Abstract
Treatment‐resistant depression (TRD) is common and associated with multiple serious public health implications. A consensus definition of TRD with demonstrated predictive utility in terms of clinical decision‐making and health outcomes does not currently exist. Instead, a plethora of definitions have been proposed, which vary significantly in their conceptual framework. The absence of a consensus definition hampers precise estimates of the prevalence of TRD, and also belies efforts to identify risk factors, prevention opportunities, and effective interventions. In addition, it results in heterogeneity in clinical practice decision‐making, adversely affecting quality of care. The US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) have adopted the most used definition of TRD (i.e., inadequate response to a minimum of two antidepressants despite adequacy of the treatment trial and adherence to treatment). It is currently estimated that at least 30% of persons with depression meet this definition. A significant percentage of persons with TRD are actually pseudo‐resistant (e.g., due to inadequacy of treatment trials or non‐adherence to treatment). Although multiple sociodemographic, clinical, treatment and contextual factors are known to negatively moderate response in persons with depression, very few factors are regarded as predictive of non‐response across multiple modalities of treatment. Intravenous ketamine and intranasal esketamine (co‐administered with an antidepressant) are established as efficacious in the management of TRD. Some second‐generation antipsychotics (e.g., aripiprazole, brexpiprazole, cariprazine, quetiapine XR) are proven effective as adjunctive treatments to antidepressants in partial responders, but only the olanzapine‐fluoxetine combination has been studied in FDA‐defined TRD. Repetitive transcranial magnetic stimulation (TMS) is established as effective and FDA‐approved for individuals with TRD, with accelerated theta‐burst TMS also recently showing efficacy. Electroconvulsive therapy is regarded as an effective acute and maintenance intervention in TRD, with preliminary evidence suggesting non‐inferiority to acute intravenous ketamine. Evidence for extending antidepressant trial, medication switching and combining antidepressants is mixed. Manual‐based psychotherapies are not established as efficacious on their own in TRD, but offer significant symptomatic relief when added to conventional antidepressants. Digital therapeutics are under study and represent a potential future clinical vista in this population.
Keywords: Depression, treatment‐resistant depression, difficult‐to‐treat depression, ketamine, esketamine, second‐generation antipsychotics, neurostimulation, electroconvulsive therapy, precision medicine, personalized medicine, patient‐reported outcomes
It is amply documented that major depressive disorder (MDD) is highly prevalent and associated with substantial burden and economic costs 1 , 2 , 3 , 4 , 5 . According to the World Health Organization (WHO), MDD is the single largest contributor to loss of healthy life, and this contribution has apparently further increased during the COVID‐19 pandemic 6 , 7 , 8 .
Notwithstanding the evidence supporting the efficacy of conventional antidepressants as well as manual‐based psychotherapies and specific neurostimulation modalities, the majority of individuals with MDD are inadequately responsive to first‐line treatments. Moreover, a substantial proportion of them fail multiple antidepressant interventions, resulting in what is described as treatment‐resistant depression (TRD) 5 , 9 , 10 , 11 , 12 , 13 , 14 , 15 , 16 .
Although non‐response is a common outcome of treatment with multiple conventional antidepressants, a consensus definition of TRD with predictive utility does not currently exist. Instead, a host of definitions have been proposed, differing in their conceptual framework, operational criteria and working assumptions. This heterogeneity of definitions has resulted in a wide range of estimates of the prevalence of TRD 16 . The proportion of people with TRD would be expected to be higher when multidimensional definitions are used, especially those including patient‐reported outcomes 17 , 18 .
There are multiple serious public health implications associated with TRD, which provide the impetus for a specific focus on its detection and algorithmic management. First, TRD is common in the general population: based on international epidemiological estimates, it is extrapolated that more than 100 million people globally meet one or more definitions of this condition 19 . In addition, cost of illness studies have documented staggering direct and indirect economic costs associated with MDD, of which more than half globally are attributable to TRD 20 .
The relatively higher cost of illness attributed to TRD is directly due to higher health care utilization and the need for higher intensity treatments 20 , 21 , 22 , 23 . Higher indirect costs are also reported in TRD as a consequence of relatively greater impairment in psychosocial function, greater need for disability benefits, higher workplace disability and absenteeism, as well as the negative impact on carers 10 , 21 , 24 , 25 , 26 , 27 , 28 , 29 , 30 , 31 , 32 , 33 , 34 , 35 . Moreover, the rate of suicidality, including completed suicide, is disproportionately higher in TRD populations 36 .
Additional public health implications of TRD relate to the established association between MDD and multiple common and chronic non‐communicable physical diseases 37 , 38 , 39 . For example, it is established that MDD is a risk factor for cardiovascular disease, obesity and type 2 diabetes mellitus, and this is especially apparent in individuals with more severe and/or persistent depressive syndromes, which are over‐represented in TRD populations 40 , 41 .
Notwithstanding the foregoing public health implications of TRD, relatively few interventions have been established as efficacious for persons having multiple failed trials with conventional antidepressants. Instead, the emphasis of treatment development in depressive disorders has been on non‐TRD populations. In addition, prevention of TRD is not a national health policy priority in any country worldwide, nor is progress in its management a quality outcome measure in any national public health care system.
Currently, more than 90 clinical practice guidelines are available that aim to provide decision support to clinicians caring for adults with mood disorders, originating from 83 countries and published in 27 languages 42 . Most of them have been produced in high‐income countries and integrate scientific evidence with expert opinion 42 , 43 , 44 , 45 . Major limitations of extant guidelines, as it specifically relates to TRD, are that they do not adopt a consensus definition of this condition, and are not consistent in their selection or sequencing of recommendations.
In addition, extant guidelines vary in how they define an adequate antidepressant regimen and frequently conflate the treatment of TRD with non‐TRD populations (i.e., partial responders to antidepressants). For example, second‐generation antipsychotics (SGAs), of which most have not been proven to be effective in TRD, are often recommended for this condition in combination with antidepressants, despite their evidentiary base comprised largely of populations defined as partial responders to antidepressants.
Herein, we aim to provide a synthesis of current definitions of TRD, with an emphasis on their limitations, and recommendations for the development of an improved consensus definition; to summarize best estimates of the prevalence of TRD on the basis of current definitions; to review the available evidence on risk factors for TRD; to provide recommendations concerning the detection and management of TRD, based on research evidence when available and opinions from international experts; and to review investigational interventions for TRD. We do not intend to review and/or supplant existing recommendations for depression which is not treatment‐resistant 44 , 45 , 46 , 47 , 48 .
DEFINITIONS OF TREATMENT‐RESISTANT DEPRESSION
The absence of a consensus and validated definition of TRD is a major limitation from the viewpoints of translational research, treatment development, as well as clinical and policy decision‐making. Indeed, the pathway towards more targeted treatments in psychiatry requires a more precise delineation of the phenotype being evaluated 49 , 50 , 51 .
The lack of a consensus definition results in the heterogeneity of populations enrolled in clinical trials evaluating new interventions for TRD, greatly limiting the interpretability and generalizability of the results. At a clinical level, the heterogeneity of patient samples contributes to differences in recommendations on the sequencing of treatments for people not responding to conventional first‐line antidepressants. Disparity in practice behavior is likely compromising optimal health outcomes amongst those living with and receiving interventions for TRD. Moreover, from a policy perspective, reimbursement and access to treatment for populations with TRD will understandably vary in the absence of a universal definition, further compromising real‐world outcomes in these patients.
The definition of TRD adopted by the US Food and Drug Administration (FDA) 52 and the European Medicines Agency (EMA) 53 is failure to respond to two or more antidepressant regimens despite adequate dose and duration and adherence to treatment. These regulatory agencies recognize the lack of precision of this definition and its overlap with definitions of “partial response” to antidepressant treatment 53 . The EMA definition, contrary to the FDA one, explicitly states that the failed antidepressants can be from the same or different mechanistic classes. Limitations of the FDA and EMA definitions are that they do not explicitly operationalize non‐response, and do not consider psychotherapeutic interventions, regarded as first‐line treatments for mild or moderate depression by most guidelines 48 .
Other definitions of TRD have tried to overcome one or more of the above drawbacks (see Table 1, 2). A commonly cited framework for the definition of inadequate response to antidepressants is the Thase and Rush staging model 54 , 55 . This model does not define TRD categorically, but instead operationalizes and tacitly implies TRD along a continuum of failed antidepressant trials. Stage I is defined by failure of at least one adequate trial of one major class of antidepressants; stage II by failure of at least two adequate trials of at least two distinctly different classes of antidepressants; stage III by stage II resistance plus failure of an adequate trial of a tricyclic antidepressant (TCA); stage IV by stage III resistance plus failure of an adequate trial of a monoamine oxidase inhibitor (MAOI); and stage V by stage IV resistance plus failure of a course of bilateral electroconvulsive therapy (ECT). In the text of the reference paper, it is made clear that the first trial should be a 4‐week one with a selective serotonin reuptake inhibitor (SSRI) in moderate dosages 54 .
Table 1.
Definitions of treatment‐resistant depression (TRD)
FDA | EMA | Thase & Rush | Maudsley Model | GSRD | DM‐TRD | MGH‐S | |
---|---|---|---|---|---|---|---|
Categorical definition | + | + | – | + | + | + | – |
Number of requested treatment failures | 2 | 2 | 1 | 1 | 2 | 1 | 1 |
Operationalization of “failure” of treatment | – | – | – | – | + | – | – |
Indication that failed antidepressants must be of different classes | – | – | + | – | + | – | – |
Indication of required duration of failed treatments | + | + | + | + | + | + | + |
Implication of a hierarchy of efficacy of antidepressants | – | + | + | – | – | – | – |
Failure of psychotherapies included | – | – | – | – | – | + | – |
Failure of ECT included | – | – | + | + | – | + | + |
Failure of augmentation/combination treatments included | – | – | – | + | – | + | + |
Patient‐reported outcomes considered | – | – | – | – | – | – | – |
Baseline severity included | – | – | – | + | + | + | – |
Duration of current episode included | – | – | – | + | + | + | – |
Baseline psychosocial impairment included | – | – | – | – | – | + | – |
Presence of comorbidities included | – | – | – | – | – | + | + |
Comorbid anxiety symptoms included | – | – | – | – | – | + | + |
Comorbid personality disorder included | – | – | – | – | – | + | + |
Quality of life included | – | – | – | – | – | – | – |
History of psychosocial stressors included | – | – | – | – | – | + | – |
History of childhood adversity included | – | – | – | – | – | – | – |
FDA – US Food and Drug Administration, EMA – European Medicines Agency, GSRD – European Group for the Study of Resistant Depression, DM‐TRD – Dutch Measure for quantification of Treatment Resistant Depression, MGH‐S – Massachusetts General Hospital Staging, ECT – electroconvulsive therapy
Table 2.
Options for management of treatment‐resistant depression (TRD)
Option | Rationale | Limitations |
---|---|---|
Extending antidepressant trial | Delayed time to response amongst subpopulations with TRD. |
Modest evidence base supporting the strategy. Unlikely to be acceptable to most patients living with TRD. Alternative strategies for TRD better established (e.g., ECT, esketamine). |
Switching antidepressants |
Mechanistically dissimilar antidepressants from different classes may offer improved health outcomes in TRD in some cases. Especially appropriate when index antidepressant class is poorly tolerated. |
Modest evidence base supporting the strategy. Newly initiated antidepressant will require at least 4 weeks before outcome can be assessed. |
Combining antidepressants |
May target symptoms not responding to index antidepressant (e.g., fatigue, cognitive impairment, sleep problems). May improve tolerability via antidote of emergent adverse events (e.g., bupropion for antidepressant‐induced sexual dysfunction). |
Limited evidence base in TRD. Potential for drug‐drug interactions. Decreased adherence with polypharmacy regimens. Greater cost of treatment. |
Ketamine |
Acute efficacy established in TRD. Beneficial effects on suicidality. Rapid onset of symptomatic improvement. |
Insufficient long‐term efficacy, tolerability and safety data. Access to treatment limited in many jurisdictions. Specialized personnel required for safe administration. Long‐term safety profile in TRD not established (e.g., abuse liability, gateway activity). |
Esketamine |
Acute and maintenance efficacy established in TRD. Beneficial effects on suicidality. Rapid onset of symptomatic improvement. Superiority to SGA (i.e., quetiapine XR) in acute and maintenance treatment of TRD. |
Access to treatment limited in many jurisdictions. Acquisition cost. Recommendation to co‐prescribe with underlying antidepressant in TRD. |
Second‐generation antipsychotics (SGAs) |
Scalable and accessible treatments. Evidence established for olanzapine‐fluoxetine combination. |
With exception of olanzapine‐fluoxetine combination, studied in partial responders rather than TRD. Short‐ and long‐term tolerability concerns. |
Electroconvulsive therapy (ECT) |
Highly effective in acute and maintenance treatment of TRD. Non‐inferiority to IV ketamine suggested by available evidence. Efficacy in TRD across the age span. |
Relative lack of availability in many contexts. Stigma and lack of acceptability to many patients with TRD. Tolerability concerns (e.g., memory deficits). |
Repetitive transcranial magnetic stimulation |
Shown to be effective in TRD. More acceptable to patients than ECT. Accelerated protocol demonstrates significant remission rates within one week. Tolerability advantages compared to ECT (i.e., persisting cognitive deficits not observed). |
Relative lack of availability in many jurisdictions. Inferiority to ECT in TRD with non‐accelerated protocols. Insufficient long‐term data in TRD. |
Vagus nerve stimulation |
Proven efficacy in TRD in persons with extensive antidepressant failure histories. Treatment does not need to be administered on a daily basis. |
Not available in most countries globally. Complexity of procedure limits scalability. Complications of implant. Cost of treatment. |
Psychotherapies |
Evidence supports efficacy when used adjunctively in TRD. Opportunity to target comorbidities. Facilitate coping strategies with improved effects on patient‐reported outcomes. Highly acceptable to persons with lived experience of TRD. Opportunity to tailor treatment targeting specific therapeutic outcomes. |
Lack of availability of treatment or adequately trained providers. Low adherence to therapy. Lack of evidence as standalone treatment in TRD. |
Strengths of the Thase and Rush model are its simplicity, pragmatism, and close proximity to behavior in everyday clinical practice. In addition, this model prioritizes treatments that are better tolerated, which is in line with clinical practice guidelines and treatment algorithms. A first limitation of the model is that “failure” of treatment trials is not operationalized. Furthermore, the model reflects some non‐validated assumptions: for instance that, in a patient initially not responding to an SSRI, a non‐classmate antidepressant is more likely to be efficacious as a next‐step treatment strategy; or that MAOI exposure should be limited to populations with treatment resistance. In addition, there is no explicit consideration of depression features such as duration and severity of the index episode, and no mention of psychotherapeutic interventions. Finally, although augmentation or combination strategies are mentioned in the text of the reference paper 54 , they are not explicitly included in the staging model.
The Maudsley Staging Model (MSM) was developed to improve upon the limitations of the Thase and Rush model 56 . It defines treatment resistance as failure to attain significant level of improvement (i.e., clinical remission) from an accurately diagnosed depressive episode following treatment with an antidepressant given at an adequate dose for a minimum of six weeks. Three dimensions of resistance are included: treatment failure, duration of the depressive episode, and severity of depression 56 .
A maximum of seven points can be assigned for the treatment dimension: one point for failure on 1‐2 medications; two points for failure on 3‐4 medications; three points for failure on 5‐6 medications; four points for failure on 7‐10 medications; five points for failure on more than 10 medications. One further point is assigned if augmentation treatment has failed, and one further point if ECT has not been effective. A maximum of three points can be assigned for the duration of the depressive episode: one if the episode is acute (up to 12 months); two if it is subacute (from 13 to 24 months); three if it is chronic (more than 24 months). A maximum of five points can be assigned for the severity of depression: one if it is subsyndromal; two if it is mild; three if it is moderate; four if it is severe without psychosis; and five if it is severe with psychosis. The overall staging of TRD is defined as mild (total score between 3 and 6), moderate (total score between 7 and 10) or severe (total score between 11 and 15).
Thus, in the MSM, resistance is assessed on the basis not only of treatment but also of illness variables, which has been reported to be useful in predicting short‐ and intermediate‐term outcomes in TRD populations 57 , 58 . Overall, the threshold for the definition of TRD is low, requiring failure of just one adequate treatment. Failure of treatment is not operationalized, although a discussion of the complexity of defining clinical remission is provided in the text of the main paper presenting the model 56 . The assignment of scorings is in some respects arbitrary: for instance, a differential weighting is assigned to populations who fail at least five vs. less than five treatments, in the absence of validation. Failure of manual‐based psychotherapies is not considered.
The European Group for the Study of Resistant Depression (GSRD) 14 separately defined non‐response (failure to respond to one trial of 6‐8 week duration of any antidepressant treatment); TRD (failure to respond to two or more adequate trials of different classes of antidepressants, with five different levels of resistance depending on the overall duration of trials); and chronic resistant depression (failure to respond to several antidepressant trials, including augmentation strategies, of the overall duration of at least 12 months) 14 .
Strengths of the GSRD staging method are the explicit definition of treatment non‐response as a reduction of less than 50% in the total score on the Hamilton Depression Rating Scale (HAM‐D) 59 or the Montgomery‐Åsberg Depression Rating Scale (MADRS) 60 , and the lack of any implicit hierarchy of efficacy of antidepressants. Limitations are the lack of validation of any of the provided time‐based subcategories, including the definition of chronic depression based on a duration of at least one year, which is considerably briefer than what is generally accepted (i.e., longer than two years).
The Dutch Measure for quantification of Treatment Resistant Depression Model (DM‐TRD) was developed to improve upon the point system proposed in the MSM 61 . To the variables considered in that system, this model adds functional impairment (with a score from 0, no impairment, to 3, severe impairment); comorbid anxiety symptoms (with a score from 0, not present, to 1, fulfilling criteria for at least one DSM‐IV anxiety disorder); comorbid personality disorder (with a score from 0, not present, to 1, present based on formal interview); psychosocial stressors (with a score of 0, no psychosocial stressor, or 1, at least one psychosocial stressor); several categories of augmentation/combination regimens (with a score from 0, not used, to 3, five or six medications); use of psychotherapy (with a score from 0, not used, to 2, at least two empirically supported psychotherapies); and intensified treatment (with a score from 0, not used, to 2, inpatient treatment). The maximum total score becomes 27.
This model is the most comprehensive in terms of variables included, although physical comorbidities and childhood adversities are not considered. As in the MSM, the threshold for the definition of TRD is low, requiring failure of just one adequate treatment, and non‐response is not operationalized. The predictive validity of the model has been supported to some extent 61 .
The Massachusetts General Hospital Staging Model (MGH‐S) definition of TRD integrates the number of failed trials with the intensity/optimization of each trial, without assumptions on the hierarchy of antidepressant classes 62 . One point is assigned for non‐response to each adequate trial of a marketed antidepressant (duration of at least six weeks and adequate dosage). Half a point is assigned for each trial based on optimization of dose, optimization of duration, or an augmentation/combination strategy. Three points are assigned for non‐ response to ECT.
Limitations of the MGH‐S include the lack of operationalization of “failure” of trials; the arbitrary scores attributed to treatments; the fact that optimization of dose or duration of treatment is weighted equally as augmentation/combination strategies (which is not empirically supported); and the assignment of one point for each failed antidepressant, which may generate a very high total score 63 .
None of the extant TRD definitions are universally accepted and/or implemented at point‐of‐care in clinical practice 11 , 32 , 64 , 65 , 66 , 67 , 68 . In addition, no existing TRD definition is supported by an external validator and/or biomarker. Most TRD definitions do not explicitly consider failure of manual‐based psychotherapies in their hierarchical characterization of treatment resistance. As psychotherapeutic interventions are recommended as first‐line treatments in persons presenting with depression of mild or moderate severity, any working definition of TRD with clinical utility will need to explicitly include non‐response to these interventions.
Also, common across most definitions of TRD is the absence of a quantifiable and consensus endpoint defining response versus non‐response to antidepressants. An additional limitation is that the definition of outcome is based on a clinician assessment, while patient‐reported outcomes are not considered. Indeed, even amongst patients classified as “responders”, many continue to manifest debilitating residual symptoms 69 , 70 . This was highlighted in the STAR*D trial, in which it was observed that only 10% of persons “in remission” were fully asymptomatic 71 . If, for example, a person is classified as “responder” to treatment but continues to experience cognitive deficits that are impairing, it would be incorrect to consider this an adequate antidepressant response 72 .
None of the extant definitions of TRD includes reference to quality of life. This is a major limitation, given the importance assigned to this variable by persons with lived experience 73 . The predictive utility of quality of life as a critical outcome measure when defining TRD is underscored by the observation that persons remitting with antidepressants who continue to report decreased quality of life are at greater risk of relapse and recurrence 74 , 75 .
Further drawbacks of existing TRD definitions are that they fail to take into consideration the social, economic, anamnestic (e.g., adverse childhood experiences) and interpersonal factors which, alone or in combinations, are known to moderate antidepressant response 1 , 44 , 47 , 71 , 75 , 76 , 77 , 78 , 79 , 80 , 81 . Furthermore, an unintended consequence of a TRD framework that is hierarchical is encouraging multiple unproven treatment strategies, with polypharmacy and the possibility of associated safety and tolerability concerns 70 , 75 .
Moreover, results of a recent analysis in the WHO World Mental Health Surveys underscores that persistence with next‐step treatments is uncommon in persons with MDD 82 . Also, in those who do switch to next‐step treatments, a considerable treatment delay (i.e., 6‐9 months) elapses before switching occurs 82 , 83 .
An example of a patient‐centric framework describing persons with multiple antidepressant failures is the construct of difficult‐to‐treat depression (DTD) 84 . This construct relies on a biopsychosocial approach when considering causal, perpetuating and treatment factors of poor outcomes in depression 70 . The therapeutic emphasis in DTD pivots away from symptomatic remission towards symptomatic control, functional recovery and quality of life improvement as part of chronic disease management 70 .
For several patients, despite non‐remission status, more modest improvement in overall depressive symptom severity may result in significant self‐assessed improvement in well‐being 85 , 86 , 87 . For example, an approximate 35% improvement from baseline in total MADRS score may be associated with significant improvement of quality of life in persons with TRD 87 . These data support the notion that more modest improvements in symptom severity in persons with TRD may be clinically meaningful, and invite the need for multidimensional definitions that are not solely dependent on threshold symptomatic improvement 86 , 88 , 89 .
Surveys of persons with lived depression experience have highlighted the importance of dimensional symptomatic outcomes in addition to categorical ones 90 , 91 . For example, alleviation of emotional blunting, anhedonia, anxiety and rumination are often prioritized by persons living with depression over full symptomatic remission 92 . Shared‐decision making, patient‐centered care focusing on specific symptoms of concern, and integrating treatment modalities become paramount in DTD, in keeping with the guiding principles of chronic disease management 84 , 93 , 94 , 95 , 96 . Although DTD is not currently recognized by regulators as a pathway for treatment approval and marketing authorization, it more closely approximates real‐world presentations and outcomes among persons with TRD, and could serve as a clinical heuristic or even a framework informing the further characterization of TRD.
Overall, there is a confluence of research, clinical, policy, and public health reasons to have a validated and universal TRD definition. Existing definitions would be best characterized as frameworks that vary in their constituent variables and working assumptions. The existing TRD frameworks reviewed herein have not provided any substantive insight into the pathogenesis, treatment discovery and development, or clinical care of persons with TRD.
Moreover, there is no compelling evidence that any of the foregoing TRD frameworks have been implemented at large scale by the clinical or research community. A consensus definition of TRD at the very least will need to provide a quantifiable endpoint defining response, integrate manual‐based psychotherapies, empirically validate assumptions surrounding differential treatment weighting, and integrate multiple factors known to influence antidepressant response. A TRD definition that is consistent across disparate clinical care ecosystems, and fulfills both research and clinical needs, is badly needed.
PREVALENCE OF TREATMENT‐RESISTANT DEPRESSION
Differences in the definition of TRD have resulted in highly variable estimates of its prevalence rate 99 . TRD is often stated to affect approximately 30% of persons receiving antidepressant treatment in research settings, while its prevalence in real world practice is estimated to range between 6 and 55% 32 , 98 , 99 , 100 , 101 .
Most individuals with MDD access mental health care initially through the primary care system, where measurement‐based care is rarely implemented 102 , 103 , 104 . A tentative estimate of the prevalence of TRD in primary care can be made only indirectly by using a “depression treatment cascade” approach 105 . Approximately 10‐15% of patients in primary care present with clinically significant depressive symptoms, and only about half of these cases are diagnosed, of which an estimated 25% are prescribed an antidepressant 106 . Replicated evidence indicates that, of those prescribed antidepressants, the majority discontinue treatment prematurely. Hence, only about 5‐7% of persons with depression treated in primary care settings would be expected to achieve remission 106 . The foregoing cascade approach – which integrates aspects of misdiagnosis, non‐adherence, inadequate treatment trials, as well as implementation gaps – underscores the high prevalence of poor outcomes of depression in primary care, of which a significant percentage would be expected to meet criteria for TRD 10 , 30 , 64 , 71 , 107 , 108 .
A more precise estimate of the prevalence of TRD can be done by referring to the Sequenced Treatment Alternatives to Relieve Depression (STAR*D) trial, a National Institute of Mental Health (NIMH)‐sponsored multisite study (18 primary care and 23 psychiatric care settings) carried out in the US 33 . All eligible subjects enrolled in the STAR*D trial initiated treatment with citalopram. After a 12‐week trial (level 1 treatment), those persons not in remission were randomly assigned to one of seven switch/combination approaches (level 2). Non‐response to a switch/combination level 2 treatment resulted in randomization to further treatments (levels 3 and 4). The FDA and EMA definitions of TRD would align with failure to level 1 and 2 treatments in the STAR*D trial. On this basis, it can be estimated that approximately 55% of persons with MDD would meet the FDA/EMA criteria for TRD (i.e., inadequate response to two or more antidepressants despite adequate treatment intensity and duration) 33 .
In summary, while it is often stated that TRD is affecting approximately 30% of persons receiving antidepressant treatment, a more stringent and multidimensional definition of this condition emphasizing symptomatic remission increases this estimate to about 55%.
RISK FACTORS FOR TREATMENT‐RESISTANT DEPRESSION
Many factors have been identified as being associated with reduced antidepressant response, but relatively few are established as risk factors specifically for TRD. In addition, most factors identified as negatively affecting antidepressant outcomes are reported in small studies and are described with a particular antidepressant intervention. Amongst the relatively few studies that have sought to identify factors associated with TRD, most are limited by the inconsistent definition of this condition, and primarily evaluate outcomes with monoamine‐based antidepressants.
Herein, we endeavour to identify factors that are associated with TRD. As most studies have evaluated factors associated with reduced response to conventional antidepressants rather than TRD, we provide clarity and attempt to separate these two aspects.
Sociodemographic factors
It is established that older persons more frequently fail multiple monoamine‐based antidepressant treatments, which may be taken as evidence that TRD is more common in this subpopulation 109 , 110 . However, there is no evidence of an attenuated response in older adults with depression receiving manual‐based psychotherapeutic treatments 111 , and the efficacy of ECT does not seem to be reduced as a function of age 112 . It is also reported that repetitive transcranial magnetic stimulation (rTMS) may have similar (or potentially greater with increased pulse dose) efficacy in older adults with MDD 113 .
It is not established whether female sex is a risk factor for TRD 114 . Whether depression during reproductive life events (e.g., peripartum onset depression) is more likely to be treatment‐resistant is also not sufficiently established 115 . It is, however, well known that females are affected by depression at twice the rate of males, and are more likely to be prescribed antidepressants 116 . Consequently, females would be expected to represent the majority within a TRD population, although it remains uncertain whether their relative risk is higher.
Socioeconomic position is a risk factor for TRD in persons receiving monoamine‐based antidepressants. For example, in the STAR*D trial, persons meeting level 2 criteria (i.e., inadequate response to two sequential antidepressant regimens) were more likely to report lower income and dependence on the public health system 117 . In addition, persons of lower educational attainment or unemployed are found to be more often resistant to multiple sequential antidepressant strategies 17 , 118 .
Future research should evaluate whether racial and/or ethnic factors contribute to the occurrence of TRD, and also endeavour to explore whether sexual orientation and/or gender identity, marital status, interpersonal connectedness, and measures of loneliness are risk factors for TRD.
Adverse experiences and trauma
It is well established that childhood maltreatment is associated with greater severity of depression, earlier age at onset, cognitive dysfunction, presence of psychotic symptoms, and physical/psychiatric comorbidities, each of which is also associated with attenuated response to antidepressants and manual‐based psychological interventions 119 , 120 , 121 , 122 , 123 .
There are also studies providing evidence that a reported history of childhood emotional abuse is associated with recurrent depression, persistent depression, as well as treatment resistance to antidepressants 124 . The international Study to Predict Optimized Treatment for Depression (iSPOT‐D) reported that, amongst adults with MDD and a history of trauma between the ages of 4 and 7 years, only 15.9% achieved remission after 8 weeks of treatment with escitalopram, sertraline or venlafaxine, compared to 84.1% in individuals with no history of childhood trauma 125 .
The attenuated response to antidepressants in persons with a history of childhood maltreatment may not, however, occur with all antidepressants. For example, preliminary evidence suggests that response to vortioxetine or ketamine treatment in depression is not reduced in persons with trauma, suggesting different outcomes as a function of the putative mechanism of action of medications 126 , 127 .
More in general, life stress events have been directly associated with a poorer response to commonly prescribed antidepressants, as well as with a greater occurrence of suicidal behavior and comorbidities and a greater severity of symptoms, which are variables that could mediate the association with an attenuated response to antidepressants and possibly to TRD 128 .
Clinical factors
Greater baseline severity is a highly replicated risk factor for TRD, and is indeed included in some frameworks as a variable in the hierarchical characterization of the condition. Illness duration is also highly associated with TRD, with replicated evidence indicating that the length of a depressive episode is inversely proportional to the probability of treatment response 129 .
Evidence also suggests that some phenomenological characteristics of depression may be associated with treatment resistance. Psychotic symptoms affect approximately 20% of adults with MDD and are highly associated with TRD 130 . Mixed features are reported to be present in approximately 25% of persons with MDD and are associated with attenuated antidepressant response, although it remains to be determined whether they are a risk factor specifically for TRD 47 , 131 .
Anhedonia is a core component of depression endorsed by 35‐75% of patients, and may be a risk factor for TRD in persons whose treatment history is delimited to SSRIs 132 , 133 . Cognitive deficits in MDD are prevalent, persistent, and often progressively increase as a function of illness severity and duration; they are associated with attenuated response to select antidepressants, and may represent a risk factor for TRD 72 , 134 , 135 , 136 .
Anxiety symptoms are frequently reported in TRD populations, and their presence in MDD is associated with a more severe illness presentation, lower probability of remission, comorbidities and suicidality 137 , 138 , 139 , 140 . Results from the STAR*D trial indicate that persons presenting with anxious depression exhibit attenuated antidepressant response and are more likely to develop TRD 141 . The GSRD study also reported that anxiety disorders were over‐represented in persons meeting criteria for TRD 142 .
It is well established that TRD populations have a higher rate of psychiatric and physical comorbidities as compared to non‐TRD populations 143 . In addition, TRD is a risk factor for incident physical comorbidities, such as cardiovascular disease, type 2 diabetes mellitus, osteoporosis, and metabolic syndrome 40 , 144 , 145 , 146 . Evidence indicates that the foregoing physical diseases are in their turn risk factors for TRD 145 , 147 , 148 , 149 , 150 , 151 , 152 .
DETECTION OF TREATMENT‐RESISTANT DEPRESSION
The assessment of an individual with MDD towards personalization of treatment selection and sequencing has been previously reviewed in this journal 13 . Herein, we specifically focus on the assessment process aimed to confirm that TRD is present, and to rule out the possibility of pseudo‐resistance.
Reviewed herein are the most common modifiable contributors to pseudo‐resistance, including inaccuracy of the MDD diagnosis, inadequacy of current and past treatment trials, inaccurate assessment of response, and individual differences in the metabolism of antidepressants 153 , 154 .
Accurate diagnosis of MDD
Inaccuracy of the MDD diagnosis is a common reason for pseudo‐resistance. It is estimated that approximately half of individuals with MDD are not correctly diagnosed 155 . A not uncommon scenario in clinical practice is the depressed patient presenting with resistance to multiple sequential antidepressants whose correct diagnosis should be bipolar disorder instead of MDD 156 .
For most individuals with bipolar disorder, depression is the index presentation, which warrants reconsideration of the MDD diagnosis in any person presenting with TRD. Indeed, it is reported that individuals prescribed multiple failed antidepressant trials (i.e., TRD) have a much greater likelihood of an underlying diagnosis of bipolar disorder as compared to persons prescribed a single antidepressant trial 157 . Furthermore, it is reported that the transition from a diagnosis of MDD to one of bipolar disorder occurs at a rate of approximately 1‐3% per year, indicating that diagnostic assessment must be reconsidered in all TRD presentations 130 , 158 , 159 .
Multiple screening tools for bipolar disorder have been validated, including the Rapid Mood Screener (RMS) 160 , the Patient Mania Questionnaire (PMQ) 161 , the Mood Disorder Questionnaire (MDQ) 162 , and the Hypomania Checklist‐32 163 . Although screening tools are not sufficient to diagnose bipolar disorder, they can be used routinely in clinical practice and, if positive, warrant a more comprehensive assessment of the possible presence of bipolar disorder.
In addition to screening for bipolar disorder, relevant comorbid conditions should be diagnosed and managed if present. They include substance and alcohol use disorders, anxiety disorders, personality disorders, and some physical diseases such as hypothyroidism.
Determining the adequacy of treatment trials
The adequacy of an antidepressant treatment refers to the choice of medication, its dose, the duration of treatment, and the patient's adherence. A comprehensive and precise characterization of current and past medication regimens is required in order to confirm the presence of TRD, and can be captured by several instruments.
The Antidepressant Treatment History Form (ATHF) is a data capture instrument suitable for implementation at point‐of‐care. It was originally developed in studies of ECT and has subsequently undergone a broader clinical and research application 164 . It has explicit criteria for evaluating response to pharmacological and neurostimulation treatments, and is also available in a shorter version (the ATHF‐Short Form, ATHF‐SF) 165 . Other instruments that capture and record current and prior antidepressant regimens are the self‐rated Massachusetts General Hospital Antidepressant Treatment Response Questionnaire (ATRQ) 166 and the Maudsley Treatment Inventory 56 .
First of all, the appropriateness of the antidepressant regimen needs to be confirmed. It is well established that a knowledge‐implementation gap exists between what are proven treatment strategies in MDD and what are actually implemented 42 . The adequacy of the dose of the medication has then to be considered: dosing recommendations are established for all approved antidepressants and are described in their respective product monographs.
The adequate duration of an antidepressant trial is generally considered to be 4‐6 weeks at optimal dosing, although 60% of persons who achieved remission in the STAR*D trial with level 1 treatment did so after week 6 of treatment, indicating that a subpopulation of adults with MDD may require longer treatment trials 167 , 168 .
Adherence to treatment has also to be assessed. A replicated observation is the high rate of non‐adherence to antidepressants in persons with MDD. Persons with less than 80% adherence to antidepressant regimen recommendations are commonly defined as non‐adherent 169 . Using this definition, about 30‐50% of persons prescribed with antidepressants are non‐adherent in acute phase treatment 169 . Assessing adherence to therapy includes pill counts and patient self‐report. Digital sensor systems have been used in academic studies to document adherence, but are not readily available for clinical implementation.
Assessing outcome of previous antidepressant trials
Defining TRD implies quantification of therapeutic outcome with previous antidepressant treatments. However, as already stated, most definitions of TRD do not provide a quantifiable and consensus endpoint defining response versus non‐response to antidepressants. An exception is the GSRD staging method 14 , which explicitly defines treatment non‐response as a reduction of less than 50% in the total score on the HAM‐D or the MADRS. This may represent a useful reference in ordinary clinical practice.
However, it is noticed that, in some patients, a reduction of total MADRS score of about 35% may be associated with significant improvement of quality of life 87 , supporting the need for multidimensional definitions that are not solely dependent on threshold symptomatic improvement 86 , 88 , 89 . The use of measures such as the World Health Organization‐Five Well‐Being Index (WHO‐5) may be suggested for this purpose 170 . More in general, therapeutic endpoints that integrate patient‐reported outcomes along with symptomatic measures may provide a more precise characterization of response to treatment 82 .
Although “failure” of one or more antidepressant trials is an integral part of all definitions of TRD, it must be acknowledged that there is no consensus in the field about how this “failure” should be defined and ascertained. Overcoming this major limitation is an obvious priority for future research on TRD.
Pharmacogenomic testing and evaluating antidepressant blood levels
Evidence indicates that a subset of MDD patients presenting with TRD may exhibit a failed antidepressant response as a consequence of a suboptimal bioavailability of the administered antidepressant, due to rapid metabolizer status 171 , 172 , 173 , 174 . Available evidence indicates that allelic variations of cytochromes P450‐2D6 (CYP2D6) and P450‐2C19 (CYP2C19) are especially associated with antidepressant outcome. In particular, CYP2D6 phenotypes may be important in some patients taking TCAs and venlafaxine, and CYP2C19 phenotypes in some individuals receiving TCAs, citalopram, escitalopram and sertraline 171 . Although pharmacogenetic testing cannot be recommended as a routine assessment in TRD, some preliminary evidence does suggest that, in select circumstances, it may be warranted.
Furthermore, blood levels should be monitored in non‐responding persons receiving some TCAs (i.e., imipramine, desipramine, nortriptyline), as therapeutic levels/windows have been established for these agents 175 , 176 , 177 .
MANAGEMENT OF TREATMENT‐RESISTANT DEPRESSION
Herein, we review tactics which can be considered for managing TRD once the presence of this condition is confirmed. These tactics include extending the current antidepressant trial, switching antidepressants, combining antidepressants, use of esketamine/ketamine, and neurostimulation (see Table 2).
Although manual‐based psychotherapies are not proven to be efficacious as a standalone intervention in TRD, their efficacy in combination with antidepressants is briefly reviewed. Also, we briefly review the evidence for other strategies (e.g., lithium, thyroid hormone) that are better established in patients with partial response to TCAs and MAOIs rather than principally studied in TRD.
We also review data for SGAs, despite the fact that – with the exception of the olanzapine‐fluoxetine combination – these medications are not approved for TRD, but only for individuals with MDD exhibiting partial response to an index antidepressant.
Extending the antidepressant trial
As mentioned earlier, results from the STAR*D trial indicated that a proportion of individuals who responded to level 1 treatment did so after week 6. A systematic review of available studies sought to evaluate the likelihood of response during weeks 5‐8 and 9‐12 in individuals with MDD not responding after four weeks 178 . It was concluded that approximately 20% of patients with MDD not responding in the first four weeks responded during weeks 5‐8, while approximately 10% responded during weeks 9‐12 178 .
However, it is not established that extending an antidepressant trial in patients defined as having TRD results in any considerable likelihood of treatment success. In addition, persons with lived depression experience prioritize rapidity of antidepressant action, so that prolonging antidepressant trials for an additional one to two months is unlikely to be acceptable in most cases of TRD 92 .
Switching antidepressants
Meta‐analytic data are conflicting as to whether switching antidepressants increases the likelihood of response in TRD 179 , 180 . A related but separate concept that would justify switching class of antidepressants is that of “broadening the spectrum of efficacy”. For example, a patient prescribed an SSRI who continues to manifest debilitating anhedonia, fatigue, and psychomotor retardation may exhibit significant improvement when switching to an antidepressant with a different mechanism of action 181 , 182 .
Overall, switching antidepressants may be considered in some cases of TRD, and the new agent should be a “non‐classmate” antidepressant.
Combining antidepressants
Persons with TRD are commonly treated with antidepressant polypharmacy, but few relevant studies have been conducted specifically in populations with TRD 183 , 184 , 185 , 186 , 187 .
Results from a meta‐analysis have supported the efficacy of adding mirtazapine or bupropion in persons with “early‐stage” TRD (i.e., non‐response to one adequate pharmacological or psychological therapy for depression) 188 . As mentioned earlier, level 2 treatment (i.e., TRD) from the STAR*D trial included seven possible switch/augmentation strategies in adults with non‐psychotic depression not achieving remission with citalopram. The three augmentation approaches were bupropion, buspirone, and cognitive therapy. The proportion of patients achieving remission after receiving bupropion combined with citalopram was 39.0%, compared to 25.5% when switching to bupropion sustained release (SR) monotherapy 33 .
A recent meta‐analysis concluded that alpha‐2 autoreceptor antagonists (i.e., mirtazapine, mianserin, trazodone) combined with SSRIs are superior to monotherapy in mixed populations including TRD, but the composition of the patient samples studied precludes any definite interpretation of the finding 189 .
Overall, data supporting the combination of antidepressants as an efficacious treatment strategy is modest in TRD populations.
Ketamine/esketamine
Intravenous (IV) racemic ketamine has been found to rapidly improve depressive symptoms and suicidal ideation in adults with TRD, and its efficacy has been confirmed in real‐world patient samples. Clinically meaningful benefit has been observed in both single and multiple infusion studies 190 , 191 , 192 , 193 . Intranasal esketamine spray co‐initiated with an antidepressant has also demonstrated rapid clinically meaningful efficacy in patients with TRD. Unlike IV ketamine, there are also data demonstrating long‐term (i.e., greater than 3‐year) safety and tolerability for esketamine 194 , 195 .
Item analysis indicates that ketamine and esketamine not only significantly improve overall symptoms of TRD, but also specific depressive symptoms that are over‐represented in adults with TRD, such as anhedonia 196 , 197 , 198 , 199 . Meta‐analytic data also indicate that glutamatergic treatment strategies may be superior to antipsychotic agents in adults with TRD 200 , 201 .
In 2019, the FDA approved intranasal esketamine spray combined with antidepressants in adults with TRD, with subsequent approvals by other regulators globally (e.g., EMA). Less evidence is available for ketamine and/or its derivatives delivered through other routes of administration 191 . Moreover, the concomitant administration of ketamine and psychological interventions (“ketamine‐assisted” therapy) is insufficiently characterized and as such cannot be recommended for TRD 202 .
Results from the recent ESCAPE‐TRD trial indicate that intranasal esketamine combined with an antidepressant is significantly more effective than quetiapine XR in TRD, with a remission rate at week 8 of 27.1% vs. 17.6% (p=0.003) 203 . Remission rates continued to increase in both arms after the primary endpoint, with a significantly greater proportion of patients in remission at week 32 in the intranasal esketamine than in the quetiapine XR arm (55% vs. 37%, p<0.001) 203 .
Preliminary evidence indicates that the effectiveness of IV ketamine in individuals with TRD and history of non‐response to neurostimulation (i.e., ECT or rTMS) is not reduced as compared to individuals with TRD and no prior neurostimulation treatment 204 . Available evidence also indicates that the efficacy of ketamine/esketamine in the acute treatment of TRD is also apparent in individuals with greater degrees of antidepressant resistance 205 .
Safety concerns attributable to long‐term ketamine/esketamine exposure include potential for abuse and misuse, tolerance and withdrawal, effects on liver function, and possibly kidney and/or urogenital toxicity 206 . The risks for the foregoing safety concerns would be expected to be mitigated when administering ketamine/esketamine under medical supervision in accordance with best practices 205 .
Second‐generation antipsychotics
The only SGA evaluated in patients failing two or more prior antidepressant treatments (i.e., TRD) is the fixed dose olanzapine‐fluoxetine combination 207 , 208 , 209 . The other SGAs assessed in MDD (i.e., aripiprazole, brexpiprazole, cariprazine, risperidone and quetiapine XR) have been studied only in patients with a partial response to at least one antidepressant 187 , 201 , 210 , 211 , 212 , 213 , 214 , 215 , 216 , 217 , 218 , 219 , 220 , 221 , 222 , 223 , 224 .
Head‐to‐head comparisons of SGAs as augmentation in TRD are not available, nor are long‐term recurrence prevention data. The absence of long‐term data with SGAs is a point of differentiation with esketamine, which has long‐term multi‐year establishment of efficacy and safety 195 . Limitations of longer‐term use of SGAs in MDD relate to tolerability and safety concerns (e.g., metabolic dysregulation, weight gain, and extrapyramidal adverse effects) 225 .
Relatively few studies have compared the antipsychotic augmentation of antidepressants versus the combination of antidepressants in patients presenting with suboptimal antidepressant response. The VA Augmentation and Switching Treatments for Improving Depression Outcomes (VAST‐D) trial was a multisite randomized, single‐blind, parallel‐assignment trial of depression unresponsive to at least one course of antidepressant treatment 226 . Eligible subjects were randomly assigned to one of three treatments: switch to bupropion SR, augmentation of current treatment with bupropion SR, or augmentation of current treatment with aripiprazole. The remission rate at week 12 was higher for the aripiprazole group (28.9%) compared with the switch to bupropion SR group (22.3%), but not with the bupropion SR add‐on group (26.9%). Response rates were significantly higher for the aripiprazole group (74.3%) than for both bupropion SR monotherapy and bupropion SR augmentation groups (62.4% and 65.6%, respectively) 226 .
The VAST‐D trial results replicate and extend the efficacy and tolerability of SGAs in individuals with MDD partially responding to antidepressants. As mentioned earlier, there are insufficient data for SGAs in TRD. However, results of the ESCAPE‐TRD trial suggest superiority of intranasal esketamine to quetiapine XR.
Neurostimulation
Neurostimulatory treatments evaluated in TRD include vagus nerve stimulation (VNS), ECT, rTMS, magnetic seizure therapy, deep brain stimulation, and transcranial direct current stimulation 227 , 228 , 229 , 230 , 231 , 232 , 233 .
VNS has proven to be efficacious in patients with higher‐order TRD (i.e., equal or greater than four prior antidepressants), and has also demonstrated durability of effect with maintenance treatment 234 , 235 , 236 . The FDA has approved VNS in TRD patients with a history of at least four prior failed antidepressants.
ECT is a well‐established therapeutic intervention in the treatment of TRD, with an average open‐label remission rate of 48% in non‐psychotic depression 237 . Efficacy may be higher in individuals with psychotic depression. Many modifications to the implementation of ECT have retained efficacy in TRD with improved tolerability profile (e.g., bilateral brief pulse ECT vs. right unilateral ultra‐brief pulse ECT) 238 .
Results from systematic reviews and meta‐analyses consistently support the efficacy of rTMS in TRD 233 . Results also indicate that greater severity at baseline and higher number of prior antidepressant failures are associated with attenuated rTMS efficacy 239 , 240 , 241 , 242 , 243 . The cost‐effectiveness of rTMS in adults with TRD is well established, and possibly higher compared to ECT, but available evidence also shows that ECT may be more effective than conventional rTMS in the acute and recurrence prevention treatment of TRD 244 , 245 , 246 .
Newer forms of rTMS are being validated, including conventional intermittent theta burst stimulation (iTBS), whose efficacy in adults with TRD when compared to sham treatment is well established 247 , 248 . An accelerated high‐dose iTBS protocol with magnetic resonance imaging (MRI)‐guided functional connectivity targeting (Stanford neuromodulation therapy, SNT) has been found, in a double‐blind randomized controlled trial (RCT), to be significantly superior relative to sham treatment four weeks after the end of the five‐day protocol. The significant benefit observed was evident despite an average of five prior antidepressant medication trials 249 . The SNT approach was recently cleared by the FDA for TRD.
In addition, results from RCTs have supported the efficacy of magnetic seizure therapy, with additional evidence demonstrating continuation of effect 250 , 251 . A Cochrane review did not identify a significant difference between this therapy and ECT in adults with TRD 252 .
Results of RCTs have not documented the efficacy of deep brain stimulation, when compared to sham treatment, in TRD 253 , 254 , 255 , 256 , 257 . Transcranial direct current stimulation is associated with variable outcomes across RCTs in the treatment of adults with TRD: the heterogeneity in response may be due to the broad range of treatment resistance included in the original trials, from treatment‐naïve to ECT failing individuals 258 .
In summary, of the foregoing neurostimulation modalities, ECT, rTMS, VNS and SNT are recommended in adults with TRD. Although there is a lack of head‐to‐head comparator data of proven treatments in TRD, preliminary evidence suggests that ECT may be non‐inferior when compared to IV racemic ketamine in adults with TRD 259 .
Psychotherapeutic interventions
There are multiple reasons for considering psychotherapeutic interventions in persons with TRD. For example, evidence indicates that these interventions are a preferred treatment option over pharmacotherapy amongst persons with lived depression experience 73 , 260 , 261 . Residual symptoms and comorbidities in persons with TRD are frequently amenable to psychological treatments. Psychotherapies, when combined with pharmacological treatments, are conceptually supported insofar as they facilitate learning, coping and resilience mechanisms that synergize with the hypothesized biological mechanisms of action of antidepressants 262 . Finally, individuals with persistent depression and history of trauma, both of which are more common in TRD populations, exhibit significant response rates with psychological interventions 263 , 264 .
Notwithstanding the rationale for use of psychotherapies in TRD, data supporting them as standalone interventions in TRD are limited 265 , 266 . Available evidence does, however, support the efficacy of adjunctive psychological interventions in persons with TRD 267 , 268 , 269 , 270 , 271 .
The psychotherapeutic modalities most frequently investigated include cognitive behavioral therapy (CBT), interpersonal psychotherapy, and mindfulness‐based cognitive therapy 272 . Meta‐analytic data have determined that psychotherapy added to ongoing treatment as usual (TAU) had a moderate and significant effect size (Hedges’ g=0.42) in comparison with TAU alone in TRD 272 .
Overall, the available evidence indicates that manual‐based psychotherapies are effective in persons with TRD when combined with antidepressants. There is insufficient evidence about combining these interventions in persons with a higher number of prior antidepressant failures and/or ECT non‐response. Patient preference, potential for scalability with digital solutions, and efficacy in the treatment of comorbidities (e.g., anxiety disorders) are additional rationales for considering psychotherapies in patients with TRD. Preliminary evidence suggests that CBT may be capable of prolonging the effect observed in adults with TRD who acutely benefited from ketamine treatment 202 .
However, a recent European study that rigorously defined TRD failed to demonstrate the efficacy of adjunctive psychological treatment 266 . It may be surmised that patient characteristics and the type of psychological intervention are critical moderators of efficacy in TRD populations.
INVESTIGATIONAL INTERVENTIONS IN TREATMENT‐RESISTANT DEPRESSION
The public health implications of TRD provide the impetus for the development of new interventions specifically for this subpopulation. It is noteworthy that enrollment in most clinical trials of investigational agents in MDD exclude patients with TRD, especially those with a high number of failed prior antidepressant trials in the current episode, or those who have failed ECT or IV ketamine in this episode.
The class of agents imprecisely referred to as psychedelics has received the most attention as a potential investigational intervention in TRD 273 . Preliminary evidence suggests that psilocybin, combined with psychotherapy, may offer rapid and possibly sustained symptom relief in adults with TRD. For example, a phase 2 double‐blind trial randomly assigned adults with TRD to receive a single dose of psilocybin 25 mg, 10 mg or 1 mg (control) along with psychological support 274 . All persons had failed at least two prior treatments before enrollment. Participants receiving the 25 mg dose, but not the 10 mg dose, exhibited a significantly greater least‐squares mean change from baseline to week 3 compared with the 1 mg dose. The response and remission rates for the participants receiving the 25 mg dose were 37% and 29%, respectively 274 .
Several methodological problems affect available controlled trials with psilocybin in TRD. Aspects of unblinding as well as expectancy are undoubtedly contributing to the observed effects, as are the psychotherapeutic modalities that are considered integral to the process of taking psychedelics. Nevertheless, the results of available RCTs with psilocybin have provided the impetus for evaluating this drug in phase 3 pivotal trials for TRD 275 . Deconstructing the contribution of psychotherapy from the psychedelic intervention will be an inexact yet necessary endeavor in order to interpret study findings and provide appropriate treatment and implementation recommendations. Moreover, the psychotherapy that is currently combined with psychedelics does not have a standardized evidence‐based protocol.
Additional investigational interventions in TRD include lithium, thyroid hormone, buspirone, L‐methylfolate, S‐adenosylmethionine, anti‐inflammatory agents (e.g., COX‐2 inhibitors, minocycline, statins, and tumor necrosis factor‐alpha antagonists), zuranolone and dextromethorphan‐bupropion combination 276 , 277 , 278 , 279 , 280 . The extant evidence supporting lithium and thyroid hormone largely refers to their combination with TCAs and MAOIs in patients with partial response to these agents. Medications that have been studied in TRD and demonstrated not to be efficacious are pindolol and buprenorphine 281 , 282 .
Despite the widespread prescription of multiple psychotropic agents off‐label in patients with TRD, there are no rigorous studies with large samples establishing the efficacy of any of the foregoing strategies.
CONCLUSIONS
Amongst individuals meeting criteria for MDD with access to high‐quality measurement‐based care, at least 30% will meet criteria for TRD. This estimate is derived from efficacy and/or effectiveness research findings. The prevalence of TRD in real world practice is not known, but would be expected to be higher, due to knowledge‐implementation gaps, barriers to access, and illness presentation complexity 283 .
With respect to illness presentation complexity, most individuals with TRD encountered in clinical practice would not be eligible for most clinical research studies, on the basis of illness characteristics (e.g., severity, number of prior episodes, suicidality), comorbidity and treatment history 13 , 284 .
Multiple definitions of TRD have been proposed and are reviewed herein. The lack of a universal definition of TRD is a barrier to advancing mechanistic and translational research, as well as to identifying innovative and precision‐based therapeutics. In addition, public policy decisions, as well as clinical decision‐making, would be benefited by a more precise and valid definition of TRD. For example, considerations for reimbursement in TRD which are critical for access to treatment are limited by the fact that multiple definitions of this condition exist. Hence, decisions by policy makers on whether to include treatments for TRD as part of a reimbursement schedule are highly variable across jurisdictions. From a clinical perspective, the lack of a universal definition of TRD contributes to heterogeneity in treatment selection and sequencing. This heterogeneity is also reflected in clinical practice guidelines for MDD, that have different recommendations with respect to selection and sequencing of treatments for adults with TRD.
Consensus exists that the lack of a clinically meaningful improvement with a minimum of two antidepressants should be retained in any working definition of TRD. A quantifiable endpoint defining non‐response should be provided. A comprehensive and conceptually valid definition of TRD with clinical utility should also include aspects of patient‐reported outcomes, psychosocial function, as well as dimensional outcomes (e.g., anhedonia) 285 .
The related, but separate, notion of DTD seems more aligned with the realities of the clinical ecosystem, and with patient experience of depression and sequential non‐response to treatments 94 , 286 . A compelling case is made that TRD is potentially judgmental insofar as it may be interpreted as blaming the patient. Instead, DTD is agnostic and represents a patient‐centered and pragmatic approach to identifying therapeutic targets 84 . The construct of DTD could serve as a useful framework informing further characterization of TRD.
The variability in antidepressant response is widely recognized 287 . A confluence of sociodemographic and clinical characteristics is known to moderate this response. Clinicians are encouraged to identify modifiable factors that attenuate antidepressant outcomes and allocate resources to these factors in patients prescribed antidepressants. For example, non‐adherence, illness and treatment illiteracy, stigma, and attitude towards treatment are modifiable with psychoeducation efforts and possibly peer‐support 73 .
In addition, psychiatric and physical comorbidities not only attenuate antidepressant response but may also be a consequence of TRD. Targeting comorbidities at the same time as depressive symptoms would be predicted to improve treatment outcomes as well as reduce cost and health resource utilization in adults with MDD. In addition, closing the implementation‐knowledge gap with fidelity to evidence‐based treatments is a near‐term cost‐effective priority in the management of MDD today.
The evidence supports select SGAs, as well as rTMS and manual‐based psychotherapies (in combination), as proven strategies in adults who have failed one prior antidepressant. For individuals with TRD (failing multiple antidepressants), evidence is best for ketamine, esketamine, adjunctive psychotherapy, ECT and rTMS. Psychotherapeutic interventions in combination with antidepressants may offer partial symptomatic relief in persons with TRD, but their efficacy as monotherapy is not established. Combination antidepressants, switching antidepressant treatment, dose optimization and the use of a host of augmentation strategies (e.g., lithium, thyroid hormone) have mixed data supporting their usefulness 288 .
Intranasal esketamine combined with an antidepressant is the most rigorously evaluated pharmacologic strategy in the acute and maintenance treatment of adults with TRD. In addition to demonstrating acute efficacy, it has established relapse prevention, tolerability and safety in persons with TRD, with more than three years of maintenance data. IV racemic ketamine has also demonstrated robust rapid antidepressant efficacy in mostly acute studies. There are relatively few controlled studies, however, that have documented maintenance efficacy of repeat‐dose IV ketamine in adults with TRD 289 .
The relative efficacy of intranasal esketamine to ECT in TRD is unknown, but is currently being evaluated. Preliminary evidence suggests that ECT may be non‐inferior to IV racemic ketamine in the acute treatment of TRD 259 . Results from large and rigorous controlled studies comparing IV ketamine to ECT are expected to provide further decision support and inform recommendations for treatment sequencing in TRD 259 .
The investigational interventions in TRD that have received the most research, media and public attention have been psychedelics. Available evidence for psilocybin suggests acute efficacy that is rapid and sustained in well‐characterized samples of persons with TRD. Unanswered questions as to the contribution of integrated psychotherapy in persons receiving psilocybin have not only conceptual and clinical relevance, but are also critical to address from an implementation perspective.
Future research vistas with respect to pharmacological treatment are testing whether ketamine derivatives or other glutamatergic agents may be useful in TRD. Additionally, GABAergic agents (e.g., zuranolone), opioid receptor modulators, orexin antagonists, voltage‐gated ion channels modulators, anti‐inflammatories, as well as agents targeting cellular metabolic processes are also under investigation in TRD 290 .
It is recognized that TRD is an under‐researched clinical population with disproportionate morbidity and mortality. Mechanistically novel interventions that offer meaningful benefit may be eligible for FDA “breakthrough status”, incentivizing treatment discovery and development in this area.
Identifying biomarkers and biosignatures associated with TRD is an important future research vista. As reviewed herein, pharmacogenomic testing has preliminary support as a tactic in assessing TRD patients, especially in cases of medication poor tolerability. Notwithstanding, it cannot be recommended as a routine assessment in all persons presenting with TRD. It is anticipated that pharmacogenomics will advance, as will the ability to computationally interrogate multi‐omic data, providing insights into the neurobiology of TRD and also potentially informing patient stratification and precision therapeutics with clinical ecosystem application potential.
Digital psychiatry encompasses aspects of health care delivery, illness surveillance, disease management and treatment 291 , 292 , 293 , 294 . Multiple proprietary and academically led product developments are underway to identify digital therapeutics that may have application in TRD populations.
The next decade can reasonably expect the regulatory approval of innovative pharmacological treatments targeting systems implicated in the pathophysiology of depression. The foregoing, along with advances in the digital delivery of psychological interventions and refinement of parameters of neurostimulation (notably rTMS with accelerated protocols), hold promise to improve general health outcomes and cost‐effectiveness of care in TRD.
The extraordinary public health burden of TRD will unlikely be extinguished in the near future, but the proportion of individuals with debilitating symptoms of depression and dissatisfaction with treatment may be reasonably expected to be decreased with successful targeting of modifiable factors, reducing the knowledge‐implementation gap, and rapid adoption of innovations across therapeutic modalities.
REFERENCES
- 1. Herrman H, Patel V, Kieling C et al. Time for united action on depression: a Lancet‐World Psychiatric Association Commission. Lancet 2022;399:957‐1022. [DOI] [PubMed] [Google Scholar]
- 2. GBD 2019 Diseases and Injuries Collaborators . Global burden of 369 diseases and injuries in 204 countries and territories, 1990‐2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet 2020;396:1204‐22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Goldberg JF, Nasrallah HA. Major depression is a serious and potentially fatal brain syndrome requiring pharmacotherapy or neuromodulation, and psychotherapy. Psychol Med 2022;52:1423‐5. [DOI] [PubMed] [Google Scholar]
- 4. Reynolds CF 3rd, Jeste DV, Sachdev PS et al. Mental health care for older adults: recent advances and new directions in clinical practice and research. World Psychiatry 2022;21:336‐63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Pérez‐Sola V, Roca M, Alonso J et al. Economic impact of treatment‐resistant depression: a retrospective observational study. J Affect Disord 2021;295:578‐86. [DOI] [PubMed] [Google Scholar]
- 6. World Health Organization . Depression and other common mental disorders: global health estimates. Geneva: World Health Organization, 2017. [Google Scholar]
- 7. Santomauro DF, Mantilla Herrera AM, Shadid J et al. Global prevalence and burden of depressive and anxiety disorders in 204 countries and territories in 2020 due to the COVID‐19 pandemic. Lancet 2021;398:1700‐12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Xiong J, Lipsitz O, Nasri F et al. Impact of COVID‐19 pandemic on mental health in the general population: a systematic review. J Affect Disord 2020;277:55‐64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Rush AJ, Sackeim HA, Conway CR et al. Research challenges in chronic diseases: difficult to treat depression. Brain Stimul 2021;14:1708‐52. [Google Scholar]
- 10. McIntyre RS, Filteau MJ, Martin L et al. Treatment‐resistant depression: definitions, review of the evidence, and algorithmic approach. J Affect Disord 2014;156:1‐7. [DOI] [PubMed] [Google Scholar]
- 11. Brown S, Rittenbach K, Cheung S et al. Current and common definitions of treatment‐resistant depression: findings from a systematic review and qualitative interviews. Can J Psychiatry 2019;64:380‐7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Henssler J, Kurschus M, Franklin J et al. Long‐term acute‐phase treatment with antidepressants, 8 weeks and beyond: a systematic review and meta‐analysis of randomized, placebo‐controlled trials. J Clin Psychiatry 2018;79:15r10545. [DOI] [PubMed] [Google Scholar]
- 13. Maj M, Stein DJ, Parker G et al. The clinical characterization of the adult patient with depression aimed at personalization of management. World Psychiatry 2020;19:269‐93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Bartova L, Dold M, Kautzky A et al. Results of the European Group for the Study of Resistant Depression (GSRD) – basis for further research and clinical practice. World J Biol Psychiatry 2019;20:427‐48. [DOI] [PubMed] [Google Scholar]
- 15. Vieta E, Alonso J, Pérez‐Sola V et al. Epidemiology and costs of depressive disorder in Spain: the EPICO study. Eur Neuropsychopharmacol 2021;50:93‐103. [DOI] [PubMed] [Google Scholar]
- 16. Kasper S. Is treatment‐resistant depression really resistant? Eur Neuropsychopharmacol 2022;58:44‐6. [DOI] [PubMed] [Google Scholar]
- 17. Zhdanava M, Pilon D, Ghelerter I et al. The prevalence and national burden of treatment‐resistant depression and major depressive disorder in the United States. J Clin Psychiatry 2021;82:20m13699. [DOI] [PubMed] [Google Scholar]
- 18. Souery D, Oswald P, Massat I et al. Clinical factors associated with treatment resistance in major depressive disorder: results from a European multicenter study. J Clin Psychiatry 2007;68:1062‐70. [DOI] [PubMed] [Google Scholar]
- 19. Institute for Health Metrics and Evaluation . GBD results. http://ghdx.healthdata.org.
- 20. McIntyre RS, Prieto R, Schepman P et al. Healthcare resource use and cost associated with timing of pharmacological treatment for major depressive disorder in the United States: a real‐world study. Curr Med Res Opin 2019;35:2169‐77. [DOI] [PubMed] [Google Scholar]
- 21. McIntyre RS, Millson B, Power GS. Burden of treatment resistant depression (TRD) in patients with major depressive disorder in Ontario using Institute for Clinical Evaluative Sciences (ICES) databases: economic burden and healthcare resource utilization. J Affect Disord 2020;277:30‐8. [DOI] [PubMed] [Google Scholar]
- 22. Sussman M, O'sullivan AK, Shah A et al. Economic burden of treatment‐resistant depression on the U.S. health care system. J Manag Care Spec Pharm 2019;25:823‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Jensen KJ, Gronemann FH, Ankarfeldt MZ et al. Healthcare resource utilization in patients with treatment‐resistant depression – A Danish national registry study. PLoS One 2022;17:e0275299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Heerlein K, De Giorgi S, Degraeve G et al. Real‐world evidence from a European cohort study of patients with treatment resistant depression: healthcare resource utilization. J Affect Disord 2022;298:442‐50. [DOI] [PubMed] [Google Scholar]
- 25. Rathod S, Denee T, Eva J et al. Health‐related quality of life burden associated with treatment‐resistant depression in UK patients: quantitative results from a mixed‐methods non‐interventional study. J Affect Disord 2022;300:551‐62. [DOI] [PubMed] [Google Scholar]
- 26. Gillain B, Degraeve G, Dreesen T et al. Real‐world treatment patterns, outcomes, resource utilization and costs in treatment‐resistant major depressive disorder: PATTERN, a retrospective cohort study in Belgium. Pharmacoecon Open 2022;6:293‐302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Perrone V, Sangiorgi D, Andretta M et al. Healthcare resource consumption and related costs of patients estimated with treatment‐resistant depression in Italy. Clinicoecon Outcomes Res 2021;13:629‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Lynch FL, Dickerson JF, O'Keeffe‐Rosetti M et al. Understanding the relationship between depression symptom severity and health care costs for patients with treatment‐resistant depression. J Clin Psychiatry 2022;83:21m13976. [DOI] [PubMed] [Google Scholar]
- 29. Olfson M, Amos TB, Benson C et al. Prospective service use and health care costs of Medicaid beneficiaries with treatment‐resistant depression. J Manag Care Spec Pharm 2018;24:226‐36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Gaynes BN, Asher G, Gartlehner G et al. Definition of treatment‐resistant depression in the Medicare population. Rockville: Agency for Healthcare Research and Quality, 2018. [PubMed] [Google Scholar]
- 31. 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;3:137‐44. [DOI] [PubMed] [Google Scholar]
- 32. Nemeroff CB. Prevalence and management of treatment‐resistant depression. J Clin Psychiatry 2007;68(Suppl. 8):17‐25. [PubMed] [Google Scholar]
- 33. Rush AJ, Trivedi MH, Wisniewski SR et al. Acute and longer‐term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Psychiatry 2006;163:1905‐17. [DOI] [PubMed] [Google Scholar]
- 34. Chan VK, Cheung EC, Chan SS et al. Mortality‐causing mechanisms and healthcare resource utilisation of treatment‐resistant depression: a six‐year population‐based cohort study. Lancet Reg Health West Pac 2022;22:100426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Denee T, Kerr C, Eva J et al. The impact of treatment‐resistant depression on the lives of carers: a mixed‐methods study. J Affect Disord 2022;325:194‐205. [DOI] [PubMed] [Google Scholar]
- 36. Mann JJ, Michel CA, Auerbach RP. Improving suicide prevention through evidence‐based strategies: a systematic review. Am J Psychiatry 2021;178:611‐24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Brydges CR, Bhattacharyya S, Dehkordi SM et al. Metabolomic and inflammatory signatures of symptom dimensions in major depression. Brain Behav Immun 2022;102:42‐52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. McIntyre RS. Surrogate markers of insulin resistance in predicting major depressive disorder: metabolism metastasizes to the brain. Am J Psychiatry 2021;178:885‐7. [DOI] [PubMed] [Google Scholar]
- 39. Armbrecht E, Shah R, Poorman GW et al. Economic and humanistic burden associated with depression and anxiety among adults with non‐communicable chronic diseases (NCCDs) in the United States. J Multidiscip Healthc 2021;14:887‐96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. McIntyre RS, Soczynska JK, Konarski JZ et al. Should depressive syndromes be reclassified as “metabolic syndrome type II”? Ann Clin Psychiatry 2007;19:257‐64. [DOI] [PubMed] [Google Scholar]
- 41. McIntyre RS, Rosenbluth M, Ramasubbu R et al. Managing medical and psychiatric comorbidity in individuals with major depressive disorder and bipolar disorder. Ann Clin Psychiatry 2012;24:163‐9. [PubMed] [Google Scholar]
- 42. Lee Y, Brietzke E, Cao B et al. Development and implementation of guidelines for the management of depression: a systematic review. Bull World Health Organ 2020;98:683‐97H. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Cleare A, Pariante CM, Young AH et al. Evidence‐based guidelines for treating depressive disorders with antidepressants: a revision of the 2008 British Association for Psychopharmacology guidelines. J Psychopharmacol 2015;29:459‐525. [DOI] [PubMed] [Google Scholar]
- 44. 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. Can J Psychiatry 2016;61:540‐60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Bauer M, Severus E, Möller HJ et al. Pharmacological treatment of unipolar depressive disorders: summary of WFSBP guidelines. Int J Psychiatry Clin Pract 2017;21:166‐76. [DOI] [PubMed] [Google Scholar]
- 46. Nutt DJ, Davidson JRT, Gelenberg AJ et al. International consensus statement on major depressive disorder. J Clin Psychiatry 2010;71(Suppl. E1):e08. [DOI] [PubMed] [Google Scholar]
- 47. McIntyre RS, Suppes T, Tandon R et al. Florida best practice psychotherapeutic medication guidelines for adults with major depressive disorder. J Clin Psychiatry 2017;78:703‐13. [DOI] [PubMed] [Google Scholar]
- 48. National Institute for Health and Care Excellence . Depression in adults: treatment and management. https://www.nice.org.uk. [PubMed]
- 49. Insel TR. The NIMH Research Domain Criteria (RDoC) project: precision medicine for psychiatry. Am J Psychiatry 2014;171:395‐7. [DOI] [PubMed] [Google Scholar]
- 50. McIntyre RS, Alda M, Baldessarini RJ et al. The clinical characterization of the adult patient with bipolar disorder aimed at personalization of management. World Psychiatry 2022;21:364‐87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Stein DJ, Shoptaw SJ, Vigo DV et al. Psychiatric diagnosis and treatment in the 21st century: paradigm shifts versus incremental integration. World Psychiatry 2022;21:393‐414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. U.S. Food and Drug Administration . Center for Drug Evaluation and Research. Major depressive disorder: developing drugs for treatment. Silver Spring: U.S. Food and Drug Administration, 2018. [Google Scholar]
- 53. European Medicines Agency . Clinical investigation of medicinal products in the treatment of depression – Scientific guideline. Amsterdam: European Medicines Agency, 2018. [Google Scholar]
- 54. Thase ME, Rush AJ. When at first you don't succeed: sequential strategies for antidepressant nonresponders. J Clin Psychiatry 1997;58(Suppl. 13):23‐9. [PubMed] [Google Scholar]
- 55. Thase ME, Rush AJ. Treatment resistant depression. In: Bloom FE, Kupfer DJ (eds). Psychopharmacology: the fourth generation of progress. New York: Raven Press, 1995:1081‐97. [Google Scholar]
- 56. Fekadu A, Donocik JG, Cleare AJ. Standardisation framework for the Maudsley staging method for treatment resistance in depression. BMC Psychiatry 2018;18:100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Fekadu A, Wooderson SC, Markopoulou K et al. The Maudsley Staging Method for treatment‐resistant depression: prediction of longer‐term outcome and persistence of symptoms. J Clin Psychiatry 2009;70:952‐7. [DOI] [PubMed] [Google Scholar]
- 58. Fekadu A, Rane LJ, Wooderson SC et al. Prediction of longer‐term outcome of treatment‐resistant depression in tertiary care. Br J Psychiatry 2012;201:369‐75. [DOI] [PubMed] [Google Scholar]
- 59. Hamilton M. A rating scale for depression. J Neurol Neurosurg Psychiatry 1960;23:56‐62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Montgomery SA, Åsberg M. A new depression scale designed to be sensitive to change. Br J Psychiatry 1979;134:382‐9. [DOI] [PubMed] [Google Scholar]
- 61. Peeters FPML, Ruhe HG, Wichers M et al. The Dutch Measure for quantification of Treatment Resistance in Depression (DM‐TRD): an extension of the Maudsley Staging Method. J Affect Disord 2016;205:365‐71. [DOI] [PubMed] [Google Scholar]
- 62. Fava M. Diagnosis and definition of treatment‐resistant depression. Biol Psychiatry 2003;53:649‐59. [DOI] [PubMed] [Google Scholar]
- 63. Sforzini L, Worrell C, Kose M et al. A Delphi‐method‐based consensus guideline for definition of treatment‐resistant depression for clinical trials. Mol Psychiatry 2022;27:1286‐99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Demyttenaere K, Van Duppen Z. The impact of (the concept of) treatment‐resistant depression: an opinion review. Int J Neuropsychopharmacol 2019;22:85‐92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Trevino K, McClintock SM, McDonald Fischer N et al. Defining treatment‐resistant depression: a comprehensive review of the literature. Ann Clin Psychiatry 2014;26:222‐32. [PubMed] [Google Scholar]
- 66. Conway CR, George MS, Sackeim HA. Toward an evidence‐based, operational definition of treatment‐resistant depression: when enough is enough. JAMA Psychiatry 2017;74:9‐10. [DOI] [PubMed] [Google Scholar]
- 67. McAllister‐Williams RH, Christmas DMB, Cleare AJ et al. Multiple‐therapy‐resistant major depressive disorder: a clinically important concept. Br J Psychiatry 2018;212:274‐8. [DOI] [PubMed] [Google Scholar]
- 68. Gabriel FC, Stein AT, de Melo DO et al. Quality of clinical practice guidelines for inadequate response to first‐line treatment for depression according to AGREE II checklist and comparison of recommendations: a systematic review. BMJ Open 2022;12:e051918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. McIntyre RS, O'Donovan C. The human cost of not achieving full remission in depression. Can J Psychiatry 2004;49(Suppl. 1):10‐16S. [PubMed] [Google Scholar]
- 70. Rush AJ, Aaronson ST, Demyttenaere K. Difficult‐to‐treat depression: a clinical and research roadmap for when remission is elusive. Aust N Z J Psychiatry 2019;53:109‐18. [DOI] [PubMed] [Google Scholar]
- 71. Nierenberg AA, Husain MM, Trivedi MH et al. Residual symptoms after remission of major depressive disorder with citalopram and risk of relapse: a STAR*D report. Psychol Med 2010;40:41‐50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. McIntyre RS, Cha DS, Soczynska JK et al. Cognitive deficits and functional outcomes in major depressive disorder: determinants, substrates, and treatment interventions. Depress Anxiety 2013;30:515‐27. [DOI] [PubMed] [Google Scholar]
- 73. Rosenblat JD, Simon GE, Sachs GS et al. Frequency of use and perceived helpfulness of wellness strategies for bipolar and unipolar depression. Ann Clin Psychiatry 2018;30:296‐304. [PubMed] [Google Scholar]
- 74. IsHak WW, Steiner AJ, Klimowicz A et al. Major depression comorbid with medical conditions: analysis of quality of life, functioning, and depressive symptom severity. Psychopharmacol Bull 2018;48:8‐25. [PMC free article] [PubMed] [Google Scholar]
- 75. IsHak WW, Mirocha J, James D et al. Quality of life in major depressive disorder before/after multiple steps of treatment and one‐year follow‐up. Acta Psychiatr Scand 2015;131:51‐60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. McIntyre RS, Florea I, Tonnoir B et al. Efficacy of vortioxetine on cognitive functioning in working patients with major depressive disorder. J Clin Psychiatry 2017;78:115‐21. [DOI] [PubMed] [Google Scholar]
- 77. IsHak WW, James DM, Mirocha J et al. Patient‐reported functioning in major depressive disorder. Ther Adv Chronic Dis 2016;7:160‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. IsHak WW, Greenberg JM, Cohen RM. Predicting relapse in major depressive disorder using patient‐reported outcomes of depressive symptom severity, functioning, and quality of life in the Individual Burden of Illness Index for Depression (IBI‐D). J Affect Disord 2013;151:59‐65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Firth J, Siddiqi N, Koyanagi A et al. The Lancet Psychiatry Commission: a blueprint for protecting physical health in people with mental illness. Lancet Psychiatry 2019;6:675‐712. [DOI] [PubMed] [Google Scholar]
- 80. de Menezes Galvão AC, Almeida RN, de Sousa GM Jr et al. Pathophysiology of major depression by clinical stages. Front Psychol 2021;12:641779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Cuijpers P, Quero S, Noma H et al. Psychotherapies for depression: a network meta‐analysis covering efficacy, acceptability and long‐term outcomes of all main treatment types. World Psychiatry 2021;20:283‐93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Kessler RC, Kazdin AE, Aguilar‐Gaxiola S et al. Patterns and correlates of patient‐reported helpfulness of treatment for common mental and substance use disorders in the WHO World Mental Health Surveys. World Psychiatry 2022;21:272‐86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Lundberg J, Cars T, Lööv SÅ et al. Association of treatment‐resistant depression with patient outcomes and health care resource utilization in a population‐wide study. JAMA Psychiatry 2023;80:167‐75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Rush AJ, Sackeim HA, Conway CR et al. Clinical research challenges posed by difficult‐to‐treat depression. Psychol Med 2022;52:419‐32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Zhang C, Virani S, Mayes T et al. Toward a definition of “no meaningful benefit” from antidepressant treatment: an equipercentile analysis with cross‐trial validation across multiple rating scales. J Clin Psychiatry 2022;83:21m14239. [DOI] [PubMed] [Google Scholar]
- 86. Turkoz I, Alphs L, Singh J et al. Clinically meaningful changes on depressive symptom measures and patient‐reported outcomes in patients with treatment‐resistant depression. Acta Psychiatr Scand 2021;143:253‐63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Conway CR, Kumar A, Xiong W et al. Chronic vagus nerve stimulation significantly improves quality of life in treatment‐resistant major depression. J Clin Psychiatry 2018;79:18m12178. [DOI] [PubMed] [Google Scholar]
- 88. Morrens J, Mathews M, Popova V et al. Use of Clinical Global Impressions‐Severity (CGI‐S) to assess response to antidepressant treatment in patients with treatment‐resistant depression. Neuropsychiatr Dis Treat 2022;18:1127‐32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. McIntyre RS, Lipsitz O, Lui LMW et al. The meaningful change threshold as measured by the 16‐item Quick Inventory of Depressive Symptomatology in adults with treatment‐resistant major depressive and bipolar disorder receiving intravenous ketamine. J Affect Disord 2021;294:592‐6. [DOI] [PubMed] [Google Scholar]
- 90. Demyttenaere K, Kiekens G, Bruffaerts R et al. Outcome in depression (I): why symptomatic remission is not good enough. CNS Spectr 2021;26:393‐9. [DOI] [PubMed] [Google Scholar]
- 91. Demyttenaere K, Kiekens G, Bruffaerts R et al. Outcome in depression (II): beyond the Hamilton Depression Rating Scale. CNS Spectr 2021;26:378‐82. [DOI] [PubMed] [Google Scholar]
- 92. Rosenblat JD, Simon GE, Sachs GS et al. Treatment effectiveness and tolerability outcomes that are most important to individuals with bipolar and unipolar depression. J Affect Disord 2019;243:116‐20. [DOI] [PubMed] [Google Scholar]
- 93. McAllister‐Williams RH, Aaronson ST, Conway CR et al. The “difficult‐to‐treat depression” and the “response paradigm” models: implications and relevance to patient management. Aust N Z J Psychiatry 2021;55:824‐5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. McAllister‐Williams RH, Arango C, Blier P et al. Reconceptualising treatment‐resistant depression as difficult‐to‐treat depression. Lancet Psychiatry 2021;8:14‐5. [DOI] [PubMed] [Google Scholar]
- 95. McCue M, Parikh SV, Mucha L et al. Adapting the goal attainment approach for major depressive disorder. Neurol Ther 2019;8:167‐76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. McCue M, Sarkey S, Eramo A et al. Using the Goal Attainment Scale adapted for depression to better understand treatment outcomes in patients with major depressive disorder switching to vortioxetine: a phase 4, single‐arm, open‐label, multicenter study. BMC Psychiatry 2021;21:622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Henssler J, Bschor T, Baethge C. Combination antidepressant therapy vs monotherapy – further considerations – reply. JAMA Psychiatry 2022;79:832‐3. [DOI] [PubMed] [Google Scholar]
- 98. Liu X, Mukai Y, Furtek CI et al. Epidemiology of treatment‐resistant depression in the United States. J Clin Psychiatry 2021;83:21m13964. [DOI] [PubMed] [Google Scholar]
- 99. Thomas L, Kessler D, Campbell J et al. Prevalence of treatment‐resistant depression in primary care: cross‐sectional data. Br J Gen Pract 2013;63:e852‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Fife D, Reps J, Cepeda MS et al. Treatment resistant depression incidence estimates from studies of health insurance databases depend strongly on the details of the operating definition. Heliyon 2018;4:e00707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Sharman Moser S, Chodick G, Gelerstein S et al. Epidemiology of treatment resistant depression among major depressive disorder patients in Israel. BMC Psychiatry 2022;22:541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Fekadu A, Demissie M, Birhane R et al. Under detection of depression in primary care settings in low and middle‐income countries: a systematic review and meta‐analysis. Syst Rev 2022;11:21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. National Institute for Health and Care Excellence . Depression in adults. London: National Institute for Health and Care Excellence, 2018. [Google Scholar]
- 104. Martin‐Cook K, Palmer L, Thornton L et al. Setting measurement‐based care in motion: practical lessons in the implementation and integration of measurement‐based care in psychiatry clinical practice. Neuropsychiatr Dis Treat 2021;17:1621‐31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Mitchell AJ, Rao S, Vaze A. International comparison of clinicians' ability to identify depression in primary care: meta‐analysis and meta‐regression of predictors. Br J Gen Pract 2011;61:e72‐80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Pence BW, O'Donnell JK, Gaynes BN. The depression treatment cascade in primary care: a public health perspective. Curr Psychiatry Rep 2012;14:328‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Ruhé HG, van Rooijen G, Spijker J et al. Staging methods for treatment resistant depression. A systematic review. J Affect Disord 2012;137:35‐45. [DOI] [PubMed] [Google Scholar]
- 108. McAllister‐Williams RH, Arango C, Blier P et al. The identification, assessment and management of difficult‐to‐treat depression: an international consensus statement. J Affect Disord 2020;267:264‐82. [DOI] [PubMed] [Google Scholar]
- 109. Azar AR, Chopra MP, Cho LY et al. Remission in major depression: results from a geriatric primary care population. Int J Geriatr Psychiatry 2011;26:48‐55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Cooper C, Katona C, Lyketsos K et al. A systematic review of treatments for refractory depression in older people. Am J Psychiatry 2011;168:681‐8. [DOI] [PubMed] [Google Scholar]
- 111. Cuijpers P, Karyotaki E, Eckshtain D et al. Psychotherapy for depression across different age groups: a systematic review and meta‐analysis. JAMA Psychiatry 2020;77:694‐702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Dominiak M, Antosik‐Wójcin´ska AZ, Wojnar M et al. Electroconvulsive therapy and age: effectiveness, safety and tolerability in the treatment of major depression among patients under and over 65 years of age. Pharmaceuticals 2021;14:582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Kaster TS, Daskalakis ZJ, Noda Y et al. Efficacy, tolerability, and cognitive effects of deep transcranial magnetic stimulation for late‐life depression: a prospective randomized controlled trial. Neuropsychopharmacology 2018;43:2231‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Lähteenvuo M, Taipale H, Tanskanen A et al. Courses of treatment and risk factors for treatment‐resistant depression in Finnish primary and special healthcare: a nationwide cohort study. J Affect Disord 2022;308:236‐42. [DOI] [PubMed] [Google Scholar]
- 115. Cepeda MS, Kern DM, Nicholson S. Treatment resistant depression in women with peripartum depression. BMC Pregnancy Childbirth 2019;19:323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Kuehner C. Why is depression more common among women than among men? Lancet Psychiatry 2017;4:146‐58. [DOI] [PubMed] [Google Scholar]
- 117. Trivedi MH, Rush AJ, Wisniewski SR et al. Evaluation of outcomes with citalopram for depression using measurement‐based care in STAR*D: implications for clinical practice. Am J Psychiatry 2006;163:28‐40. [DOI] [PubMed] [Google Scholar]
- 118. IsHak WW, Bonifay W, Collison K et al. The recovery index: a novel approach to measuring recovery and predicting remission in major depressive disorder. J Affect Disord 2017;208:369‐74. [DOI] [PubMed] [Google Scholar]
- 119. Teicher MH, Gordon JB, Nemeroff CB. Recognizing the importance of childhood maltreatment as a critical factor in psychiatric diagnoses, treatment, research, prevention, and education. Mol Psychiatry 2022;27:1331‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Wang T, Li L, Yue Y et al. The interaction of P11 methylation and early‐life stress impacts the antidepressant response in patients with major depressive disorder. J Affect Disord 2022;312:128‐35. [DOI] [PubMed] [Google Scholar]
- 121. Yang JZ, Kang CY, Yuan J et al. Effect of adverse childhood experiences on hypothalamic‐pituitary‐adrenal (HPA) axis function and antidepressant efficacy in untreated first episode patients with major depressive disorder. Psychoneuroendocrinology 2021;134:105432. [DOI] [PubMed] [Google Scholar]
- 122. Menke A, Nitschke F, Hellmuth A et al. Stress impairs response to antidepressants via HPA axis and immune system activation. Brain Behav Immun 2021;93:132‐40. [DOI] [PubMed] [Google Scholar]
- 123. Williams LM, Debattista C, Duchemin AM et al. Childhood trauma predicts antidepressant response in adults with major depression: data from the randomized international study to predict optimized treatment for depression. Transl Psychiatry 2016;6:e799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Kaplan MJ, Klinetob NA. Childhood emotional trauma and chronic posttraumatic stress disorder in adult outpatients with treatment‐resistant depression. J Nerv Ment Dis 2000;188:596‐601. [DOI] [PubMed] [Google Scholar]
- 125. McAllister‐Williams RH. When depression is difficult to treat. Eur Neuropsychopharmacol 2022;56:89‐91. [DOI] [PubMed] [Google Scholar]
- 126. Christensen MC, Florea I, Loft H et al. Efficacy of vortioxetine in patients with major depressive disorder reporting childhood or recent trauma. J Affect Disord 2020;263:258‐66. [DOI] [PubMed] [Google Scholar]
- 127. O'Brien B, Lijffijt M, Wells A et al. The impact of childhood maltreatment on intravenous ketamine outcomes for adult patients with treatment‐resistant depression. Pharmaceuticals 2019;12:133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Yrondi A, Vaiva G, Walter M et al. Childhood trauma increases suicidal behaviour in a treatment‐resistant depression population: a FACE‐DR report. J Psychiatr Res 2021;135:20‐7. [DOI] [PubMed] [Google Scholar]
- 129. Scott J, Eccleston D, Boys R. Can we predict the persistence of depression? Br J Psychiatry 1992;161:633‐7. [DOI] [PubMed] [Google Scholar]
- 130. McIntyre RS, Berk M, Brietzke E et al. Bipolar disorders. Lancet 2020;396:1841‐56. [DOI] [PubMed] [Google Scholar]
- 131. McIntyre RS, Ng‐Mak D, Chuang CC et al. Major depressive disorder with subthreshold hypomanic (mixed) features: a real‐world assessment of treatment patterns and economic burden. J Affect Disord 2017;210:332‐7. [DOI] [PubMed] [Google Scholar]
- 132. Cao B, Zhu J, Zuckerman H et al. Pharmacological interventions targeting anhedonia in patients with major depressive disorder: a systematic review. Prog Neuropsychopharmacol Biol Psychiatry 2019;92:109‐17. [DOI] [PubMed] [Google Scholar]
- 133. Goodwin GM, Price J, De Bodinat C et al. Emotional blunting with antidepressant treatments: a survey among depressed patients. J Affect Disord 2017;221:31‐5. [DOI] [PubMed] [Google Scholar]
- 134. Millan MJ, Agid Y, Brüne M et al. Cognitive dysfunction in psychiatric disorders: characteristics, causes and the quest for improved therapy. Nat Rev Drug Discov 2012;11:141‐68. [DOI] [PubMed] [Google Scholar]
- 135. Rosenblat JD, Kakar R, McIntyre RS. The cognitive effects of antidepressants in major depressive disorder: a systematic review and meta‐analysis of randomized clinical trials. Int J Neuropsychopharmacol 2015;19:pyv082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Ang YS, Bruder GE, Keilp JG et al. Exploration of baseline and early changes in neurocognitive characteristics as predictors of treatment response to bupropion, sertraline, and placebo in the EMBARC clinical trial. Psychol Med 2022;52:2441‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Kessler RC, Sampson NA, Berglund P et al. Anxious and non‐anxious major depressive disorder in the World Health Organization World Mental Health Surveys. Epidemiol Psychiatr Sci 2015;24:210‐26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Robinson OJ, Vytal K, Cornwell BR et al. The impact of anxiety upon cognition: perspectives from human threat of shock studies. Front Hum Neurosci 2013;7:203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Trombello JM, Pizzagalli DA, Weissman MM et al. Characterizing anxiety subtypes and the relationship to behavioral phenotyping in major depression: results from the EMBARC study. J Psychiatr Res 2018;102:207‐15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Yoo I, Woo JM, Lee SH et al. Influence of anxiety symptoms on improvement of neurocognitive functions in patients with major depressive disorder: a 12‐week, multicenter, randomized trial of tianeptine versus escitalopram, the CAMPION study. J Affect Disord 2015;185:24‐30. [DOI] [PubMed] [Google Scholar]
- 141. Fava M, Rush AJ, Alpert JE et al. Difference in treatment outcome in outpatients with anxious versus nonanxious depression: a STAR*D report. Am J Psychiatry 2008;165:342‐51. [DOI] [PubMed] [Google Scholar]
- 142. Kautzky A, Dold M, Bartova L et al. Clinical factors predicting treatment resistant depression: affirmative results from the European multicenter study. Acta Psychiatr Scand 2019;139:78‐88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. McIntyre RS, Schaffer A, Beaulieu S. The Canadian Network for Mood and Anxiety Treatments (CANMAT) task force recommendations for the management of patients with mood disorders and comorbid conditions. Ann Clin Psychiatry 2012;24:2‐3. [PubMed] [Google Scholar]
- 144. Cao B, Xu J, Li R et al. Interventions targeting comorbid depression and overweight/obesity: a systematic review. J Affect Disord 2022;314:222‐32. [DOI] [PubMed] [Google Scholar]
- 145. Grigolon RB, Trevizol AP, Gerchman F et al. Is obesity a determinant of success with pharmacological treatment for depression? A systematic review, meta‐analysis and meta‐regression. J Affect Disord 2021;287:54‐68. [DOI] [PubMed] [Google Scholar]
- 146. Rashidian H, Subramaniapillai M, Park C et al. Changes in insulin resistance following antidepressant treatment mediate response in major depressive disorder. J Psychopharmacol 2023;37:313‐7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Toups MSP, Myers AK, Wisniewski SR et al. Relationship between obesity and depression: characteristics and treatment outcomes with antidepressant medication. Psychosom Med 2013;75:863‐72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Woo YS, Seo HJ, McIntyre RS et al. Obesity and its potential effects on antidepressant treatment outcomes in patients with depressive disorders: a literature review. Int J Mol Sci 2016;17:80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Vogelzangs N, Beekman ATF, van Reedt Dortland AKB et al. Inflammatory and metabolic dysregulation and the 2‐year course of depressive disorders in antidepressant users. Neuropsychopharmacology 2014;39:1624‐34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Fanelli G, Serretti A. Depression, antidepressants, and insulin resistance: which link? Eur Neuropsychopharmacol 2022;60:4‐6. [DOI] [PubMed] [Google Scholar]
- 151. Beran M, Muzambi R, Geraets A et al. The bidirectional longitudinal association between depressive symptoms and HbA1c: a systematic review and meta‐analysis. Diabet Med 2022;39:e14671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Pan A, Keum N, Okereke OI et al. Bidirectional association between depression and metabolic syndrome: a systematic review and meta‐analysis of epidemiological studies. Diabetes Care 2012;35:1171‐80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Nierenberg AA, Amsterdam JD. Treatment‐resistant depression: definition and treatment approaches. J Clin Psychiatry 1990;51(Suppl.):39‐47. [PubMed] [Google Scholar]
- 154. Rush AJ, Thase ME. Improving depression outcome by patient‐centered medical management. Am J Psychiatry 2018;175:1187‐98. [DOI] [PubMed] [Google Scholar]
- 155. Mitchell AJ, Vaze A, Rao S. Clinical diagnosis of depression in primary care: a meta‐analysis. Lancet 2009;374:609‐19. [DOI] [PubMed] [Google Scholar]
- 156. McIntyre RS, Calabrese JR. Bipolar depression: the clinical characteristics and unmet needs of a complex disorder. Curr Med Res Opin 2019;35:1993‐2005. [DOI] [PubMed] [Google Scholar]
- 157. Li CT, Bai YM, Huang YL et al. Association between antidepressant resistance in unipolar depression and subsequent bipolar disorder: cohort study. Br J Psychiatry 2012;200:45‐51. [DOI] [PubMed] [Google Scholar]
- 158. Elefante C, Brancati GE, Petrucci A et al. Risk of conversion to bipolar disorder in patients with late‐onset major depression. Int Clin Psychopharmacol 2022;37:234‐41. [DOI] [PubMed] [Google Scholar]
- 159. Goldberg JF, Nierenberg AA, Iosifescu DV. Wrestling with antidepressant use in bipolar disorder: the ongoing debate. J Clin Psychiatry 2021;82:19ac13181. [DOI] [PubMed] [Google Scholar]
- 160. McIntyre RS, Patel MD, Masand PS et al. The Rapid Mood Screener (RMS): a novel and pragmatic screener for bipolar I disorder. Curr Med Res Opin 2021;37:135‐44. [DOI] [PubMed] [Google Scholar]
- 161. Cerimele JM, Russo J, Bauer AM et al. The Patient Mania Questionnaire (PMQ‐9): a brief scale for assessing and monitoring manic symptoms. J Gen Intern Med 2022;37:1680‐7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Villagonzalo KA, Dodd S, Ng F et al. The utility of the Mood Disorder Questionnaire as a screening tool in a methadone maintenance treatment program. Int J Psychiatry Clin Pract 2010;14:150‐3. [DOI] [PubMed] [Google Scholar]
- 163. Meyer TD, Hammelstein P, Nilsson L‐G et al. The Hypomania Checklist (HCL‐32): its factorial structure and association to indices of impairment in German and Swedish nonclinical samples. Compr Psychiatry 2007;48:79‐87. [DOI] [PubMed] [Google Scholar]
- 164. Sackeim HA. The definition and meaning of treatment‐resistant depression. J Clin Psychiatry 2001;62(Suppl. 16):10‐7. [PubMed] [Google Scholar]
- 165. Sackeim HA, Aaronson ST, Bunker MT et al. The assessment of resistance to antidepressant treatment: rationale for the Antidepressant Treatment History Form: Short Form (ATHF‐SF). J Psychiatr Res 2019;113:125‐36. [DOI] [PubMed] [Google Scholar]
- 166. Chandler GM, Iosifescu DV, Pollack MH et al. Validation of the Massachusetts General Hospital Antidepressant Treatment History Questionnaire (ATRQ). CNS Neurosci Ther 2010;16:322‐5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Rush AJ, Trivedi MH, Wisniewski SR et al. Acute and longer‐term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Psychiatry 2006;163:1905‐17. [DOI] [PubMed] [Google Scholar]
- 168. Szegedi A, Jansen WT, van Willigenburg APP et al. Early improvement in the first 2 weeks as a predictor of treatment outcome in patients with major depressive disorder: a meta‐analysis including 6562 patients. J Clin Psychiatry 2009;70:344‐53. [DOI] [PubMed] [Google Scholar]
- 169. Pedrosa‐Naudín MA, Gutiérrez‐Abejón E, Herrera‐Gómez F et al. Non‐adherence to antidepressant treatment and related factors in a region of Spain: a population‐based registry study. Pharmaceutics 2022;14:2696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Topp CW, Østergaard SD, Søndergaard S et al. The WHO‐5 Well‐Being Index: a systematic review of the literature. Psychother Psychosom 2015;84:167‐76. [DOI] [PubMed] [Google Scholar]
- 171. Arnone D, Omar O, Arora T et al. Effectiveness of pharmacogenomic tests including CYP2D6 and CYP2C19 genomic variants for guiding the treatment of depressive disorders: systematic review and meta‐analysis of randomised controlled trials. Neurosci Biobehav Rev 2023;144:104965. [DOI] [PubMed] [Google Scholar]
- 172. Greden JF, Parikh SV, Rothschild AJ et al. Impact of pharmacogenomics on clinical outcomes in major depressive disorder in the GUIDED trial: a large, patient‐ and rater‐blinded, randomized, controlled study. J Psychiatr Res 2019;111:59‐67. [DOI] [PubMed] [Google Scholar]
- 173. Rothschild AJ, Parikh SV, Hain D et al. Clinical validation of combinatorial pharmacogenomic testing and single‐gene guidelines in predicting psychotropic medication blood levels and clinical outcomes in patients with depression. Psychiatry Res 2021;296:113649. [DOI] [PubMed] [Google Scholar]
- 174. Rosenblat JD, Lee Y, McIntyre RS. Does pharmacogenomic testing improve clinical outcomes for major depressive disorder? A systematic review of clinical trials and cost‐effectiveness studies. J Clin Psychiatry 2017;78:720‐9. [DOI] [PubMed] [Google Scholar]
- 175. Piacentino D, Bianchi E, De Donatis D et al. Therapeutic drug monitoring of antidepressants: an underused but potentially valuable tool in primary care. Front Psychiatry 2022;13:867840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Funk CSM, Hart XM, Gründer G et al. Is therapeutic drug monitoring relevant for antidepressant drug therapy? implications from a systematic review and meta‐analysis with focus on moderating factors. Front Psychiatry 2022;13:826138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Cellini L, De Donatis D, Zernig G et al. Antidepressant efficacy is correlated with plasma levels: mega‐analysis and further evidence. Int Clin Psychopharmacol 2022;37:29‐37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Henssler J, Kurschus M, Franklin J et al. Trajectories of acute antidepressant efficacy: how long to wait for response? A systematic review and meta‐analysis of long‐term, placebo‐controlled acute treatment trials. J Clin Psychiatry 2018;79:17r11470. [DOI] [PubMed] [Google Scholar]
- 179. Brignone M, Diamand F, Painchault C et al. Efficacy and tolerability of switching therapy to vortioxetine versus other antidepressants in patients with major depressive disorder. Curr Med Res Opin 2016;32:351‐66. [DOI] [PubMed] [Google Scholar]
- 180. Bschor T, Kern H, Henssler J et al. Switching the antidepressant after nonresponse in adults with major depression: a systematic literature search and meta‐analysis. J Clin Psychiatry 2018;79:16r10749. [DOI] [PubMed] [Google Scholar]
- 181. Papakostas GI, Thase ME, Fava M et al. Are antidepressant drugs that combine serotonergic and noradrenergic mechanisms of action more effective than the selective serotonin reuptake inhibitors in treating major depressive disorder? A meta‐analysis of studies of newer agents. Biol Psychiatry 2007;62:1217‐27. [DOI] [PubMed] [Google Scholar]
- 182. Fagiolini A, Florea I, Loft H et al. Effectiveness of vortioxetine on emotional blunting in patients with major depressive disorder with inadequate response to SSRI/SNRI treatment. J Affect Disord 2021;283:472‐9. [DOI] [PubMed] [Google Scholar]
- 183. Grover D, Tom M, Maguire G et al. Polypharmacy – purpose, benefits and limitations. Curr Med Chem 2022;29:5606‐14. [DOI] [PubMed] [Google Scholar]
- 184. Mojtabai R, Olfson M. National trends in psychotropic medication polypharmacy in office‐based psychiatry. Arch Gen Psychiatry 2010;67:26‐36. [DOI] [PubMed] [Google Scholar]
- 185. Dold M, Kautzky A, Bartova L et al. Pharmacological treatment strategies in unipolar depression in European tertiary psychiatric treatment centers – A pharmacoepidemiological cross‐sectional multicenter study. Eur Neuropsychopharmacol 2016;26:1960‐71. [DOI] [PubMed] [Google Scholar]
- 186. Taylor RW, Marwood L, Oprea E et al. Pharmacological augmentation in unipolar depression: a guide to the guidelines. Int J Neuropsychopharmacol 2020;23:587‐625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Strawbridge R, Carter B, Marwood L et al. Augmentation therapies for treatment‐resistant depression: systematic review and meta‐analysis. Br J Psychiatry 2019;214:42‐51. [DOI] [PubMed] [Google Scholar]
- 188. Scott F, Hampsey E, Gnanapragasam S et al. Systematic review and meta‐analysis of augmentation and combination treatments for early‐stage treatment‐resistant depression. J Psychopharmacol 2023;37:268‐78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Henssler J, Alexander D, Schwarzer G et al. Combining antidepressants vs antidepressant monotherapy for treatment of patients with acute depression: a systematic review and meta‐analysis. JAMA Psychiatry 2022;79:300‐12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Zhou Y, Wang C, Lan X, et al. The effectiveness of repeated intravenous ketamine on subjective and objective psychosocial function in patients with treatment‐resistant depression and suicidal ideation. J Affect Disord 2022;304:78‐84. [DOI] [PubMed] [Google Scholar]
- 191. McIntyre RS, Carvalho IP, Lui LMW et al. The effect of intravenous, intranasal, and oral ketamine in mood disorders: a meta‐analysis. J Affect Disord 2020;276:576‐84. [DOI] [PubMed] [Google Scholar]
- 192. Fava M, Freeman MP, Flynn M et al. Double‐blind, placebo‐controlled, dose‐ranging trial of intravenous ketamine as adjunctive therapy in treatment‐resistant depression (TRD). Mol Psychiatry 2020;25:1592‐603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. McIntyre RS, Rodrigues NB, Lee Y et al. The effectiveness of repeated intravenous ketamine on depressive symptoms, suicidal ideation and functional disability in adults with major depressive disorder and bipolar disorder: results from the Canadian Rapid Treatment Center of Excellence. J Affect Disord 2020;274:903‐10. [DOI] [PubMed] [Google Scholar]
- 194. Daly EJ, Singh JB, Fedgchin M et al. Efficacy and safety of intranasal esketamine adjunctive to oral antidepressant therapy in treatment‐resistant depression: a randomized clinical trial. JAMA Psychiatry 2018;75:139‐48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Wajs E, Aluisio L, Holder R et al. Esketamine nasal spray plus oral antidepressant in patients with treatment‐resistant depression: assessment of long‐term safety in a phase 3, open‐label study (SUSTAIN‐2). J Clin Psychiatry 2020;81:19m12891. [DOI] [PubMed] [Google Scholar]
- 196. Zheng W, Yang XH, Gu LM et al. Antianhedonic effects of serial intravenous subanaesthetic ketamine in anxious versus nonanxious depression. J Affect Disord 2022;313:72‐6. [DOI] [PubMed] [Google Scholar]
- 197. Nogo D, Jasrai AK, Kim H et al. The effect of ketamine on anhedonia: improvements in dimensions of anticipatory, consummatory, and motivation‐related reward deficits. Psychopharmacology 2022;239:2011‐39. [DOI] [PubMed] [Google Scholar]
- 198. Zheng W, Gu LM, Zhou YL et al. Association of VEGF with antianhedonic effects of repeated‐dose intravenous ketamine in treatment‐refractory depression. Front Psychiatry 2021;12:780975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Wilkowska A, Wiglusz MS, Gałuszko‐Wegielnik M et al. Antianhedonic effect of repeated ketamine infusions in patients with treatment resistant depression. Front Psychiatry 2021;12:704330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Dold M, Bartova L, Kasper S. Treatment response of add‐on esketamine nasal spray in resistant major depression in relation to add‐on second‐generation antipsychotic treatment. Int J Neuropsychopharmacol 2020;23:440‐5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Vázquez GH, Bahji A, Undurraga J et al. Efficacy and tolerability of combination treatments for major depression: antidepressants plus second‐generation antipsychotics vs. esketamine vs. lithium. J Psychopharmacol 2021;35:890‐900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Wilkinson ST, Rhee TG, Joormann J et al. Cognitive behavioral therapy to sustain the antidepressant effects of ketamine in treatment‐resistant depression: a randomized clinical trial. Psychother Psychosom 2021;90:318‐27. [DOI] [PubMed] [Google Scholar]
- 203. Reif A. Esketamine nasal spray improves short and long term outcomes compared with quetiapine extended release in patients with treatment resistant depression: First results from ESCAPE TRD, a randomised, multi centre phase IIIb clinical trial. Presented at the Congress of the German Society of Psychiatry and Psychotherapy, Berlin, November 2022. [Google Scholar]
- 204. Rodrigues NB, Siegel A, Lipsitz O et al. Effectiveness of intravenous ketamine in mood disorder patients with a history of neurostimulation. CNS Spectr 2022;27:315‐21. [DOI] [PubMed] [Google Scholar]
- 205. McIntyre RS, Rosenblat JD, Nemeroff CB et al. Synthesizing the evidence for ketamine and esketamine in treatment‐resistant depression: an international expert opinion on the available evidence and implementation. Am J Psychiatry 2021;178:383‐99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Schatzberg AF. A word to the wise about intranasal esketamine. Am J Psychiatry 2019;176:422‐4. [DOI] [PubMed] [Google Scholar]
- 207. Corya SA, Sanger TM, Van Campen LE et al. Olanzapine/fluoxetine combination for treatment‐resistant depression: a controlled study of SSRI and nortriptyline resistance. J Clin Psychiatry 2005;66:1289‐97. [DOI] [PubMed] [Google Scholar]
- 208. Corya SA, Williamson D, Sanger TM et al. A randomized, double‐blind comparison of olanzapine/fluoxetine combination, olanzapine, fluoxetine, and venlafaxine in treatment‐resistant depression. Depress Anxiety 2006;23:364‐72. [DOI] [PubMed] [Google Scholar]
- 209. 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;68:224‐36. [DOI] [PubMed] [Google Scholar]
- 210. 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;68:843‐53. [DOI] [PubMed] [Google Scholar]
- 211. Mahmoud RA, Pandina GJ, Turkoz I et al. Risperidone for treatment‐refractory major depressive disorder: a randomized trial. Ann Intern Med 2007;147:593‐602. [DOI] [PubMed] [Google Scholar]
- 212. McIntyre A, Gendron A, McIntyre A. Quetiapine adjunct to selective serotonin reuptake inhibitors or venlafaxine in patients with major depression, comorbid anxiety, and residual depressive symptoms: a randomized, placebo‐controlled pilot study. Depress Anxiety 2007;24:487‐94. [DOI] [PubMed] [Google Scholar]
- 213. 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;28:156‐65. [DOI] [PubMed] [Google Scholar]
- 214. Reeves H, Batra S, May RS et al. Efficacy of risperidone augmentation to antidepressants in the management of suicidality in major depressive disorder: a randomized, double‐blind, placebo‐controlled pilot study. J Clin Psychiatry 2008;69:1228‐36. [DOI] [PubMed] [Google Scholar]
- 215. Bauer M, Pretorius HW, Constant EL et al. Extended‐release quetiapine as adjunct to an antidepressant in patients with major depressive disorder: results of a randomized, placebo‐controlled, double‐blind study. J Clin Psychiatry 2009;70:540‐9. [DOI] [PubMed] [Google Scholar]
- 216. 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;14:197‐206. [DOI] [PubMed] [Google Scholar]
- 217. Keitner GI, Garlow SJ, Ryan CE et al. A randomized, placebo‐controlled trial of risperidone augmentation for patients with difficult‐to‐treat unipolar, non‐psychotic major depression. J Psychiatr Res 2009;43:205‐14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218. 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;13:917‐32. [DOI] [PubMed] [Google Scholar]
- 219. Fava M, Mischoulon D, Iosifescu D et al. A double‐blind, placebo‐controlled study of aripiprazole adjunctive to antidepressant therapy among depressed outpatients with inadequate response to prior antidepressant therapy (ADAPT‐A Study). Psychother Psychosom 2012;81:87‐97. [DOI] [PubMed] [Google Scholar]
- 220. 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. J Clin Psychiatry 2015;76:1232‐40. [DOI] [PubMed] [Google Scholar]
- 221. 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. J Clin Psychiatry 2015;76:1224‐31. [DOI] [PubMed] [Google Scholar]
- 222. Durgam S, Earley W, Guo H et al. Efficacy and safety of adjunctive cariprazine in inadequate responders to antidepressants: a randomized, double‐blind, placebo‐controlled study in adult patients with major depressive disorder. J Clin Psychiatry 2016;77:371‐8. [DOI] [PubMed] [Google Scholar]
- 223. Earley WR, Guo H, Németh G et al. Cariprazine augmentation to antidepressant therapy in major depressive disorder: results of a randomized, double‐blind, placebo‐controlled trial. Psychopharmacol Bull 2018;48:62‐80. [PMC free article] [PubMed] [Google Scholar]
- 224. Fava M, Durgam S, Earley W et al. Efficacy of adjunctive low‐dose cariprazine in major depressive disorder: a randomized, double‐blind, placebo‐controlled trial. Int Clin Psychopharmacol 2018;33:312‐21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Cha DS, McIntyre RS. Treatment‐emergent adverse events associated with atypical antipsychotics. Expert Opin Pharmacother 2012;13:1587‐98. [DOI] [PubMed] [Google Scholar]
- 226. 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;318:132‐45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Li H, Cui L, Li J et al. Comparative efficacy and acceptability of neuromodulation procedures in the treatment of treatment‐resistant depression: a network meta‐analysis of randomized controlled trials. J Affect Disord 2021;287:115‐24. [DOI] [PubMed] [Google Scholar]
- 228. Spurny‐Dworak B, Godbersen GM, Reed MB et al. The impact of theta‐burst stimulation on cortical GABA and glutamate in treatment‐resistant depression: a surface‐based MRSI analysis approach. Front Mol Neurosci 2022;15:913274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229. Holtzheimer PE, Husain MM, Lisanby SH et al. Subcallosal cingulate deep brain stimulation for treatment‐resistant depression: a multisite, randomised, sham‐controlled trial. Lancet Psychiatry 2017;4:839‐49. [DOI] [PubMed] [Google Scholar]
- 230. Bulteau S, Laurin A, Pere M et al. Intermittent theta burst stimulation (iTBS) versus 10 Hz high‐frequency repetitive transcranial magnetic stimulation (rTMS) to alleviate treatment‐resistant unipolar depression: a randomized controlled trial (THETA‐DEP). Brain Stimul 2022;15:870‐80. [DOI] [PubMed] [Google Scholar]
- 231. Chen L, Thomas EHX, Kaewpijit P et al. Accelerated theta burst stimulation for the treatment of depression: a randomised controlled trial. Brain Stimul 2021;14:1095‐105. [DOI] [PubMed] [Google Scholar]
- 232. Daskalakis ZJ, McClintock SM, Hadas I et al. Confirmatory Efficacy and Safety Trial of Magnetic Seizure Therapy for Depression (CREST‐MST): protocol for identification of novel biomarkers via neurophysiology. Trials 2021;22:906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Adu MK, Shalaby R, Chue P et al. Repetitive transcranial magnetic stimulation for the treatment of resistant depression: a scoping review. Behav Sci 2022;12:195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Kumar A, Bunker MT, Aaronson ST et al. Durability of symptomatic responses obtained with adjunctive vagus nerve stimulation in treatment‐resistant depression. Neuropsychiatr Dis Treat 2019;15:457‐68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Sackeim HA, Dibué M, Bunker MT et al. The long and winding road of vagus nerve stimulation: challenges in developing an intervention for difficult‐to‐treat mood disorders. Neuropsychiatr Dis Treat 2020;16:3081‐93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Kraus C, Quach D, Sholtes DM et al. Setting up a successful vagus nerve stimulation service for patients with difficult‐to‐treat depression. Neuromodulation 2022;25:316‐26. [DOI] [PubMed] [Google Scholar]
- 237. Zandi PP, Morreale M, Reti IM et al. National Network of Depression Centers' recommendations on harmonizing clinical documentation of electroconvulsive therapy. J ECT 2022;38:159‐64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Sackeim HA, Prudic J, Nobler MS et al. Effects of pulse width and electrode placement on the efficacy and cognitive effects of electroconvulsive therapy. Brain Stimul 2008;1:71‐83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Grammer GG, Kuhle AR, Clark CC et al. Severity of depression predicts remission rates using transcranial magnetic stimulation. Front Psychiatry 2015;6:114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Fitzgerald PB, Hoy KE, Anderson RJ et al. A study of the pattern of response to rTMS treatment in depression. Depress Anxiety 2016;33:746‐53. [DOI] [PubMed] [Google Scholar]
- 241. Kar SK. Predictors of response to repetitive transcranial magnetic stimulation in depression: a review of recent updates. Clin Psychopharmacol Neurosci 2019;17:25‐33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. Lisanby SH, Husain MM, Rosenquist PB et al. Daily left prefrontal repetitive transcranial magnetic stimulation in the acute treatment of major depression: clinical predictors of outcome in a multisite, randomized controlled clinical trial. Neuropsychopharmacology 2009;34:522‐34. [DOI] [PubMed] [Google Scholar]
- 243. Levkovitz Y, Isserles M, Padberg F et al. Efficacy and safety of deep transcranial magnetic stimulation for major depression: a prospective multicenter randomized controlled trial. World Psychiatry 2015;14:64‐73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Zemplényi A, Józwiak‐Hagymásy J, Kovács S et al. Repetitive transcranial magnetic stimulation may be a cost‐effective alternative to antidepressant therapy after two treatment failures in patients with major depressive disorder. BMC Psychiatry 2022;22:437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245. Zhao YJ, Tor PC, Khoo AL et al. Cost‐effectiveness modeling of repetitive transcranial magnetic stimulation compared to electroconvulsive therapy for treatment‐resistant depression in Singapore. Neuromodulation 2018;21:376‐82. [DOI] [PubMed] [Google Scholar]
- 246. Health Quality Ontario . Repetitive transcranial magnetic stimulation for treatment‐resistant depression: a systematic review and meta‐analysis of randomized controlled trials. Ont Health Technol Assess Ser 2016;16:1‐66. [PMC free article] [PubMed] [Google Scholar]
- 247. Hsu JH, Downar J, Vila‐Rodriguez F et al. Impact of prior treatment on remission with intermittent theta burst versus high‐frequency repetitive transcranial magnetic stimulation in treatment resistant depression. Brain Stimul 2019;12:1553‐5. [DOI] [PubMed] [Google Scholar]
- 248. Blumberger DM, Vila‐Rodriguez F, Thorpe KE et al. Effectiveness of theta burst versus high‐frequency repetitive transcranial magnetic stimulation in patients with depression (THREE‐D): a randomised non‐inferiority trial. Lancet 2018;391:1683‐92. [DOI] [PubMed] [Google Scholar]
- 249. Cole EJ, Phillips AL, Bentzley BS et al. Stanford Neuromodulation Therapy (SNT): a double‐blind randomized controlled trial. Am J Psychiatry 2022;179:132‐41. [DOI] [PubMed] [Google Scholar]
- 250. Daskalakis ZJ, Tamminga C, Throop A et al. Confirmatory Efficacy and Safety Trial of Magnetic Seizure Therapy for Depression (CREST‐MST): study protocol for a randomized non‐inferiority trial of magnetic seizure therapy versus electroconvulsive therapy. Trials 2021;22:786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. Tang VM, Blumberger DM, Throop A et al. Continuation magnetic seizure therapy for treatment‐resistant unipolar or bipolar depression. J Clin Psychiatry 2021;82:20m13677. [DOI] [PubMed] [Google Scholar]
- 252. Jiang J, Zhang C, Li C et al. Magnetic seizure therapy for treatment‐resistant depression. Cochrane Database Syst Rev 2021;6:CD013528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Hitti FL, Cristancho MA, Yang AI et al. Deep brain stimulation of the ventral capsule/ventral striatum for treatment‐resistant depression: a decade of clinical follow‐up. J Clin Psychiatry 2021;82:21m13973. [DOI] [PubMed] [Google Scholar]
- 254. Holtzheimer PE, Kelley ME, Gross RE et al. Subcallosal cingulate deep brain stimulation for treatment‐resistant unipolar and bipolar depression. Arch Gen Psychiatry 2012;69:150‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255. Dougherty DD, Rezai AR, Carpenter LL et al. A randomized sham‐controlled trial of deep brain stimulation of the ventral capsule/ventral striatum for chronic treatment‐resistant depression. Biol Psychiatry 2015;78:240‐8. [DOI] [PubMed] [Google Scholar]
- 256. Kisely S, Li A, Warren N et al. A systematic review and meta‐analysis of deep brain stimulation for depression. Depress Anxiety 2018;35:468‐80. [DOI] [PubMed] [Google Scholar]
- 257. Zhou C, Zhang H, Qin Y et al. A systematic review and meta‐analysis of deep brain stimulation in treatment‐resistant depression. Prog Neuropsychopharmacol Biol Psychiatry 2018;82:224‐32. [DOI] [PubMed] [Google Scholar]
- 258. Ramasubramanian V, Mathumathi S, Rajendhiran G et al. A comparative study of the effect of electroconvulsive therapy and transcranial direct current stimulation in the treatment of persons suffering from treatment‐resistant depression. Ind Psychiatry J 2022;31:68‐73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259. Rhee TG, Shim SR, Forester BP et al. Efficacy and safety of ketamine vs electroconvulsive therapy among patients with major depressive episode: a systematic review and meta‐analysis. JAMA Psychiatry 2022;79:1162‐72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260. McHugh RK, Whitton SW, Peckham AD et al. Patient preference for psychological vs pharmacologic treatment of psychiatric disorders: a meta‐analytic review. J Clin Psychiatry 2013;74:595‐602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261. Furukawa TA, Shinohara K, Sahker E et al. Initial treatment choices to achieve sustained response in major depression: a systematic review and network meta‐analysis. World Psychiatry 2021;20:387‐96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262. Boschloo L, Bekhuis E, Weitz ES et al. The symptom‐specific efficacy of antidepressant medication vs. cognitive behavioral therapy in the treatment of depression: results from an individual patient data meta‐analysis. World Psychiatry 2019;18:183‐91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263. Nemeroff CB, Heim CM, Thase ME et al. Differential responses to psychotherapy versus pharmacotherapy in patients with chronic forms of major depression and childhood trauma. Proc Natl Acad Sci USA 2003;100:14293‐6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264. Yrondi A, Aouizerate B, Bennabi D et al. Childhood maltreatment and clinical severity of treatment‐resistant depression in a French cohort of outpatients (FACE‐DR): one‐year follow‐up. Depress Anxiety 2020;37:365‐74. [DOI] [PubMed] [Google Scholar]
- 265. Ijaz S, Davies P, Williams CJ et al. Psychological therapies for treatment‐resistant depression in adults. Cochrane Database Syst Rev 2018;5:CD010558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266. Bartova L, Fugger G, Dold M et al. Combining psychopharmacotherapy and psychotherapy is not associated with better treatment outcome in major depressive disorder – evidence from the European Group for the Study of Resistant Depression. J Psychiatr Res 2021;141:167‐75. [DOI] [PubMed] [Google Scholar]
- 267. Markowitz JC, Wright JH, Peeters F et al. The neglected role of psychotherapy for treatment‐resistant depression. Am J Psychiatry 2022;179:90‐3. [DOI] [PubMed] [Google Scholar]
- 268. Eisendrath SJ, Gillung E, Delucchi KL et al. A randomized controlled trial of mindfulness‐based cognitive therapy for treatment‐resistant depression. Psychother Psychosom 2016;85:99‐110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269. Fonagy P, Rost F, Carlyle JA et al. Pragmatic randomized controlled trial of long‐term psychoanalytic psychotherapy for treatment‐resistant depression: the Tavistock Adult Depression Study (TADS). World Psychiatry 2015;14:312‐21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270. Hauksson P, Ingibergsdóttir S, Gunnarsdóttir T et al. Effectiveness of cognitive behaviour therapy for treatment‐resistant depression with psychiatric comorbidity: comparison of individual versus group CBT in an interdisciplinary rehabilitation setting. Nord J Psychiatry 2017;71:465‐72. [DOI] [PubMed] [Google Scholar]
- 271. McPherson S, Cairns P, Carlyle J et al. The effectiveness of psychological treatments for treatment‐resistant depression: a systematic review. Acta Psychiatr Scand 2005;111:331‐40. [DOI] [PubMed] [Google Scholar]
- 272. van Bronswijk S, Moopen N, Beijers L et al. Effectiveness of psychotherapy for treatment‐resistant depression: a meta‐analysis and meta‐regression. Psychol Med 2019;49:366‐79. [DOI] [PubMed] [Google Scholar]
- 273. Reiff CM, Richman EE, Nemeroff CB et al. Psychedelics and psychedelic‐assisted psychotherapy. Am J Psychiatry 2020;177:391‐410. [DOI] [PubMed] [Google Scholar]
- 274. Goodwin GM, Aaronson ST, Alvarez O et al. Single‐dose psilocybin for a treatment‐resistant episode of major depression. N Engl J Med 2022;387:1637‐48. [DOI] [PubMed] [Google Scholar]
- 275. Siegel AN, Meshkat S, Benitah K et al. Registered clinical studies investigating psychedelic drugs for psychiatric disorders. J Psychiatr Res 2021;139:71‐81. [DOI] [PubMed] [Google Scholar]
- 276. Baune BT, Sampson E, Louise J et al. No evidence for clinical efficacy of adjunctive celecoxib with vortioxetine in the treatment of depression: a 6‐week double‐blind placebo controlled randomized trial. Eur Neuropsychopharmacol 2021;53:34‐46. [DOI] [PubMed] [Google Scholar]
- 277. Crossley NA, Bauer M. Acceleration and augmentation of antidepressants with lithium for depressive disorders: two meta‐analyses of randomized, placebo‐controlled trials. J Clin Psychiatry 2007;68:935‐40. [DOI] [PubMed] [Google Scholar]
- 278. Zazula R, Husain MI, Mohebbi M et al. Minocycline as adjunctive treatment for major depressive disorder: pooled data from two randomized controlled trials. Aust N Z J Psychiatry 2021;55:784‐98. [DOI] [PubMed] [Google Scholar]
- 279. Walker AJ, Kim Y, Borissiouk I et al. Statins: neurobiological underpinnings and mechanisms in mood disorders. Neurosci Biobehav Rev 2021;128:693‐708. [DOI] [PubMed] [Google Scholar]
- 280. Nettis MA, Lombardo G, Hastings C et al. Augmentation therapy with minocycline in treatment‐resistant depression patients with low‐grade peripheral inflammation: results from a double‐blind randomised clinical trial. Neuropsychopharmacology 2021;46:939‐48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281. Liu Y, Zhou X, Zhu D et al. Is pindolol augmentation effective in depressed patients resistant to selective serotonin reuptake inhibitors? A systematic review and meta‐analysis. Hum Psychopharmacol 2015;30:132‐42. [DOI] [PubMed] [Google Scholar]
- 282. Lee HH, Blumberger DM, Lenze EJ et al. Low‐dose augmentation with buprenorphine for treatment‐resistant depression: a multisite randomized controlled trial with multimodal assessment of target engagement. Biol Psychiatry Glob Open Sci 2022;2:127‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283. Leavy MB, Boussios C, Phillips RL Jr et al. Outcome measure harmonization and data infrastructure for patient‐centered outcomes research in depression: final report. Rockville: Agency for Healthcare Research and Quality, 2022. [PubMed] [Google Scholar]
- 284. Zimmerman M, Balling C, Chelminski I et al. Applying the inclusion/exclusion criteria in placebo‐controlled studies to a clinical sample: a comparison of medications. J Affect Disord 2020;260:483‐8. [DOI] [PubMed] [Google Scholar]
- 285. McIntyre RS, Ismail Z, Watling CP et al. Patient‐reported outcome measures for life engagement in mental health: a systematic review. J Patient Rep Outcomes 2022;6:62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286. Costa T, Menzat B, Engelthaler T et al. The burden associated with, and management of, difficult‐to‐treat depression in patients under specialist psychiatric care in the United Kingdom. J Psychopharmacol 2022;36:545‐56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287. Leichsenring F, Steinert C, Rabung S et al. The efficacy of psychotherapies and pharmacotherapies for mental disorders in adults: an umbrella review and meta‐analytic evaluation of recent meta‐analyses. World Psychiatry 2022; 21:133‐45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288. McKeown L, Taylor RW, Day E et al. Patient perspectives of lithium and quetiapine augmentation treatment in treatment‐resistant depression: a qualitative assessment. J Psychopharmacol 2022;36:557‐65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289. Smith‐Apeldoorn SY, Veraart JK, Spijker J et al. Maintenance ketamine treatment for depression: a systematic review of efficacy, safety, and tolerability. Lancet Psychiatry 2022;9:907‐21. [DOI] [PubMed] [Google Scholar]
- 290. Ragguett RM, Tamura JK, McIntyre RS. Keeping up with the clinical advances: depression. CNS Spectr 2019;24(Suppl. 1):25‐37. [DOI] [PubMed] [Google Scholar]
- 291. Torous J, Bucci S, Bell IH et al. The growing field of digital psychiatry: current evidence and the future of apps, social media, chatbots, and virtual reality. World Psychiatry 2021;20:318‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292. Chekroud AM, Bondar J, Delgadillo J et al. The promise of machine learning in predicting treatment outcomes in psychiatry. World Psychiatry 2021;20:154‐70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293. Lee Y, Ragguett RM, Mansur RB et al. Applications of machine learning algorithms to predict therapeutic outcomes in depression: a meta‐analysis and systematic review. J Affect Disord 2018; 241:519‐32. [DOI] [PubMed] [Google Scholar]
- 294. Venkatesan A, Forster B, Rao P et al. Improvements in depression outcomes following a digital cognitive behavioral therapy intervention in a polychronic population: retrospective study. JMIR Form Res 2022;6:e38005. [DOI] [PMC free article] [PubMed] [Google Scholar]