139
Views
2
CrossRef citations to date
0
Altmetric
Review

Neuromodulation therapies and treatment-resistant depression

&
Pages 53-65 | Published online: 13 Jul 2012

Abstract

Background

Patients with treatment-resistant depression (TRD) who showed partial response to pharmacological and psychotherapeutic interventions need a trial of neuromodulation therapies (NTs).

Objective

This paper aims to review evidence-based data on the use of NTs in TRD.

Method

Using keywords and combined-word strategy, multiple computer searches of PubMed, Google Scholar, Quertle(R), and Medline were conducted for retrieving relevant articles published in English-language peer-reviewed journals (2000–2012). Those papers that addressed NTs in TRD were retained for extensive review.

Results

Despite methodological challenges, a range of 30%–93% of TRD patients showed substantial improvement to one of the NTs. One hundred–percent improvement was reported in two single-case studies on deep brain stimulation. Some studies reported no benefits from transcranial direct current stimulation. NTs were reported to have good clinical efficacy, better safety margin, and benign side-effect profile. Data are limited regarding randomized clinical trials, long-term efficacy, and cost-effectiveness of these approaches. Both modified electroconvulsive therapy and magnetic seizure therapy were associated with reversible but disturbing neurocognitive adverse effects. Besides clinical utility, NTs including approaches on the horizon may unlock the biological basis underlying mood disorders including TRD.

Conclusion

NTs are promising in patients with TRD, as the majority of them show good clinical response measured by standardized depression scales. NTs need further technological refinements and optimization together with continuing well-designed studies that recruit larger numbers of participants with TRD.

Introduction

It is estimated that depression afflicts about 121 million people worldwide. Major depression (MD) is the main cause of disability and the fourth-leading contributor to the global burden of disease. By the year 2020, MD is projected to reach second place in the ranking of disability-adjusted life years. Trials of available antidepressant medications alone or combined with psychotherapies are effective for 60%–80% of those affected with MD.Citation1 Conversely, up to 40% of patients with MD do not show satisfactory improvement attributable to multiple biopsychosocial factors. At its worst, MD can lead to suicide, and as a consequence about 850,000 lives are lost every year.Citation2

Treatment-resistant depression (TRD) evades universal definition; however, a poor response to two adequate (optimal dosage and 6–12 weeks duration) trials of two different classes of antidepressants has been proposed as its operational characterization.Citation3 Researchers have categorized TRD in accordance to antidepressant trials: stage 0, has not had a single adequate trial of medication; stage 1, failure of an adequate trial of one class of an antidepressant that is monotherapy; stage 2, failure of adequate trials of two distinctly different classes – that is, selective serotonin reuptake inhibitors (SSRIs) and tricyclic antidepressants – of antidepressant, involving two monotherapy trials; stage 3, stage 2 plus failure to respond to one augmentation strategy of lithium or thyroid augmentation of one of the monotherapies; stage 4, stage 3 plus a failure to a second augmentation strategy in terms of monoamine oxidase inhibitors; and stage 5, stage 4 plus failure of an adequate course of ECT.Citation4 There are other staging methods of TRD.Citation5 These staging methods help researchers and clinicians to understand TRD patients and accordingly plan interventions for enhancing the response, remission rate, and quality of life. However, TRD continues to challenge mental health care providers despite the understanding of psychosocial and biological markers and psychopharmacology of mood disorders and also the availability of multiple therapeutic options including optimization, switching, and combination of antidepressants. Notably, currently there is an increasing interest in the utilization of several neuromodulation therapies (NTs) in the management of patients with TRD.Citation6 This is because psychopharmacological therapy exposes the entire body to a potentially therapeutic substance in order to treat a relatively small region of the brain, whereas NTs are designed to target specific brain circuits that are important in the pathogenesis of MD. Additionally, NTs are not systemic and, therefore, the side-effect profile is limited and different from medications, and there are minimal, if any, drug interactions.Citation7 Furthermore, evidence-based data has been emerging continuously about FDA-approved and yet-to-be-approved NTs in the TRD population over the past decade. This paper summarizes these data on the role of NTs in TRD patients.

Search method

Multiple computer searches were conducted using PubMed, Google Scholar, Quertle(R), and Medline databases for the years 2000–2012. A number of keywords were used: treatment-resistant depression, treatment-refractory depression, partial-response depression, nonresponse depression, neuromodulation techniques, neurostimulation approaches, and somatic therapies. These words were combined with modified electroconvulsive therapy (mECT), repetitive transcranial magnetic stimulation (rTMS), vagus nerve stimulation (VNS), magnetic seizure therapy (MST), deep brain stimulation (DBS), transcranial direct current stimulation, cranial electric stimulation (CES), epidural cortical stimulation (ECS), focused ultrasound (FUS), near-infrared light therapy (NIR), low-field magnetic stimulation (LFMS), and optogenetic stimulation (OS) for a second round of computer searches. A third round of searches included words such as mechanisms, brain areas involved, and outcomes combined with aforesaid therapies. As a corollary, relevant articles published in English-language peer-reviewed journals were retrieved. Only clinical trials, systematic reviews, and meta-analyses that addressed TRD and NTs were retained for extensive review and inclusion in this study. Some exceptions were made with regard to some unique case reports, open and controlled studies, and small and large case series describing usefulness of NTs in patients with TRD and MD. Studies addressing non-TRD populations were excluded from this review. Similarly, studies focusing on neurosurgical ablation approaches in TRD populations were not considered for inclusion. References of selected articles were also reviewed for identifying relevant TRD trials, which were also included in this review. A couple of important TRD studies conducted prior to 2000 were also included.

Categorization of NTs

NTs for neuropsychiatric disorders including MD are categorized into the following: (1) seizure therapies, including mECT and MST, (2) noninvasive therapies, including rTMS, TDCS, and CES, (3) neurosurgical approaches, including VNS, ECS, and DBS, and (4) new approaches on the horizon, including FUS, NIR, LFMS, and OS.Citation8 Another category represents neurosurgical ablation therapies, including cingulotomy and limbic leucotomy used in TRD. Such technical details as invasiveness, anesthesia needed, seizures induced, target related to deep brain structures, contactness, stimulation being focal or generalized and form of stimulation of each neuromodulation therapy are presented in .

Table 1 Technical information of neuromodulation therapies

Mechanisms of action of NTs

There is an increasing focus on exploring biomarkers underlying the pathogenesis of mood disordersCitation9 that help in the development of new drugs and NTs. In several related studies, overactive subcallosal cingulate gyrus (SCG) glucose metabolism has been reported in MD that is reduced with successful antidepressant therapies.Citation10 Interestingly, DBS is reported to modulate neural pathways linked with SCG in relieving MD.Citation8,Citation11,Citation12 According to some studies, antidepressant effects were also found when DBS targeted ventral capsule/ventral striatum (VC/VS) in patients with severe obsessive-compulsive disorder (OCD) and MD.Citation13,Citation14 In a study of single patients with dystonia suffering from depression, DBS of globus pallidus internus (GPI) showed improvement in dystonia but also showed antidepressant effects through modulation of mesolimbic dopamine pathways.Citation15 In another study, also of single patients with tardive dyskinesia (TD) and MD, DBS brought about improvement in depressive mood.Citation16 Other studies have also reported improvement in both depression and TD after DBS of the inferior thalamic peduncle (ITP), which modulates orbitofrontal cortex hyperactivity.Citation17,Citation18 Bewernick and colleagues reported that DBS of the nucleus accumbens (NAc) was associated with decreased ratings of depression and anxiety in TRD patients.Citation19

Rush and colleaguesCitation20 noticed antidepressant effects when VNS was used for epilepsy. VNS modulates neural pathways associated with mood regulation: the nucleus tractus solitaries, raphe nucleus, and locus ceruleus.Citation21 In fact, the VNS device stimulates left cervical vagus nerve containing afferent neurons tracking through the brain stem to cortical and subcortical networks.Citation20Citation23 Furthermore, some neurobiological studies reported disruptions in right and left dorsolateral prefrontal cortex (R/LDLPFC) in mood disorders. Also, rTMS of R/LDLPFC results in antidepressive effects coupled with increasing cerebral blood supply to this brain areas.Citation23Citation25 Certainly, NTs target more specific, localized regions in the brain, which are somehow dysfunctional in MD. It remains uncertain how the depression is relieved; this is yet to be understood well, and hence basic neurobiological studies are needed. Similarly with regard to ECT, though no exact mechanism is known, debate and research continues in this field.Citation26Citation30

Electroconvulsive therapy

Modified ECT has been used extensively in psychotic depression, schizophrenia, mania, and other mental disorders. It requires light anesthesia and is a recognized mode of treatment for TRD.Citation31,Citation32 It remains the most effective therapy in TRD patients with a response rate of 50%–70%, though the strength of recommendation of ECT is C.Citation33,Citation34 It targets nonspecific, broad regions of the cortex, and its mechanism of action is elusive. Notably, high post-ECT relapse rate and safety profile are of great concern for TRD patients and health providers as well. In a study of patients with nonpsychotic MD that tested whether pre-ECT medication resistance is associated with post-ECT relapse rates, it was observed that 34.6% of nonmedication-resistant patients who were not exposed to at least one antidepressant medication trial relapsed, while 50.0% of medication-resistant patients relapsed, a difference that was not statistically significant but clinically relevant.Citation35 Furthermore, in the first week after acute remission, 9.8% of patients not having at least one antidepressant medication trial met relapse criteria, while 31.4% of medication-resistant patients met relapse criteria, a difference that was statistically significant. It was concluded that MD patients who have had at least one adequate antidepressant medication trial or no such trial before ECT may be especially prone to early relapse after successful acute remission with mECT.Citation35 Research is needed to develop strategies in order to prevent relapse following successful ECT in MD, which may be maintenance ECT and a combination of pharmacotherapy and mECT.

Furthermore, it is also important to identify the predictors of nonresponse to mECT. In a large sample of patients with TRD, mECT was effective in 66% of patients. mECT nonresponse was associated with bipolar subtype, mixed features, slightly less severe depressive symptoms, and longer duration of the depressive episode.Citation36 In another study that aimed to investigate whether the clinical course of TRD patients following a course of mECT might be associated with changes of plasma brain-derived neurotrophic factor (BDNF) concentrations, it was shown that at baseline, plasma BDNF levels of patients were significantly lower than those of control subjects, and those after ECT were significantly increased in parallel with the decrease of the Hamilton Depression Rating Scale (HDRS) total score. Only remitter patients who showed higher baseline BDNF levels than nonremitters reached normalized BDNF levels after mECT. These findings suggested the potential usefulness of baseline plasma BDNF levels as predictors of response to mECT in TRD patients.Citation37 In an earlier study of 18 patients with TRD, levels of BDNF and 3-methoxy-4-hydroxyphenylglycol but not homovanillic acid were increased following mECT in responders, which suggested that dopamine and BDNF might be involved in the mechanism of action of mECT.Citation38 In a recent study of adolescents with TRD, both continuation and maintenance of mECT were useful and safe for selected adolescents with severe TRD, and symptom remission was achieved without experiencing cognitive impairment;Citation39 the latter is a surprising finding and needs replication studies. Interestingly, in another development, data support the use of ketamine as anesthetic agent prior to ECT for increasing its antidepressant effect as compared to propofol. In a related study, 31 inpatients with TRD underwent eight mECT sessions for 4 weeks. The HDRS was used to evaluate these patients before ECT and after the completion of the second, fourth, sixth, and eighth ECT sessions. The HDRS scores improved earlier in the ketamine group, with decreases in HDRS scores that were significantly greater in the ketamine group. The implication of this finding is that the symptoms of MD might be alleviated rapidly if ketamine anesthesia is used in TRD patients during ECT.Citation40

A retrospective evaluation of 5482 ECT treatments in 455 patients with TRD found therapeutic advantages in combination therapies versus ECT. A total of 18.2% of treatments were ECT monotherapy, 8.87% were done with one antidepressant. Results revealed that seizure duration was unaffected by most antidepressants, but SSRI caused a lengthened seizure activity. Postictal suppression was lower in mirtazapine and higher in SSRI and SNRI-treated patients. A significant enhancement of therapeutic effectiveness was seen in the patient group receiving tricyclics, SSRI, or mirtazapine, with no serious adverse events. This study supported the use of mirtazapine in enhancing the therapeutic effectiveness of ECT. Baghai and colleagues suggested that controlled studies are necessary to investigate further the possible advantages of ECT and pharmacotherapy combinations, especially the use of modern dual-acting antidepressants, which also have proven their efficacy in TRD.Citation41 Although mECT is effective in TRD, it significantly produces transient confusion, anterograde amnesia, and retrograde amnesia. Therefore, scientists have focused attention on technological refinements in ECT and also developing techniques that do not cause cognitive impairment and at the same time remain effective in MD and TRD.Citation42Citation46

Repetitive transcranial magnetic stimulation

The FDA has approved rTMS for the treatment of MD and TRD in adolescents and adults. It is a noninvasive technique with good efficacy in TDR. Its other indications include chronic pain, movement disorders, stroke, epilepsy, tinnitus, and other psychiatric disorders. Notably, rTMS is safer on long-term use and acts more selectively than mECT on brain areas implicated in the pathogenesis of MD.Citation47,Citation48 The rTMS has two forms: high-frequency rapid (HFR) (>1 Hz) and low-frequency slow (LFS) (≤1 Hz). Furthermore, HFR rTMS is preferred over LFS sychronized TMS, as the former was associated with more antidepressant effects in depressed patients as reflected by significant increases in blood supply to prefrontal cortical and limbic regions.Citation23 A sequential bilateral rTMS (LF right [LFR] then HF left [HFL]) is also effective in TRD patients but not more effective than unilateral HFL rTMS.Citation49,Citation50

In an open-label study, 21 patients who failed two antidepressant trials were given rTMS (HF, 10 Hz and intensity of 110%) for 4 weeks, keeping the dose of preexisting antidepressants unchanged. The majority of patients (n = 19) completed the 4-week study and were assessed. In intention-to-treat analysis, the mean HDRS scores were reduced from 30.80 ± 5.00 to 19.00 ± 6.37. No patient discontinued rTMS due to adverse effects, including headache, which was reported by 16% of patients. The study indicated the potential utility of rTMS as an augmenting agent in TRD.Citation51 Like LFR then HFL sequential bilateral rTMS, HFL and LFR unilateral rTMS are also efficacious in TRD. In a 6-week double-blind, randomized, sham-controlled trial in 50 patients with TRD, three trains of LF rTMS to the right prefrontal cortex of 140 seconds’ duration at 1 Hz were applied daily, followed immediately by 15 trains of 5 seconds’ duration of HFL rTMS at 10 Hz. Sham stimulation was applied with the coil angled at 45° from the scalp. The primary outcome variable was the score on the Montgomery–Åsberg Depression Rating Scale (MADRS). According to this study, there was a significantly greater response to active than sham stimulation at 2 weeks and across the full duration of the study. A significant proportion of the study group receiving active treatment met response (44%) or remission (36%) criteria by study end compared to the sham stimulation group (8%), and none remitted (0%). It was noted that sequentially applying both HFL rTMS and LFR rTMS to the right prefrontal cortex resulted in substantial improvement in patients with TRD. Furthermore, the treatment response accumulated to a clinically meaningful level over 4–6 weeks of active treatment.Citation23 In another controlled investigation, patients with TRD were randomized to receive 15 sessions of active or sham rTMS delivered to the LDLPFC at 110% the estimated prefrontal cortex threshold. Each session consisted of 32 trains of 10-Hz rTMS delivered in 5-second trains. The results showed response rate (≥50% decrease in HDRS score) for the rTMS group was 30.6%, significantly greater than the 6.1% rate in the sham group. The remission rate (an HDRS score < 8) for the rTMS group was 20%, significantly greater than the 3% rate in the sham group. The authors concluded that rTMS to LDLPFC can produce statistically and clinically significant antidepressant effects in patients with TRD.Citation25

In another study, subjects between the ages of 18 and 85 years were recruited from a tertiary care university hospital. Seventy-four subjects with TRD and an HDRS score > 21 were randomized to receive unilateral, bilateral, or sham rTMS. According to this study, the remission rate was significantly higher in the bilateral group than the sham group, but the remission rate in the unilateral group did not differ from either group. These findings warrant larger controlled studies that compare the efficacy of sequential bilateral rTMS and HFL/LFR rTMS in MD and TRD.Citation50,Citation52 From a safety perspective, rTMS can rarely induce accidental seizures, especially among patients with brain insult and on medications that reduce seizure threshold. However, this major side effect could be curtailed if expert guidelines are followed.Citation53,Citation54 Over the past 10 years, a number of meta-analyses of rTMS efficacy studies were conducted and the summary of these studies is as follows: a minimum of five to a maximum of 33 studies included; almost all included studies except one focused on depression rather than TRD; rTMS was more effective than sham rTMS; quality of studies improved successively; and rTMS designs also improved and effect size of rTMS was comparable to antidepressant drugs.Citation55Citation60 Finally, Moreines and colleaguesCitation61 have reviewed the neuropsychological effects of somatic therapies including rTMS that were associated with reversible mild reductions in sustained attention, spatial planning, and verbal retention.

Vagus nerve stimulation

The FDA approved the use of VNS in patients with MD and TRD in 2005.Citation62,Citation63 VNS principally stimulates the left cervical vagus nerve with a programmable neurostimulator. Observations of mood elevation during VNS for resistant epilepsy have suggested its potential role in TRD.Citation21,Citation22,Citation64 VNS targets the nucleus tractus solitarius, frontolimbic network, the locus ceruleus, and dorsal raphe nucleus, which regulate mood. Notably, initial studies on VNS reported inconsistent findings regarding reduced metabolism and blood flow in targeted brain networks with no putative antidepressant mechanism.Citation21,Citation63,Citation65 Similarly, a multicenter study on VNS found no significant reductions in depression scores for the experimental group as a whole, but antidepressant responses were observed among 40% of 30 recruited patients with TRD.Citation20 However, subsequent studies on VNS reported positive results. In a naturalistic, 1-year, follow-up study of 30 TRD patients who received VNS, the results were as follows: response rate of 40%–46% was sustained and the remission rate significantly increased, from 17% to 29% with an additional 9 months of long-term VNS. It was concluded that long-term VNS was associated with sustained benefit linked with good functional status.Citation66 Another naturalistic study with 2 years’ follow-up of 74 European patients with TRD showed a significant reduction at all the three time points, ie, 3, 12, and 24 months of VNS in the HDRS scores. After 2 years, 53.1% of the patients responded well, and 38.9% fulfilled the remission criteria. The proportion of patients with remission remained constant as the duration of VNS increased, with no concomitant antidepressant medication significant impact. This 2-year open-label trial of VNS suggested a clinical response and a benign adverse-effect profile among patients with TRD.Citation67 In a recent study of 15 consecutive outpatients with TRD, VNS significantly decreased Beck Depression Inventory (BDI) scores compared to baseline at 6 and 12 months, from a mean of 37.8 ± 7.8 before VNS activation to a mean of 24.6 ± 11.4 at 12 months. By 1 year, 28.6% of patients responded to VNS and 7.1% remitted. HDRS showed similar improvement at 1 year, with a 43% response rate and 14.3% remission rate. Reported side effects of VNS in decreasing frequency were hoarseness, dyspnea, nausea, pain, and anxiety, and no patient terminated treatment due to side effects. According to this study, a substantial minority of patients with TRD benefited from VNS.Citation68 VNS also induces cough, neck or jaw pain, and rarely infection. But it has no adverse neuropsychological effects.Citation61 In a study of single patients, VNS produced good results, with cost savings over mECT.Citation69 According to a systematic review, VNS examined in four clinical trials with 355 patients demonstrated steadily increasing improvement with full benefit after 6–12 months, sustained up to 2 years. But the primary results of the only controlled trial were negative and attributed to small sample size. Further controlled studies with large sample size are warranted to establish its efficacy and tolerability in future.Citation62,Citation70 The issue of predictors of response to VNS is addressed sparsely. In an open-label study of TRD, the predictors of response to VNS were history of resistant depression, mild to moderate resistant depression, not-severe resistant depression, and no history of use of ECT.Citation71 Trials of VNS in combination with pharmacotherapy are also needed in TRD populations.

Transcranial direct current stimulation

Transcranial direct current stimulation, a noninvasive technique with no FDA approval, has been used in patients with MD with mixed results. TDCS of the prefrontal cortex has been proposed as a therapeutic intervention in MD.Citation72,Citation73 In a parallel-group, double-blind clinical trial, 40 patients with MD who were medication-free were randomized into three groups. They were assessed by a blind rater using HDRS and BDI after ten sessions of TDCS during a 2-week period. According to this investigation, significantly larger reductions in depression scores after DLPFC TDCS were observed as compared to occipital and sham TDCS. Moreover, the beneficial effects of TDCS in the DLPFC group persisted for 1 month after the end of treatment. The authors suggested further investigation on the effects of TDCS for the treatment of MD.Citation72 Another double-blind, randomized study tested TDCS in 40 depressed participants and used the following parameters: 1-mA current strength, five treatment sessions, active or sham, and given on alternate days. Anodal stimulation was centered over the left DLPFC, with the cathode placed on the lateral aspect of the contralateral orbit. TDCS was continued up to a total of ten active sessions per participant. Overall, depression scores improved significantly over ten TDCS treatments, but there was no between-group difference in the five-session, sham-controlled phase. According to this study,Citation73 TDCS was found to be safe, with no adverse effects on a variety of assessed neuropsychological functions.Citation61 It was recommended that the efficacy of TDCS in MD be further evaluated over a longer treatment period, using enhanced stimulation parameters.Citation73

In another study, 22 patients with TRD were randomly assigned to a crossover protocol comparing TDCS and placebo stimulation add-on to a stable antidepressant medication. The parameters of active TDCS were 1 or 2 mA for 20 minutes/day, anode over the left DLPFC, and cathode over the contralateral supraorbital region. Active and placebo TDCS were applied for 2 weeks using indistinguishable DC stimulators. The results showed that there was no significant difference in depression scores after 2 weeks of real compared with 2 weeks of sham TDCS. In contrast, subjective mood ratings showed an increase in positive emotions after real TDCS compared with sham TDCS. Anodal TDCS, applied for 2 weeks, was not superior to placebo stimulation in patients with TRD. The authors suggested that modified and improved TDCS protocols should be carried out in controlled trials to develop TDCS with better efficacy in TRD.Citation74 All aforementioned studies except oneCitation74 addressed the usefulness of TDCS in MD, and hence more controlled trials are needed in TRD patients.

Deep brain stimulation

Deep brain stimulation, yet to be approved by the FDA, is a reversible invasive technique that involves stereotactical implantation of electrodes powered by a pulse generator into the specific dysfunctional brain regions implicated in mood disorders, Parkinson’s disease, Alzheimer’s disease, movement disorders, and other neuropsychiatric disorders. High frequency DBS of motor, mood, and cognitive neuronal circuits is reported to improve these conditions.Citation75 DBS therapy, dose- and site-dependent, is a less invasive and less extreme alternative to ablative psychosurgeries.Citation76 Research data supports DBS that targets cortico-striatal-pallido-thalamocortical loop, the VC/VS, and other neuronal networks in patients with MD, TRD, OCD, and Tourette’s syndrome.Citation77Citation81 Additionally, NAc that contains dopamine, a reward system and involved in the pathogenesis of MD, is a promising target for DBS. In a study, ten patients with severe TRD were implanted with bilateral DBS electrodes in the NAc. Twelve months later, five patients reached 50% reduction of the HDRS score, with significantly increased pleasure activities. Furthermore, the [18F]-2-fluoro-2-deoxy-d-glucose positron emission tomography data revealed that DBS decreased metabolism in the SCG, orbital prefrontal cortex, and amygdala. This study supported antidepressant and antianhedonic effects of DBS in patients with TRD. However, the small sample size limits the interpretation of results, and further research recruiting larger samples is needed.Citation82 In a multicenter study of 21 TRD patients who received DBS, it was found that patients treated with SCG DBS had variable response with time: 57% at 1 month, 48% at 6 months, and 29% at 12 months. The response rate after 12 months of DBS increased to 62% when redefined as a reduction in the baseline HRSD of 40% or more. Additionally, reductions in depressive symptoms were associated with amelioration in disease severity in patients who responded to surgery. Overall, this study corroborated the results of other research that the outcome of SCG DBS may be replicated across multiple centers.Citation83

In two influential review articles, researchers have provided greater details of somatic treatments in terms of target structures, motivation, response rates, mechanism of action, and technical issues.Citation8,Citation9 Accordingly, somatic therapies targeted SCG, VC/VS, left cervical vagus nerve, R/L DLPFC, GPI, lateral habenula, and ITP in MD and TRD patients, and improvement reported ranged from 30.6% to 66.7%.Citation8,Citation10,Citation12,Citation14 ().

Table 2 Summary of treatment-resistant depression studies

Furthermore, an improvement of 100% was reported in two DBS studies that included one patient with dystonia and TRD and another patient with MD and tardive dyskinesia.Citation16,Citation17 On a long-term basis (≤6 years), DBS is safe and effective in patients with TRD, as substantiated by recent data.Citation87Citation89 According to these studies,Citation87Citation89 chronic DBS SCG was effective in TRD and bipolar patients and well tolerated with minor hemorrhagic events,Citation86,Citation90,Citation91 but no neurocognitive impairment was reportedCitation61 (). As a mechanism of action, overactive SCG glucose metabolism seen in MD is reduced with antidepressant therapies and DBS.Citation10,Citation11

Table 3 Side effects of deep brain stimulation

Magnetic seizure therapy

Magnetic seizure therapy, also known as magnetic convulsion therapy and yet to be approved by the FDA, has antidepressant effects. It uses magnetic fields to induce therapeutic seizures. It has a better side-effect profile than modified ECT. Studies conducted in humans and primates suggest that cognitive side effects of MST are more benign than those of mECT. Notably, postictal orientation recovery time is short and rapid with MST.Citation61,Citation92,Citation93 Furthermore, several studies have corroborated improved cognitive outcomes with MST as compared to mECT. However, neither therapy causes structural changes, ie, volume, total number, or numerical density in neurons or glia in the frontal cortex, hippocampus, and their subregions in human and nonhuman brain.Citation94Citation96 Overall, magnetic seizures with benign side-effect profile are therapeutically better than mECT seizures. Other than adverse neurocognitive effects, ECT is also associated with reversible bradycardia and tachycardia immediate post-ECT and ictal and postictal stages, respectively. In nonhuman studies of MST, these effects were minimal, reflecting a more superficial cortical site of action with less impact on deep brain structures, which are implicated in sympathetic and parasympathetic nervous system control, relative to ECT.Citation97 Both antidepressant activity and cognitive side-effect profile of MST were further addressed in an open-label study, which tested whether it is associated with clinically significant antidepressant effects in TRD as an add-on therapy to controlled pharmacotherapy.Citation85 Twenty patients with TRD were randomly assigned to receive either MST or ECT for more than 2 years. The primary outcome measure was antidepressant response assessed by MADRS, and secondary outcome measures included HDRS, Hamilton Anxiety Scale, BDI, and 90-Item Symptom Checklist. Antidepressant response as defined by 50% improvement in MADRS ratings was statistically significant and of similar size in both treatment groups with no cognitive side effects. Characteristics in MST- and ECT-induced seizures were comparable, especially regarding ictal activity and postictal suppression. Kayser and colleagues suggested that MST may be a potential alternative to ECT if efficacy and safety are validated in larger clinical trials.Citation85 MST is reported to result in minimal retrograde and anterograde amnesia.Citation61 In summary, more studies are needed to further substantiate the efficacy of MST in mood disorder, including TRD patients.

Notably, there is converging evidence that NTs have a lower risk of neurocognitive side effects compared to mECT, which are benign.Citation61 (). By and large, short- and long-term research is needed to establish the efficacy, safety, and cost-effectiveness of neurostimulation therapies.Citation98 In addition, these therapies in general need proper selection of patients in line with tailored treatment guidelines.Citation99 Also, treatment teams should strictly follow ethical guidelines, especially those concerning autonomy, voluntary consent, beneficence, and nonmaleficence prior to using NTs in individual patients.Citation48,Citation80,Citation100

Table 4 Neurocognitive effects of somatic therapiesTable Footnote*

There are other NTs, including CES and ECS, used uncommonly for a variety of disorders, such as anxiety, headaches, pain, stroke recovery, movement disorders, insomnia, and depression, but the data are largely limited in TRD patients.Citation101,Citation102 In a systematic review, Rosa and Lisanby have described the technical details of all NTs, including indications, safety, and effectiveness of ECS and CES.Citation8 At the neurophysiological level, CES is quite different from tDCS.Citation103 In one study, with ECS that used prefrontal cortical modulation, an average 55% improvement in depression scores was demonstrated.Citation104 CES is associated with headache and nausea followed by skin irritation.Citation105 Unlike DBS, epidural cortical stimulation has fewer side effects.Citation8

Newer neurostimulation therapies

There are other neuromodulation therapies on the horizon, which include FUs, LFMS, and NIR.Citation106Citation109 The data about these approaches are limited and need further research, especially concerning their role in mood disorders, including TRD populations. With regard to OS, microbial light-sensitive proteins called opsins are introduced into neurons and function as ion channels that open or close according to light exposure. Channelrhodopsin-2 is one that allows Na+ ions to enter the cell following exposure to ~470 nm blue light.Citation110 According to Rosa and Lisanby,Citation8 the advent of this technique has multiple implications: targeting specific fiber tracts that overlap in space; selectively activating or inactivating specific projection neurons to the same target; being a contactless form of stimulation relying on photoactivation; and its potential use in treating mood disorders. Like DBS, OS will also require surgical implantation of the light-emitting electrode; however, OS certainly has other advantages over DBS.Citation8 In one nonhuman study, antidepressant effects of OS of medial prefrontal cortex have already been reported in a chronic social defeat stress model in rodents.Citation111 More studies on newer NTs are needed in human subjects with MD and TRD.

Discussion

This is a qualitative review of literature on somatic therapies used in the management of MD and refractory depression. About 30% of patients with TRD not responding to several intervention approaches, including optimization, augmentation and a combination of antidepressant drugs, are the principle candidates for NTs.Citation5Citation9 Among these therapies, mECT is most extensively and effectively used in severe depression and TRD but associated with serious neurocognitive adverse effects because of nonspecific, broad excitation of cortical and deeper structures of the brain, and its mechanism of action is continuingly debatable.Citation26Citation33 Other noninvasive somatic treatments such as rTMS, tDCS, MST, and CES target more specific neuronal networks in the brain that are dysfunctional in MD, TRD, and other neuropsychiatric disorders, and reported to have fairly good safety and clinical profiles with more benign neuropsychological side effects.Citation8,Citation9,Citation23Citation25,Citation48Citation61,Citation72Citation74,Citation85 Invasive NTs, ie, VNS, DBS, and ECS with nonserious adverse effect profile, are also reported to be effective in patients with MD and TRD.Citation8Citation10,Citation12Citation22,Citation61,Citation104 New NTs on the horizon are also promising in patients with MD and TRD. Although short- and long-term evidence-based comparative-effectiveness data on the role of NTs in adult patients TRD is emerging at a rapid pace,Citation112 further research on their technical optimization, mechanisms of action, efficacy, side effect profile, and cost-effectiveness in larger populations of TRD patients are warranted in future.

Conclusion

There is converging evidence that up to 40% patients with MD fail to respond to an initial antidepressant therapy. Modified ECT has a definite place in the management of patients with TRD; however, it carries well-known potential for neurocognitive impairment. Like ECT, MST also has neuropsychological adverse effects but of a milder nature. The role of other neuromodulation methods, including VNS, rTMS, DBS, and tDCS, in TRD patients is expanding with greater efficacy and fewer side effects. These neuromodulatory approaches rather tend to improve neurocognitive functions. These treatment modalities could be used alone or in combination with antidepressant therapy and/or psychotherapy. Besides their therapeutic utility, neuromodulation techniques can further open windows into the biological basis of disordered neurocircuits related to MD and TRD.

Recommendations

  1. Most studies on somatic therapies are of small sample size and hence reflect less reliable and valid results. Therefore, collaborative, multisite and/or multicountry studies that use the same protocols and also recruit larger samples with TRD are urgently needed.

  2. Another observation is that multiple hypotheses were tested in most neuromodulatory intervention trials. This methodological dilemma could be circumvented by determining a hypothesis a priori and others as exploratory.

  3. Most importantly, TRD evades a universally accepted definition, and hence tools to measure refractoriness of depression and strict eligibility criteria need to be developed.

  4. Evidently, poor results of recent MD and TRD trials indicate the heterogeneous nature of depression and TRD as well. Therefore, treatment trials of somatic therapies should target more specific subpopulations together with the detection of endophenotypes to predict their response

  5. Another challenge is blinding, which is vulnerable, and both the use of external raters and avoiding contact between subjects will solve this problem.

  6. Additionally, open-label studies, especially of VNS and DBS, tend to produce weak results, and therefore alternative designs including partial crossover and comparison against waiting list are needed.

  7. There is a relative lack of follow-up studies on somatic therapies, and hence more naturalistic studies are required in future.

  8. It is observed that the optimal parameters of somatic therapies are not defined, which could be managed by the use of adaptive designs and collaborative networks.

  9. Finally, unlike nonpharmacologic research in adults with TRD,Citation112 there is a relative lack of direct comparison with antidepressant drugs, and hence comparative research is needed. Most of these recommendations were constructed closely matching the challenges reported in the literature on NTs, MD, and TRD populations.Citation8,Citation72,Citation113

Disclosure

The authors disclose no conflicts of interest in this manuscript.

References

  • PrestonJDIntroduction to Psychopharmacology: A Practical Clinician’s Guide2010 Available from: http://www.continuingedcourses.net/active/courses/course015.phpAccessed May 24, 2012
  • World Health OrganizationMental health: depression Available from: http://www.who.int/mental_health/management/depression/definition/enAccessed February 12, 2012
  • SoueryDPapakostasGITrivediMHTreatment-resistant depressionJ Clin Psychiatry200667Suppl 6162216848672
  • ThaseMERushJATreatment-resistant depressionBloomFEKupferDJPsychopharmacologyNew YorkRaven199510811097
  • RuhéHGvan RooijenGSpijkerJPeetersFPScheneAHStaging methods for treatment resistant depression. A systematic reviewJ Affect Disord20121371–3354521435727
  • BerlimMTFleckMPTureckiGCurrent trends in the assessment and somatic treatment of resistant/refractory major depression: an overviewAnn Med200840214915918293145
  • GeorgeMSBrain stimulation treatments for depression: shifting the paradigm in treatment-resistant depression: assessing barriers to responses and applying adjunctive therapies for better patient results www.vindicomeded.com/cmelc/psych_monograph1209.aspAccessed on 12 May 2012
  • RosaMALisanbySHSomatic treatments for mood disordersNeuropsychopharmacology20123710211621976043
  • WardMPIrazoquiPPEvolving refractory major depressive disorder diagnostic and treatment paradigms: toward closed-loop therapeuticsFront Neuroeng20103720631824
  • LozanoAMMaybergHSGiacobbePHamaniCCraddockRCKennedySHSubcallosal cingulate gyrus deep brain stimulation for treatment-resistant depressionBiol Psychiatry200864646146718639234
  • MaybergHSBrannanSKTekellJLRegional metabolic effects of fluoxetine in major depression: serial changes and relationship to clinical responseBiol Psychiatry200048883084311063978
  • MaybergHSLozanoAMVoonVDeep brain stimulation for treatment-resistant depressionNeuron200545565166015748841
  • GreenbergBDMaloneDAFriehsGMThree-year outcomes in deep brain stimulation for highly resistant obsessive-compulsive disorderNeuropsychopharmacology200631112384239316855529
  • MaloneDAJrDoughertyDDRezaiARDeep brain stimulation of the ventral capsule/ventral striatum for treatment-resistant depressionBiol Psychiatry200965426727518842257
  • HälbigTDGruberDKoppUASchneiderGHTrottenbergTKupschAPallidal stimulation in dystonia: effects on cognition, mood, and quality of lifeJ Neurol Neurosurg Psychiatry200576121713171616291900
  • KoselMSturmVFrickCMood improvement after deep brain stimulation of the internal globus pallidus for tardive dyskinesia in a patient suffering from major depressionJ Psychiatr Res200741980180316962613
  • JimenezFVelascoFSalin-PascualRA patient with a resistant major depression disorder treated with deep brain stimulation in the inferior thalamic peduncleNeurosurgery20055758559316145540
  • VelascoFVelascoMJiménezFVelascoALSalin-PascualRNeurobiological background for performing surgical intervention in the inferior thalamic peduncle for treatment of major depression disordersNeurosurgery20055743944816145522
  • BewernickBHHurlemannRMatuschANucleus accumbens deep brain stimulation decreases ratings of depression and anxiety in treatment–resistant depressionBiol Psychiatry20096711011619914605
  • RushAJGeorgeMSSackeimHAVagus nerve stimulation (VNS) for treatment-resistant depressions: a multicenter studyBiol Psychiatry20004727628610686262
  • NemeroffCBMaybergHSKrahlSEVNS therapy in treatment-resistant depression: clinical evidence and putative neurobiological mechanismsNeuropsychopharmacology20063171345135516641939
  • GeorgeMSSackeimHAMarangellLBVagus nerve stimulation. A potential therapy for resistant depression?Psychiatr Clin North Am200023475778311147246
  • FitzgeraldPBBenitezJde CastellaADaskalakisZJBrownTLKulkarniJA randomized, controlled trial of sequential bilateral repetitive transcranial magnetic stimulation for treatment-resistant depressionAm J Psychiatry20061631889416390894
  • SpeerAMKimbrellTAWassermannEMOpposite effects of high and low frequency rTMS on regional brain activity in depressed patientsBiol Psychiatry2000481133114111137053
  • AveryDHHoltzheimerPE3rdFawazWA controlled study of repetitive transcranial magnetic stimulation in medication-resistant major depressionBiol Psychiatry200659218719416139808
  • FraisATElectroconvulsive therapy: a theory for the mechanism of actionJ ECT2010261606119458535
  • KatoNNeurophysiological mechanisms of electroconvulsive therapy for depressionNeurosci Res200964131119321135
  • Sánchez GonzálezRAlcoverroOPagerolsJRojoJEElectrophysiological mechanisms of action of electroconvulsive therapyActas Esp Psiquiatr200937634335120066586
  • BärKJEbertABoettgerMKIs successful electroconvulsive therapy related to stimulation of the vagal system?J Affect Disord20101251–332332920202688
  • NordanskogPDahlstrandULarssonMRLarssonEMKnutssonLJohansonAIncrease in hippocampal volume after electroconvulsive therapy in patients with depression: a volumetric magnetic resonance imaging studyJ ECT2010261626720190603
  • FinkMConvulsive therapy: a review of the first 55 yearsJ Affect Disord20016311511246075
  • KhalidNAtkinsMTredgetJGilesMChampney-SmithKKirovGThe effectiveness of electroconvulsive therapy in treatment-resistant depression: a naturalistic studyJ ECT200824214114518580559
  • SheltonRCOsuntokunOHeinlothANCoryaSATherapeutic options for treatment-resistant depressionCNS Drugs201024213116120088620
  • National Guideline ClearinghouseClinical Practice Guideline on Major Depression in Childhood and Adolescence2009 Available from: http://www.guideline.gov/content.aspx?id=25659Accessed March 28, 2012
  • RasmussenKGMuellerMRummansTAIs baseline medication resistance associated with potential for relapse after successful remission of a depressive episode with ECT? Data from the Consortium for Research on Electroconvulsive Therapy (CORE)J Clin Psychiatry200970223223719192459
  • PerugiGMeddaPZanelloSToniCCassanoGBEpisode length and mixed features as predictors of ECT nonresponse in patients with medication-resistant major depressionBrain Stimul201251182422037132
  • PiccinniADel DebbioAMeddaPPlasma brain-derived neurotrophic factor in treatment-resistant depressed patients receiving electroconvulsive therapyEur Neuropsychopharmacol200919534935519223156
  • OkamotoTYoshimuraRIkenouchi-SugitaAEfficacy of electroconvulsive therapy is associated with changing blood levels of homovanillic acid and brain-derived neurotrophic factor (BDNF) in refractory depressed patients: a pilot studyProg Neuropsychopharmacol Biol Psychiatry20083251185119018403081
  • GhaziuddinNDumasSHodgesEUse of continuation or maintenance electroconvulsive therapy in adolescents with severe treatment-resistant depressionJ ECT201127216817421233763
  • OkamotoNNakaiTSakamotoKNagafusaYHiguchiTNishikawaTRapid antidepressant effect of ketamine anesthesia during electroconvulsive therapy of treatment-resistant depression: comparing ketamine and propofol anesthesiaJ ECT201026322322719935085
  • BaghaiTCMarcuseABroschMThe influence of concomitant antidepressant medication on safety, tolerability and clinical effectiveness of electroconvulsive therapyWorld J Biol Psychiatry200672829016684680
  • FujitaANakaakiSSegawaKMemory, attention, and executive functions before and after sine and pulse wave electroconvulsive therapies for treatment-resistant major depressionJ ECT20062210711216801825
  • CrowleyKPickleJDaleRFattalOA critical examination of bifrontal electroconvulsive therapy: clinical efficacy, cognitive side effects, and directions for future researchJ ECT20082426827118648318
  • DumitriuDCollinsKAltermanRMathewSJNeurostimulatory therapeutics in management of treatment-resistant depression with focus on deep brain stimulationMt Sinai J Med20087526327518704979
  • GeorgeMSNahasZBorckardtJJBrain stimulation for the treatment of psychiatric disordersCurr Opin Psychiatry200720325025417415078
  • DoughertyDDRauchSLSomatic therapies for treatment-resistant depression: new neurotherapeutic interventionsPsychiatr Clin North Am2007301313717362801
  • JanicakPGO’ReardonJPSampsonSMTranscranial magnetic stimulation in the treatment of major depressive disorder: a comprehensive summary of safety experience from acute exposure, extended exposure, and during reintroduction treatmentJ Clin Psychiatry20086922223218232722
  • CroarkinPEWallCAMcClintockSMKozelFAHusainMMSampsonSMThe emerging role for repetitive transcranial magnetic stimulation in optimizing the treatment of adolescent depressionJ ECT201026432332920418774
  • FitzgeraldPBHoyKEHerringSEA double blind randomized trial of unilateral left and bilateral prefrontal cortex transcranial magnetic stimulation in treatment resistant major depressionJ Affect Disord Epub March 5, 2012
  • BlumbergerDMMulsantBHFitzgeraldPBA randomized double-blind sham-controlled comparison of unilateral and bilateral repetitive transcranial magnetic stimulation for treatment-resistant major depressionWorld J Biol Psychiatry Epub July 8, 2011
  • JhanwarVGBishnoiRJJhanwarMRUtility of repetitive transcranial stimulation as an augmenting treatment method in treatment-resistant depressionIndian J Psychol Med201133929622021964
  • Dell’ossoBCamuriGCastellanoFMeta-review of metanalytic studies with repetitive transcranial magnetic stimulation (rTMS) for the treatment of major depressionClin Pract Epidemiol Ment Health2011716717722135698
  • RossiSHallettMRossiniPMPascual-LeoneASafety of TMS Consensus GroupSafety, ethical considerations, and application guidelines for the use of transcranial magnetic stimulation in clinical practice and researchClin Neurophysiol2009120122008203919833552
  • LefaucheurJPAndré-ObadiaNPouletEFrench guidelines on the use of repetitive transcranial magnetic stimulation (rTMS): safety and therapeutic indicationsNeurophysiol Clin2011415–622129522153574
  • MartinJLBarbanojMJSchlaepferTERepetitive transcranial magnetic stimulation for the treatment of depression. Systematic review and meta-analysisBr J Psychiatry200318248049112777338
  • CouturierJLEfficacy of rapid-rate repetitive transcranial magnetic stimulation in the treatment of depression: a systematic review and meta-analysisJ Psychiatry Neurosci200530839015798783
  • SchutterDJAntidepressant efficacy of high-frequency transcranial magnetic stimulation over the left dorsolateral prefrontal cortex in double-blind sham-controlled designs: a meta-analysisPsychol Med200939657518447962
  • GrossMNakamuraLPascual-LeoneAHas repetitive transcranial magnetic stimulation (rTMS) treatment for depression improved? A systematic review and meta-analysis comparing the recent vs the earlier rTMS studiesActa Psychiatr Scand200711616517317655557
  • LamRWChanPWilkins-HoMRepetitive transcranial magnetic stimulation for treatment-resistant depression: a systematic review and metaanalysisCan J Psychiatry20085362163118801225
  • HerrmannLLEbmeierKPFactors modifying the efficacy of transcranial magnetic stimulation in the treatment of depression: a reviewJ Clin Psychiatry2006671870187617194264
  • MoreinesJLMcClintockSMHoltzheimerPENeuropsychological effects of neuromodulation techniques for treatment-resistant depression: a reviewBrain Stimul201141172721255751
  • NahasZBurnsCFoustMJShortBHerbsmanTGeorgeMSVagus nerve stimulation (VNS) for depression: what do we know now and what should be done next?Curr Psychiatry Rep20068644545117094924
  • O’ReardonJPCristanchoPPeshekADVagus nerve stimulation (VNS) and treatment of depression: to the brainstem and beyondPsychiatry200635546321103178
  • GoodmanWKInselTRDeep brain stimulation in psychiatry: concentrating on the road aheadBiol Psychiatry200965426326619167964
  • CarpenterLLMorenoFAKlingMAEffect of vagus nerve stimulation on cerebrospinal fluid monoamine metabolites, norepinephrine, and gamma-aminobutyric acid concentrations in depressed patientsBiol Psychiatry20045641842615364040
  • MarangellLBRushAJGeorgeMSVagus nerve stimulation (VNS) for major depressive episodes: one year outcomesBiol Psychiatry200251428028711958778
  • BajboujMMerklASchlaepferTETwo-year outcome of vagus nerve stimulation in treatment-resistant depressionJ Clin Psychopharmacol201030327328120473062
  • CristanchoPCristanchoMABaltuchGHThaseMEO’ReardonJPEffectiveness and safety of vagus nerve stimulation for severe treatment-resistant major depression in clinical practice after FDA approval: outcomes at 1 yearJ Clin Psychiatry201172101376138221295002
  • WarnellRLElahiNIntroduction of vagus nerve stimulation into a maintenance electroconvulsive therapy regimen: a case study and cost analysisJ ECT200723211411917548984
  • MohrPRodriguezMSlavíčkováAHankaJThe application of vagus nerve stimulation and deep brain stimulation in depressionNeuropsychobiology201164317018021811087
  • SackeimHARushAJGeorgeMSVagus nerve stimulation (VNS) for treatment-resistant depression: efficacy, side effects, and predictors of outcomeNeuropsychopharmacology20012571372811682255
  • BoggioPSRigonattiSPRibeiroRBA randomized, double-blind clinical trial on the efficacy of cortical direct current stimulation for the treatment of major depressionInt J Neuropsychopharmacol200811224925417559710
  • LooCKSachdevPMartinDA double-blind, sham-controlled trial of transcranial direct current stimulation for the treatment of depressionInt J Neuropsychopharmacol2010131616919671217
  • PalmUSchillerCFintescuZTranscranial direct current stimulation in treatment resistant depression: a randomized double-blind, placebo-controlled studyBrain Stimul Epub September 7, 2011
  • TierneyTSSankarTLozanoAMDeep brain stimulation emerging indicationsProg Brain Res2011194839521867796
  • HardestyDESackeimHADeep brain stimulation in movement and psychiatric disordersBiol Psychiatry20076183183517126303
  • KopellBHGreenbergBDAnatomy and physiology of the basal ganglia: implications for DBS in psychiatryNeurosci Biobehav Rev200832340842217854894
  • MaybergHSTargeted electrode-based modulation of neural circuits for depressionJ Clin Invest2009119471772519339763
  • TyeSJFryeMALeeKHDisrupting disordered neurocircuitry: treating refractory psychiatric illness with neuromodulationMayo Clin Proc200984652253219483169
  • SchlaepferTEBewernickBKayserSLenzDModulating affect, cognition, and behavior – prospects of deep brain stimulation for treatment-resistant psychiatric disordersFront Integr Neurosci201152921738500
  • LujanJLChaturvediAMaloneDARezaiARMachadoAGMcIntyreCCAxonal pathways linked to therapeutic and nontherapeutic outcomes during psychiatric deep brain stimulationHum Brain Mapp201233495896821520343
  • BewernickBHHurlemannRMatuschANucleus accumbens deep brain stimulation decreases ratings of depression and anxiety in treatment-resistant depressionBiol Psychiatry201067211011619914605
  • LozanoAMGiacobbePHamaniCA multicenter pilot study of subcallosal cingulate area deep brain stimulation for treatment-resistant depressionJ Neurosurg2012116231532222098195
  • KleinEKreininIChistyakovATherapeutic efficacy of right prefrontal slow repetitive transcranial magnetic stimulation in major depression: a double-blind controlled studyArch Gen Psychiatry19995631532010197825
  • KayserSBewernickBHGrubertCHadrysiewiczBLAxmacherNSchlaepferTEAntidepressant effects, of magnetic seizure therapy and electroconvulsive therapy, in treatment-resistant depressionJ Psychiatr Res201145556957620951997
  • BlomstedtPSjobergRLHanssonMBodlundOHarizMIDeep brain stimulation in the treatment of depressionActa Psychiatr Scand201112341120961293
  • HoltzheimerPEKelleyMEGrossRESubcallosal cingulate deep brain stimulation for treatment-resistant unipolar and bipolar depressionArch Gen Psychiatry201269215015822213770
  • KennedySHGiacobbePRizviSJDeep brain stimulation for treatment-resistant depression: follow-up after 3 to 6 yearsAm J Psychiatry2011168550251021285143
  • RizviSJDonovanMGiacobbePPlacenzaFRotzingerSKennedySHNeurostimulation therapies for treatment resistant depression: a focus on vagus nerve stimulation and deep brain stimulationInt Rev Psychiatry201123542443622200132
  • BerneyAVingerhoetsFPerrinAEffect on mood of subthalamic DBS for Parkinson’s disease: a consecutive series of 24 patientsNeurology2002591427142912427897
  • BinderDKRauGStarrPAHemorrhagic complications of microelectrode-guided deep brain stimulationStereotact Funct Neurosurg200380283114745205
  • KirovGEbmeierKPScottAIFQuick recovery of orientation after 100 Hz magnetic seizure therapy (MST) for major depressive disorderBr J Psychiatry2008193215215518670002
  • ZyssTZiebaAHeseRTMagnetic seizure therapy (MST) – a safer method for evoking seizure activity than current therapy with a confirmed antidepressant efficacyNeuro Endocrinol Lett201031442543720802450
  • MoscripTDTerraceHSSackeimHALisanbySHRandomized controlled trial of the cognitive side-effects of magnetic seizure therapy (MST) and electroconvulsive shock (ECS)Int J Neuropsychopharmacol20069111116045810
  • CycowiczYMLuberBSpellmanTLisanbySHNeurophysiological characterization of high-dose magnetic seizure therapy: comparisons with electroconvulsive shock and cognitive outcomesJ ECT200925315716419300292
  • DworkAJChristensenJRLarsenKBUnaltered neuronal and glial counts in animal models of magnetic seizure therapy and electroconvulsive therapyNeuroscience200916441557156419782728
  • RownySBCycowiczYMMcClintockSMTruesdaleMDLuberBLisanbySHDifferential heart rate response to magnetic seizure therapy (MST) relative to electroconvulsive therapy: a nonhuman primate modelNeuroimage20094731086109119497373
  • CarpenterLLNeurostimulation in resistant depressionJ Psychopharmacol200620Suppl 3354016644770
  • KennedySHMilevRGiacobbePCanadian Network for Mood and Anxiety Treatments (CANMAT) clinical guidelines for the management of major depressive disorder in adults. IV. Neurostimulation therapiesJ Affect Disord2009117Suppl 1S44S5319656575
  • WardHEHwynnNOkunMSUpdate on deep brain stimulation for neuropsychiatric disordersNeurobiol Dis201038334635320096357
  • BystritskyAKerwinLFeusnerJA pilot study of cranial electrotherapy stimulation for generalized anxiety disorderJ Clin Psychiatry20086941241718348596
  • KlawanskySYeungABerkeyCShahNPhanHChalmersTCMeta–analysis of randomized controlled trials of cranial electrostimulation. Efficacy in treating selected psychological and physiological conditionsJ Nerv Ment Dis19951834784847623022
  • StaggCJNitscheMAPhysiological basis of transcranial direct current stimulationNeuroscientist201117375321343407
  • NahasZAndersonBSBorckardtJBilateral epidural prefrontal cortical stimulation for treatment-resistant depressionBiol Psychiatry20106710110919819427
  • KirschDLSmithRBThe use of cranial electrotherapy stimulation in the management of chronic pain: a reviewNeuro Rehabilitation200014859411455071
  • TylerWJTufailYFinsterwaldMTauchmannMLOlsonEJMajesticCRemote excitation of neuronal circuits using low-intensity, low-frequency ultrasoundPLoS One20083e351118958151
  • VolkowNDTomasiDWangGJEffects of low-field magnetic stimulation on brain glucose metabolismNeuroimage20105162362820156571
  • KatzEJIlevIKKrauthamerVKim doHWeinreichDExcitation of primary afferent neurons by near-infrared light in vitroNeuroreport20102166266620495496
  • MathewMAmat-RoldanIAndresRSignalling effect of NIR pulsed lasers on axonal growthJ Neurosci Methods201018619620119945486
  • ZhangFAravanisAMAdamantidisAde LeceaLDeisserothKCircuit-breakers: optical technologies for probing neural signals and systemsNat Rev Neurosci2007857758117643087
  • CovingtonHE3rdLoboMKMazeIAntidepressant effect of optogenetic stimulation of the medial prefrontal cortexJ Neurosci201030160821609021123555
  • GaynesBNLuxLLloydSNonpharmacologic Interventions for Treatment-Resistant Depression in Adults2011 Available from: http://www.effectivehealthcare.ahrq.gov/reports/final.cfmAccessed April 3, 2012
  • BrunoniARTengCTCorreaCNeuromodulation approaches for the treatment of major depression: challenges and recommendations from a working group meetingArq Neuropsiquiatr201068343345120602051