotherapy for patients with treatment resistant depression: A systematic review. Journal of General Internal Medicine. 2011;26:643
  106. 106. Eisendrath SJ, Gillung E, Delucchi KL, et al. A randomized controlled trial of mindfulness-based cognitive therapy for treatment-resistant depression. Psychotherapy and Psychosomatics. 2016;85:99
  107. 107. Watkins ER, Mullan E, Wingrove J. et al, Rumination-focused cognitive-behavioural therapy for residual depression: Phase II randomised controlled trial. The British Journal of Psychiatry. 2011;199:317
  108. 108. Wiles NJ, Thomas L, Turner N, et al. Long-term effectiveness and cost-effectiveness of cognitive behavioural therapy as an adjunct to pharmacotherapy for treatment-resistant depression in primary care: Follow-up of the CoBalT randomised controlled trial. Lancet Psychiatry. 2016;3:137
  109. 109. Thase ME, Friedman ES, Biggs MM, et al. Cognitive therapy versus medication in augmentation and switch strategies as second-step treatments: A STAR*D report. The American Journal of Psychiatry. 2007;164:739
  110. 110. Keller MB, McCullough JP, Klein DN, et al. A comparison of nefazodone, the cognitive behavioral-analysis system of psychotherapy, and their combination for the treatment of chronic depression. The New England Journal of Medicine. 2000;342:1462
  111. 111. Barker WA, Scott J, Eccleston D. The Newcastle chronic depression study: Results of a treatment regime. International Clinical Psychopharmacology. 1987;2:261
  112. 112. Scott J. Chronic depression: Can cognitive therapy succeed when other treatments fail? Behavioural Psychotherapy. 1992;20:25
  113. 113. Thomas SP, Nandhra HS, Jayaraman A. Systematic review of lamotrigine augmentation of treatment resistant unipolar depression (TRD). Review published. Journal of Mental Health. 2010;19(2):168-175
  114. 114. Ravindran AV, Kennedy SH, O'Donovan MC, Fallu A, Camacho F, Binder CE. Osmotic-release oral system methylphenidate augmentation of antidepressant monotherapy in major depressive disorder: Results of a double-blind, randomized, placebo-controlled trial. The Journal of Clinical Psychiatry. 2008;69(1):87-94
  115. 115. Fava M, Thase ME, De Battista C. A multicenter, placebo-controlled study of modafinil augmentation in partial responders to selective serotonin reuptake inhibitors with persistent fatigue and sleepiness. The Journal of Clinical Psychiatry. 2005;66(1):85-93
  116. 116. Sokolski KN, Conney JC, Brown BJ, De Met EM. Once-daily high-dose pindolol for SSRI-refractory depression. Psychiatry Research. 2004;125(2):81-86
  117. 117. Zarate CA, Singh JB, Quiroz JA, De Jesus G, Denicoff KK, Luckenbaugh DA, et al. A double-blind, placebo-controlled study of memantine in the treatment of major depression. The American Journal of Psychiatry. 2006;163(1):153-155
  118. 118. Zarate CA, Singh JB, Carlson PJ, Brutsche NE, Ameli R, Luckenbaugh DA, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Archives of General Psychiatry. 2006;63(8):856-864
  119. 119. Mathew SJ, Murrough JW, Aan Het Rot M, Collins KA, Reich DL, Charney DS. Riluzole for relapse prevention following intravenous ketamine in treatment-resistant depression: A pilot randomized, placebo-controlled continuation trial. The International Journal of Neuropsychopharmacology. 2009;17:1-12
  120. 120. Kennedy SH, Lam RW, McIntyre RS, et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) 2016 Clinical guidelines for the management of adults with major depressive disorder: Section 3. Pharmacological treatments. Canadian Journal of Psychiatry. 2016;61:540
  121. 121. Spijker J, Nolen WA. An algorithm for the pharmacological treatment of depression. Acta Psychiatrica Scandinavica. 2010;121:180
  122. 122. Gaynes BN, Lux L, Lloyd S, et al. Nonpharmacologic Interventions for Treatment-Resistant Depression in Adults. Comparative Effectiveness Review No. 33. AHRQ Publication No. 11-EHC056-EF. Rockville, MD: Agency for Healthcare Research and Quality; 2011. Available from: http://www.effectivehealthcare.ahrq.gov/ or 800-358-9295. PMID: 22091472
  123. 123. Fang Y, Yuan C, Xu Y, et al. A pilot study of the efficacy and safety of paroxetine augmented with risperidone, valproate, buspirone, trazodone, or thyroid hormone in adult Chinese patients with treatment-resistant major depression. Journal of Clinical Psychopharmacology. 2011;31:638
  124. 124. Fang Y, Yuan C, Xu Y, et al. Comparisons of the efficacy and tolerability of extended-release venlafaxine, mirtazapine, and paroxetine in treatment-resistant depression: A double-blind, randomized pilot study in a Chinese population. Journal of Clinical Psychopharmacology. 2010;30:357
  125. 125. Thase ME, Trivedi MH, Nelson JC, et al. Examining the efficacy of adjunctive aripiprazole in major depressive disorder: A pooled analysis of 2 studies. The Primary Care Companion to The Journal of Clinical Psychiatry. 2008;10:440
  126. 126. Osterberg L, Blaschke T. Adherence to medication. The New England Journal of Medicine. 2005;353:487
  127. 127. Bauer M, Dell'osso L, Kasper S, et al. Extended-release quetiapine fumarate (quetiapine XR) monotherapy and quetiapine XR or lithium as add-on to antidepressants in patients with treatment-resistant major depressive disorder. Journal of Affective Disorders. 2013;151:209
  128. 128. Drevets WC, Zarate CA Jr, Furey ML. Antidepressant effects of the muscarinic cholinergic receptor antagonist scopolamine: A review. Biological Psychiatry. 2013;73:1156-1163
  129. 129. George TP, Sacco KA, Vessicchio JC, Weinberger AH, Shytle RD. Nicotinic antagonist augmentation of selective serotonin reuptake inhibitor-refractory major depressive disorder: A preliminary study. Journal of Clinical Psychopharmacology. 2008;28:340-344
  130. 130. Furey ML, Drevets WC. Antidepressant efficacy of the antimuscarinic drug scopolamine: A randomized, placebo-controlled clinical trial. Archives of General Psychiatry. 2006;63(10):1121-1129
  131. 131. Lapidus KA, Levitch CF, Perez AM, Brallier JW, Parides MK, Soleimani L, et al. A randomized controlled trial of intranasal ketamine in major depressive disorder. Biological Psychiatry. 2014;76:970-976
  132. 132. Daly EJ, Singh JB, Fedgchin M, Cooper K, Lim P, Shelton RC, et al. Efficacy and safety of intranasal esketamine adjunctive to oral antidepressant therapy in treatment-resistant depression. A randomized clinical trial. JAMA Psychiatry. 2018;75(2):139-148
  133. 133. Cusin C, Dougherty DD. Somatic therapies for treatment-resistant depression: ECT, TMS, VNS, DBS. Biology of Mood & Anxiety Disorders. 2012;2:14
  134. 134. Fu CH, Williams SC, Cleare AJ, et al. Attenuation of the neural response to sad faces in major depression by antidepressant treatment: A prospective, event-related functional magnetic resonance imaging study. Archives of General Psychiatry. 2004;61:877-889
  135. 135. Tansey KE, Guipponi M, Hu X, Domenici E, Lewis G, Malafosse A, et al. Contribution of common genetic variants to antidepressant response. Biological Psychiatry. 2013;73:679-682
  136. 136. Binder EB, Holsboer F. Pharmacogenomics and antidepressant drugs. Annals of Medicine. 2006;38:82-94
  137. 137. de Leon J, Armstrong SC, Cozza KL. Clinical guidelines for psychiatrists for the use of pharmacogenetic testing for CYP450 2D6 and CYP450 2C19. Psychosomatics. 2006;47:75-85
  138. 138. Abaut AY, Chevanne F, Le Corre P. Oral bioavailability and intestinal secretion of amitriptyline: Role of P-glycoprotein? International Journal of Pharmaceutics. 2007;330:121-128
  139. 139. Peters EJ, Slager SL, McGrath PJ, et al. Investigation of serotonin-related genes in antidepressant response. Molecular Psychiatry. 2004;9:879-889
  140. 140. Choi MJ, Kang RH, Ham BJ, et al. Serotonin receptor 2A gene polymorphism (−1438A/G) and short-term treatment response to citalopram. Neuropsychobiology. 2005;52:155-162
  141. 141. McMahon FJ, Buervenich S, Charney D, et al. Variation in the gene encoding the serotonin 2A receptor is associated with outcome of antidepressant treatment. American Journal of Human Genetics. 2006;78:804-814
  142. 142. Calabrese F, van der Doelen RH, Guidotti G, Racagni G, Kozicz T, Homberg JR, et al. Exposure to early life stress regulates Bdnf expression in SERT mutant rats in ananatomically selective fashion. Journal of Neurochemistry. 2015;132:146-154
  143. 143. Tsai SJ, Cheng CY, Yu YW, et al. Association study of a brain-derived neurotrophic-factor genetic polymorphism and major depressive disorders, symptomatology, and antidepressant response. American Journal of Medical Genetics. Part B, Neuropsychiatric Genetics. 2003;123:19-22
  144. 144. Kirchheiner J, Grundemann D, Schomig E. Contribution of allelic variations in transporters to the phenotype of drug response. Journal of Psychopharmacology. 2006;20:27-32
  145. 145. Serretti A, Zanardi R, Cusin C, et al. Tryptophan hydroxylase gene associated with paroxetine antidepressant activity. European Neuropsychopharmacology. 2001;11:375-380
  146. 146. Serretti A, Cusin C, Rossini D, et al. Further evidence of a combined effect of SERTPR and TPH on SSRIs response in mood disorders. American Journal of Medical Genetics. Part B, Neuropsychiatric Genetics. 2004;129:36-40
  147. 147. Reul J, Holsboer F. Corticotrophin-releasing factor receptor 1 and 2 in anxiety and depression. Current Opinion in Pharmacology. 2002;2(1):23-33
  148. 148. Liu Z, Zhu F, Wang G, et al. Association study of corticotropin-releasing hormone receptor1 gene polymorphisms and antidepressant response in major depressive disorders. Neuroscience Letters. 2007;414:155-158
  149. 149. Brouwer JP, Appelhof BC, van Rossum EF, et al. Prediction of treatment response by HPA-axis and glucocorticoid receptor polymorphisms in major depression. Psychoneuroendocrinology. 2006;31:1154-1163
  150. 150. Zanardi R, Benedetti F, Di Bella D, et al. Efficacy of paroxetine in depression is influenced by a functional polymorphism within the promoter of the serotonin transporter gene. Journal of Clinical Psychopharmacology. 2000;20:105-107
  151. 151. Zanardi R, Serretti A, Rossini D, et al. Factors affecting fluvoxamine antidepressant activity: Influence of pindolol and 5-HTTLPR in delusional and nondelusional depression. Biological Psychiatry. 2001;50:323-330
  152. 152. Serretti A, Benedetti F, Zanardi R, Smeraldi E. The influence of Serotonin Transporter Promoter Polymorphism (SERTPR) and other polymorphisms of the serotonin pathway on the efficacy of antidepressant treatments. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2005;29:1074-1084
  153. 153. Kraft JB, Peters EJ, Slager SL, et al. Analysis of association between the serotonin transporter and antidepressant response in a large clinical sample. Biological Psychiatry. 2007;61:734-742
  154. 154. Caspi A, Sugden K, Moffitt TE, Taylor A, Craig IW, Harrington H, et al. Influence of life stress on depression: Moderation by a polymorphism in the 5-HTT gene. Science. 2003;301(5631):386-389
  155. 155. Mancama D, Kerwin RW. Role of pharmacogenomics in individualising treatment with SSRIs. CNS Drugs. 2003;17:143-151
  156. 156. Carvalho LA, Torre JP, Papadopoulos AS, Poon L, Juruena MF, Markopoulou K, et al. Lack of clinical therapeutic benefit of antidepressants is associated overall activation of the inflammatory system. Journal of Affective Disorders. 2013;148:136-140
  157. 157. Gennarelli M, Uher R, Breen G, Farmer A, Aitchison KJ, Craig IW, et al. Candidate genes expression profile associated with antidepressants response in the GENDEP study: Differentiating between baseline ‘predictors’ and longitudinal ‘targets’. Neuropsychopharmacology. 2013;38:377-385
  158. 158. Kiraly DD, Horn SR, Van Dam NT, Costi S, Schwartz J, Kim-Schulze S, et al. Altered peripheral immune profiles in treatment-resistant depression: Response to ketamine and prediction of treatment outcome. Translational Psychiatry. 2017;7:e1065
  159. 159. Abbasi SH, Hosseini F, Modabbernia A, Ashrafi M, Akhondzadeh S. Effect of celecoxib add-on treatment on symptoms and serum IL-6 concentrations in patients with major depressive disorder: Randomized double-blind placebo-controlled study. Journal of Affective Disorders. 2012;141:308-314
  160. 160. Green AI, Schatzberg AF. Obituary. Joseph J. Schildkraut, 1934-2006. Neuropsychopharmacology. 2007;32:1855-1856
  161. 161. Schildkraut JJ. The catecholamine hypothesis of affective disorders: A review of supporting evidence. The American Journal of Psychiatry. 1965;122:509-522
  162. 162. Humble M. Noradrenaline and serotonin reuptake inhibition as clinical principle: A review of antidepressant efficacy. Acta Psychiatrica Scandinavica. 2000;101(Suppl 402):28-36
  163. 163. Ontiveros A. Response to serotonergic and noradrenergic antidepressants: A crossover study of fluoxetine and desipramine in patients with first major depression episode. Gaceta Médica de México. 2017;153(6):188-194
  164. 164. Coplan JD, Gopinath S, Abdallah C. Neurobiological hypothesis of treatment-resistant depression—Mechanisms for selective serotonin reuptake inhibitor non-efficacy. Frontiers in Behavioral Neuroscience. 2014;8:189. DOI: 10.3389/fnbeh.2014.00189. Published online: May 20. 2014. PMCID: PMC4033019. PMID: 24904340A
  165. 165. Thase M, Connolly KR, Roy-Byrne PP, Solomon D. Unipolar Depression in Adults: Treatment of Resistant Depression. Waltham, MA: Wolters Kluwer Health; 2015

Written By

Jose Alfonso Ontiveros

Submitted: 31 August 2018 Reviewed: 15 November 2018 Published: 07 June 2019