Early interventions for psychosis

Article Type
Changed
Tue, 11/02/2021 - 12:36
Display Headline
Early interventions for psychosis

Neuroscience research over the past half century has failed to significantly advance the treatment of severe mental illness.1,2 Hence, evidence that a longer duration of untreated psychosis (DUP) aggravates—and early intervention with medication and social supports ameliorates—the long-term adverse consequences of psychotic disorders generated a great deal of interest.3,4 This knowledge led to the development of diverse early intervention services worldwide aimed at this putative “critical window.” It raised the possibility that appropriate interventions could prevent the long-term disability that makes chronic psychosis one of the most debilitating disorders.5,6 However, even beyond the varied cultural and economic confounds, it is difficult to assess, compare, and optimize program effectiveness.7 Obstacles include paucity of sufficiently powered, well-designed randomized controlled trials (RCTs), the absence of diagnostic biomarkers or other prognostic indicators to better account for the inherent heterogeneity in the population and associated outcomes, and the absence of modifiable risk factors that can guide interventions and provide intermediate outcomes.4,8-10

To better appreciate these issues, it is important to distinguish whether a program is designed to prevent psychosis, or to mitigate the effects of psychosis. Two models include the:

  • Prevention model, which focuses on young individuals who are not yet overtly psychotic but at high risk
  • First-episode recovery model, which focuses on those who have experienced a first episode of psychosis (FEP) but have not yet developed a chronic disorder.

Both models share long-term goals and are hampered by many of the same issues summarized above. They both deviate markedly from the standard medical model by including psychosocial services designed to promote restoration of a self-defined trajectory to greater independence.11-14 The 2 differ, however, in the challenges they must overcome to produce their sample populations and establish effective interventions.10,15,16

In this article, we provide a succinct overview of these issues and a set of recommendations based on a “strength-based” approach. This approach focuses on finding common ground between patients, their support system, and the treatment team in the service of empowering patients to resume responsibility for transition to adulthood.

The prevention model

While most prevention initiatives in medicine rely on the growing ability to target specific pathophysiologic pathways,3 preventing psychosis relies on clinical evidence showing that DUP and early interventions predict a better course of severe mental illness.17 In contrast, initiatives such as normalizing neonatal neuronal pathways are more consistent with the strategy utilized in other fields but have yet to yield a pathophysiologic target for psychosis.3,18

Initial efforts to identify ‘at-risk’ individuals

The prevention model of psychosis is based on the ability to identify young individuals at high risk for developing a psychotic disorder (Figure). The first screening measures were focused on prodromal psychosis (eg, significant loss of function, family history, and “intermittent” and “attenuated” psychotic symptoms). When applied to referred (ie, pre-screened) samples, 30% to 40% of this group who met criteria transitioned to psychosis over the next 1 to 3 years despite antidepressant and psychosocial interventions.19 Comprising 8 academic medical centers, the North American Prodrome Longitudinal Study (NAPLS) produced similar results using the Structured Interview for Prodromal Syndromes (SIPS).17 Thus, 30% to 50% of pre-screened individuals referred by school counselors and mental health professionals met SIPS criteria, and 35% of these individuals transitioned to psychosis over 30 months. The validity of this measure was further supported by the fact that higher baseline levels of unusual thought content, suspicion/paranoia, social impairment, and substance abuse successfully distinguished approximately 80% of those who transitioned to psychosis. The results of this first generation of screening studies were exciting because they seemed to demonstrate that highly concentrated samples of young persons at high risk of developing psychosis could be identified, and that fine-tuning the screening criteria could produce even more enriched samples (ie, positive predictive power).

CP02010012_f1.PNG

Initial interventions produced promising results

The development of effective screening measures led to reports of effective treatment interventions. These were largely applied in a clinical staging model that restricted antipsychotic medications to those who failed to improve after receiving potentially “less toxic” interventions (eg, omega-3 polyunsaturated fatty acids and other antioxidants; psychotherapy; cognitive-behavioral therapy [CBT]; family therapy).5 While study designs were typically quasi-experimental, the interventions appeared to dramatically diminish the transition to psychosis (ie, approximately 50%).

Continue to: The first generation...

 

 

The first generation of RCTs appeared to confirm these results, although sample sizes were small, and most study designs assessed only a single intervention. Initial meta-analyses of these data reported that both CBT and antipsychotics appeared to prevent approximately one-half of individuals from becoming psychotic at 12 months, and more than one-third at 2 to 4 years, compared with treatment as usual.20

While some researchers challenged the validity of these findings,21-23 the results generated tremendous international enthusiasm and calls for widespread implementation.6 The number of early intervention services (EIS) centers increased dramatically worldwide, and in 2014 the National Institute for Health and Care Excellence released standards for interventions to prevent transition to psychosis.24 These included close monitoring, CBT and family interventions, and avoiding antipsychotics when possible.24

Focusing on sensitivity over specificity

The first generation of studies generated by the prevention model relied on outreach programs or referrals, which produced small samples of carefully selected, pre-screened individuals (Figure, Pre-screened) who were then screened again to establish the high-risk sample.25 While approximately 33% of these individuals became psychotic, the screening process required a very efficient means of eliminating those not at high-risk (given the ultimate target population represented only approximately .5% of young people) (Figure). The pre-screening and screening processes in these first-generation studies were labor-intensive but could only identify approximately 5% of those individuals destined to become psychotic over the next 2 or 3 years. Thus, alternative methods to enhance sensitivity were needed to extend programming to the general population.

Second-generation pre-screening (Figure; Step 1). New pre-screening methods were identified that captured more individuals destined to become psychotic. For example, approximately 90% of this population were registered in health care organizations (eg, health maintenance organizations) and received a psychiatric diagnosis in the year prior to the onset of psychosis (true positives).8 These samples, however, contained a much higher percentage of persons not destined to become psychotic, and somehow the issue of specificity (decreasing false positives) was minimized.8,9 For example, pre-screened samples contained 20 to 50 individuals not destined to become psychotic for each one who did.26 Since screening measures could only eliminate approximately 20% of this group (Figure, Step 2, page 25), second-generation transition rates fell from 30% to 40% to 2% to 10%.27,28

Other pre-screening approaches were introduced, but they also focused on capturing more of those destined to become psychotic (sensitivity) than eliminating those who would not (specificity). For instance, Australia opened more than 100 “Headspace” community centers nationwide designed to promote engagement and self-esteem in youth experiencing anxiety; depression; stress; relationship, work, or school problems; or bullying.13 Most services were free and included mental health staff who screened for psychosis and provided a wide range of services in a destigmatized setting. These methods identified at least an additional 5% to 7% of individuals destined to become psychotic, but to our knowledge, no data have been published on whether they helped eliminate those who did not.

Continue to: Second-generation screening

 

 

Second-generation screening (Figure, Step 2). A second screening aims to retain those pre-screened individuals who will become psychotic (ie, minimizing false negatives) while further minimizing those who do not (ie, minimizing false positives). The addition of cognitive, neural (eg, structural MRI; neurophysiologic), and biochemical (eg, inflammatory immune and stress) markers to the risk calculators have produced a sensitivity close to 100%.8,9 Unfortunately, these studies downplayed specificity, which remained approximately 20%.8,9 Specificity is critical not just because of concerns about stigma (ie, labeling people as pre-psychotic when they are not) but also because of the adverse effects of antipsychotic medications and the effects on future program development (interventions are costly and labor-intensive). Also, diluting the pool with individuals not at risk makes it nearly impossible to identify effective interventions (ie, power).27,28

While some studies focused on increasing specificity (to approximately 75%), this leads to an unacceptable loss of sensitivity (from 90% to 60%),29 with 40% of pre-screened individuals who would become psychotic being eliminated from the study population. The addition of other biological markers (eg, salivary cortisol)30 and use of learning health systems may be able to enhance these numbers (initial reports of specificity = 87% and sensitivity = 85%).8,9 This is accomplished by integrating artificial and human intelligence measures of clinical (symptom and neurocognitive measures) and biological (eg, polygenetic risk scores; gray matter volume) variables.31 However, even if these results are replicated, more effective pre-screening measures will be required.

Identifying a suitable sample population for prevention program studies is clearly more complicated than for FEP studies, where one can usually identify many of those in the at-risk population by their first hospitalization for psychotic symptoms. The issues of false positives (eg, substance-induced psychosis) and negatives (eg, slow deterioration, prominent negative symptoms) are important concerns, but proportionately far less significant.

 

Prevention and FEP interventions

Once a study sample is constituted, 1 to 3 years of treatment interventions are initiated. Interventions for prevention programs typically include CBT directed at attenuated psychosis (eg, reframing or de-catastrophizing unusual thoughts and minimizing distress associated with unusual perceptions); case management to facilitate personal, educational, and vocational goals; and family therapy in single or multi-group formats to educate one’s support system about the risk state and to minimize adverse familial responses.14 Many programs also include supported education or employment services to promote reintegration in age-appropriate activities; group therapy focused on substance abuse and social skills training; cognitive remediation to ameliorate the cognitive dysfunction; and an array of pharmacologic interventions designed to delay or prevent transition to psychosis or to alleviate symptoms. While most interventions are similar, FEP programs have recently included peer support staff. This appears to instill hope in newly diagnosed patients, provide role models, and provide peer supporters an opportunity to use their experiences to help others and earn income.32

The breadth and depth of these services are critical because retention in the program is highly dependent on participant engagement, which in turn is highly dependent on whether the program can help individuals get what they want (eg, friends, employment, education, more autonomy, physical health). The setting and atmosphere of the treatment program and the willingness/ability of staff to meet participants in the community are also important elements.11,12 In this context, the Headspace community centers are having an impact far beyond Australia and may prove to be a particularly good model.13

Continue to: Assessing prevention and FEP interventions

 

 

Assessing prevention and FEP interventions

The second generation of studies of prevention programs has not confirmed, let alone extended, the earlier findings and meta-analyses. A 2020 report concluded CBT was still the most promising intervention; it was more effective than control treatments at 12 and 18 months, although not at 6, 24, or 48 months.33 This review included controlled, open-label, and naturalistic studies that assessed family therapy; omega-3 polyunsaturated fatty acids; integrated psychological therapy (a package of interventions that included family education, CBT, social skills training, and cognitive remediation); N-methyl-D-aspartate receptor modulators; mood stabilizers; and antipsychotics. In addition to the evidence supporting CBT, the results also indicated nonsignificant trends favoring family and integrated psychological therapy. Neither a 2019 Cochrane review34 nor a 2020 “umbrella” assessment of 42 meta-analyses9 found convincing evidence for the efficacy of any program components.

While these disappointing findings are at least partly attributable to the methodological challenges described above and in the Figure, other factors may hinder establishing effective interventions. In contrast to FEP studies, those focused on prevention had a very ambitious agenda (eliminating psychosis) and tended to downplay more modest intermediate outcomes. These studies also tended to assess new ideas with small samples rather than pursue promising findings with larger multi-site studies focused on a group of interventions. The authors of a Cochrane review observed “There is the impression that in this whole area there is a triumph of hope over adversity. There is the repeated hope invested in another—often unique—study question and then a study of fewer than 100 participants are completed. This results in the set of comparisons reported here, all 9 of which are too underpowered to really highlight clear differences.”34 To use a baseball analogy, it seems that investigators are “swinging for the fence” when a few singles are what’s really needed.

From the outset, the goals of FEP studies were more modest, largely ignoring the task of developing consensus definitions of recovery that require following patients for up to 5 to 10 years. Instead, they use intermediate endpoints based on adapting treatments that already appeared effective in patients with chronic mental disorders.35 As a consequence, researchers examining FEP demonstrated clear, albeit limited, salutary effects using large multi-site trials and previously established outcome measures.3,10,36 For instance, the Recovery After an Initial Schizophrenia Episode-Early Treatment Program (RAISE-ETP) study was a 2-year, multi-site RCT (N = 404) funded by the National Institute of Mental Health (NIMH). The investigators reported improved indices of social function (eg, quality of life; education and work participation) and total ratings of psychopathology and depression compared with treatment as usual. Furthermore, they established that DUP predicted treatment response.35 The latter finding was underscored by improvement being limited to the 50% with <74 weeks DUP. Annual costs of the program per 1 standard deviation improvement in quality of life were approximately $1,000 for patients with <74 weeks DUP and $40,000 for those with >74 weeks DUP. Concurrent meta-analyses confirmed and extended these findings,16 showing higher remission rates; diminished relapses and hospital admissions; greater engagement in programming; greater involvement in work and school; improved quality of life; and other steps toward recovery. These studies were also able to establish a clear benefit of antipsychotic medications, particularly a high acceptance of long-acting injectable antipsychotic formulations, which promoted adherence and decreased some adverse events37; and early use of clozapine therapy, which improved remission rates and longer-term outcomes.38 Other findings underscored the need to anticipate and address new problems associated with effective antipsychotic therapy (eg, antipsychotic response correlates with weight gain, a particularly intolerable adverse event for this age group).39 Providing pre-emptive strategies such as exercise groups and nutritional education may be necessary to maintain adherence.

 

Limitations of FEP studies

The effect sizes in these FEP studies were small to medium on outcome measures tracking recovery and associated indicators (eg, global functioning, school/work participation, treatment engagement); the number needed to treat for each of these was >10. There is no clear evidence that recovery programs such as RAISE-ETP actually reduce longer-term disability. Most studies showed disability payments increased while clinical benefits tended to fade over time. In addition, by grouping interventions together, the studies made it difficult to identify effective vs ineffective treatments, let alone determine how best to personalize therapy for participants in future studies.

The next generation of FEP studies

While limited in scope, the results of the recent FEP studies justify a next generation of recovery interventions designed to address these shortcomings and optimize program outcomes.39 Most previous FEP studies were conducted in community mental health center settings, thus eliminating the need to transition services developed in academia into the “real world.” The next generation of NIMH studies will be primarily conducted in analogous settings under the Early Psychosis Intervention Network (EPINET).40 EPINET’s study design echoes that responsible for the stepwise successes in the late 20th century that produced cures for the deadliest childhood cancer, acute lymphoblastic leukemia (ALL). This disease was successfully treated by modifying diverse evidence-based practices without relying on pharmacologic or other major treatment breakthroughs. Despite this, the effort yielded successful personalized interventions that were not obtainable for other severe childhood conditions.40 EPINET hopes to automate much of these stepwise advances with a learning health system. This program relies on data routinely collected in clinical practice to drive the process of scientific discovery. Specifically, it determines the relationships between clinical features, biologic measures, treatment characteristics, and symptomatic and functional outcomes. EPINET aims to accelerate our understanding of biomarkers of psychosis risk and onset, as well as factors associated with recovery and cure. Dashboard displays of outcomes will allow for real-time comparisons within and across early intervention clinics. This in turn identifies performance gaps and drives continuous quality improvement.

Continue to: Barriers to optimizing program efficacy for both models

 

 

Barriers to optimizing program efficacy for both models

Unfortunately, there are stark differences between ALL and severe mental disorders that potentially jeopardize the achievement of these aims, despite the advances in data analytic abilities that drive the learning health system. Specifically, the heterogeneity of psychotic illnesses and the absence of reliable prognostic and modifiable risk markers (responsible for failed efforts to enhance treatment of serious mental illness over the last half century1,2,41) are unlikely to be resolved by a learning health system. These measures are vital to determine whether specific interventions are effective, particularly given the absence of a randomized control group in the EPINET/learning health system design. Fortunately, however, the National Institutes for Health has recently initiated the Accelerating Medicines Partnership–Schizophrenia (AMP-SCZ). This approach seeks “promising biological markers that can help identify those at risk of developing schizophrenia as early as possible, track the progression of symptoms and other outcomes and ultimately define targets for treatment development.”42 The Box1,4,9,10,36,41,43-45 describes some of the challenges involved in identifying biomarkers of severe mental illness.

Box

Identifying biomarkers of severe mental illness

Biomarkers and modifiable risk factors4,9,10,41,43 are at the core of personalized medicine and its ultimate objective (ie, theragnostics). This is the ability to identify the correct intervention for a disorder based on a biomarker of the illness.10,36 The inability to identify biomarkers of severe mental illness is multifactorial but in part may be attributable to “looking in all the wrong places.”41 By focusing on neural processes that generate psychiatric symptomatology, investigators are assuming they can bridge the “mind gap”1 and specifically distinguish between pathological, compensatory, or collateral measures of poorly characterized limbic neural functions.41

It may be more productive to identify a pathological process within the limbic system that produces a medical condition as well as the mental disorder. If one can isolate the pathologic limbic circuit activity responsible for a medical condition, one may be able to reproduce this in animal models and determine whether analogous processes contribute to the core features of the mental illness. Characterization of the aberrant neural circuit in animal models also could yield targets for future therapies. For example, episodic water intoxication in a discrete subset of patients with schizophrenia44 appears to arise from a stress diathesis produced by anterior hippocampal pathology that disrupts regulation of antidiuretic hormone, oxytocin, and hypothalamic-pituitary-adrenal axis secretion. These patients also exhibit psychogenic polydipsia that may be a consequence of the same hippocampal pathology that disrupts ventral striatal and lateral hypothalamic circuits. These circuits, in turn, also modulate motivated behaviors and cognitive processes likely relevant to psychosis.45

A strength-based approach

The absence of sufficiently powered RCTs for prevention studies and the reliance on intermediate outcomes for FEP studies leaves unanswered whether such programs can effectively prevent chronic psychosis at a cost society is willing to pay. Still, substantial evidence indicates that outreach, long-acting injectable antipsychotics, early consideration of clozapine, family therapy, CBT for psychosis/attenuated psychosis, and services focused on competitive employment can preserve social and occupational functioning.16,34 Until these broader questions are more definitively addressed, it seems reasonable to apply what we have learned (Table11,12,35,37-39,46).

CP02010012_t1.PNG

Simply avoiding the most divisive aspects of the medical model that inadvertently promote stigma and undercut self-confidence may help maintain patients’ willingness to learn how best to apply their strengths and manage their limitations.11 The progression to enduring psychotic features (eg, fixed delusions) may reflect ongoing social isolation and alienation. A strength-based approach seeks first to establish common goals (eg, school, work, friends, family support, housing, leaving home) and then works to empower the patient to successfully reach those goals.35 This typically involves giving them the opportunity to fail, avoiding criticism when they do, and focusing on these experiences as learning opportunities from which success can ultimately result.

It is difficult to offer all these services in a typical private practice setting. Instead, it may make more sense to use one of the hundreds of early intervention services programs in the United States.46 If a psychiatric clinician is dedicated to working with this population, it may also be possible to establish ongoing relationships with primary care physicians, family and CBT therapists, family support services (eg, National Alliance on Mental Illness), caseworkers and employment counselors. In essence, a psychiatrist may be able re-create a multidisciplinary effort by taking advantage of the expertise of these various professionals. The challenge is to create a consistent message for patients and families in the absence of regular meetings with the clinical team, although the recent reliance on and improved sophistication of virtual meetings may help. Psychiatrists often play a critical role even when the patient is not prescribed medication, partly because they are most comfortable handling the risks and may have the most comprehensive understanding of the issues at play. When medications are appropriate and patients with FEP are willing to take them, early consideration of long-acting injectable antipsychotics and clozapine may provide better stabilization and diminish the risk of earlier and more frequent relapses.

Bottom Line

Early interventions for psychosis include the prevention model and the first-episode recovery model. It is difficult to assess, compare, and optimize the effectiveness of such programs. Current evidence supports a ‘strength-based’ approach focused on finding common ground between patients, their support system, and the treatment team.

Related Resources

  • Early Assessment and Support Alliance. National Early Psychosis Directory. https://easacommunity.org/nationaldirectory.php
  • Kane JM, Robinson DG, Schooler NR, et al. Comprehensive versus usual community care for first-episode psychosis: 2-year outcomes from the NIMH RAISE Early Treatment Program. Am J Psychiatry. 2016 ;173(4):362-372

Drug Brand Name

Clozapine • Clozaril

References

1. Hyman SE. Revolution stalled. Sci Transl Med. 2012;4(155):155cm11. doi: 10.1126/scitranslmed.3003142

2. Harrington A. Mind fixers: psychiatry’s troubled search for the biology of mental illness. W.W. Norton & Company; 2019.

3. Millan MJ, Andrieux A, Bartzokis G, et al. Altering the course of schizophrenia: progress and perspectives. Nat Rev Drug Discov. 2016;15(7):485-515.

4. Lieberman JA, Small SA, Girgis RR. Early detection and preventive intervention in schizophrenia: from fantasy to reality. Am J Psychiatry. 2019;176(10):794-810.

5. McGorry PD, Nelson B, Nordentoft M, et al. Intervention in individuals at ultra-high risk for psychosis: a review and future directions. J Clin Psychiatry. 2009;70(9):1206-1212.

6. Csillag C, Nordentoft M, Mizuno M, et al. Early intervention in psychosis: From clinical intervention to health system implementation. Early Interv Psychiatry. 2018;12(4):757-764.

7. McGorry PD, Ratheesh A, O’Donoghue B. Early intervention—an implementation challenge for 21st century mental health care. JAMA Psychiatry. 2018;75(6):545-546.

8. Rosenheck R. Toward dissemination of secondary prevention for psychosis. Am J Psychiatry. 2018;175(5):393-394.

9. Fusar-Poli P, Salazar de Pablo G, Correll CU, et al. Prevention of psychosis: advances in detection, prognosis, and intervention. JAMA Psychiatry. 2020;77(7):755-765.

10. Oliver D, Reilly TJ, Baccaredda Boy O, et al. What causes the onset of psychosis in individuals at clinical high risk? A meta-analysis of risk and protective factors. Schizophr Bull. 2020;46(1):110-120.

11. Tindall R, Simmons M, Allott K, et al. Disengagement processes within an early intervention service for first-episode psychosis: a longitudinal, qualitative, multi-perspective study. Front Psychiatry. 2020;11:565-565.

12. Dixon LB, Holoshitz Y, Nossel I. Treatment engagement of individuals experiencing mental illness: review and update. World Psychiatry. 2016;15(1):13-20.

13. Rickwood D, Paraskakis M, Quin D, et al. Australia’s innovation in youth mental health care: The headspace centre model. Early Interv Psychiatry. 2019;13(1):159-166.

14. Woodberry KA, Shapiro DI, Bryant C, et al. Progress and future directions in research on the psychosis prodrome: a review for clinicians. Harv Rev Psychiatry. 2016;24(2):87-103.

15. Gupta T, Mittal VA. Advances in clinical staging, early intervention, and the prevention of psychosis. F1000Res. 2019;8:F1000 Faculty Rev-2027. doi: 10.12688/f1000research.20346.1

16. Correll CU, Galling B, Pawar A, et al. Comparison of early intervention services vs treatment as usual for early-phase psychosis: a systematic review, meta-analysis, and meta-regression. JAMA Psychiatry. 2018;75(6):555-565.

17. Cannon TD, Cadenhead K, Cornblatt B, et al. Prediction of psychosis in youth at high clinical risk: a multisite longitudinal study in North America. Arch Gen Psychiatry. 2008;65(1):28-37.

18. Sommer IE, Bearden CE, van Dellen E, et al. Early interventions in risk groups for schizophrenia: what are we waiting for? NPJ Schizophr. 2016;2(1):16003-16003.

19. McGorry PD, Nelson B. Clinical high risk for psychosis—not seeing the trees for the wood. JAMA Psychiatry. 2020;77(7):559-560.

20. van der Gaag M, Smit F, Bechdolf A, et al. Preventing a first episode of psychosis: meta-analysis of randomized controlled prevention trials of 12 month and longer-term follow-ups. Schizophr Res. 2013;149(1):56-62.

21. Marshall M, Rathbone J. Early intervention for psychosis. Cochrane Database Syst Rev. 2011;(6):CD004718. doi: 10.1002/14651858.CD004718.pub3

22. Heinssen RK, Insel TR. Preventing the onset of psychosis: not quite there yet. Schizophr Bull. 2015;41(1):28-29.

23. Amos AJ. Evidence that treatment prevents transition to psychosis in ultra-high-risk patients remains questionable. Schizophr Res. 2014;153(1):240.

24. National Institute for Health and Care Excellence. Psychosis and schizophrenia in adults: prevention and management. Clinical guideline [CG178]. 1.3.7 How to deliver psychological interventions. Published February 12, 2014. Updated March 1, 2014. Accessed August 30, 2021. https://www.nice.org.uk/guidance/cg178/chapter/recommendations#how-to-deliver-psychological-interventions

25. Fusar-Poli P, Werbeloff N, Rutigliano G, et al. Transdiagnostic risk calculator for the automatic detection of individuals at risk and the prediction of psychosis: second replication in an independent National Health Service Trust. Schizophr Bull. 2019;45(3):562-570.

26. Fusar-Poli P, Oliver D, Spada G, et al. The case for improved transdiagnostic detection of first-episode psychosis: electronic health record cohort study. Schizophr Res. 2021;228:547-554.

27. Fusar-Poli P. Negative psychosis prevention trials. JAMA Psychiatry. 2017;74(6):651.

28. Cuijpers P, Smit F, Furukawa TA. Most at‐risk individuals will not develop a mental disorder: the limited predictive strength of risk factors. World Psychiatry. 2021;20(2):224-225.

29. Carrión RE, Cornblatt BA, Burton CZ, et al. Personalized prediction of psychosis: external validation of the NAPLS-2 psychosis risk calculator with the EDIPPP Project. Am J Psychiatry. 2016;173(10):989-996.

30. Worthington MA, Walker EF, Addington J, et al. Incorporating cortisol into the NAPLS2 individualized risk calculator for prediction of psychosis. Schizophr Res. 2021;227:95-100.

31. Koutsouleris N, Dwyer DB, Degenhardt F, et al. Multimodal machine learning workflows for prediction of psychosis in patients with clinical high-risk syndromes and recent-onset depression. JAMA Psychiatry. 2021;78(2):195-209.

32. Simmons MB, Grace D, Fava NJ, et al. The experiences of youth mental health peer workers over time: a qualitative study with longitudinal analysis. Community Ment Health J. 2020;56(5):906-914.

33. Devoe DJ, Farris MS, Townes P, et al. Interventions and transition in youth at risk of psychosis: a systematic review and meta-analyses. J Clin Psychiatry. 2020;81(3):17r12053. doi: 10.4088/JCP.17r12053

34. Bosnjak Kuharic D, Kekin I, Hew J, et al. Interventions for prodromal stage of psychosis. Cochrane Database Syst Rev. 2019;2019(11):CD012236

35. Dixon LB, Goldman HH, Srihari VH, et al. Transforming the treatment of schizophrenia in the United States: The RAISE Initiative. Annu Rev Clin Psychol. 2018;14:237-258.

36. Friedman-Yakoobian MS, Parrish EM, Eack SM, et al. Neurocognitive and social cognitive training for youth at clinical high risk (CHR) for psychosis: a randomized controlled feasibility trial. Schizophr Res. 2020;S0920-9964(20)30461-8. doi: 10.1016/j.schres.2020.09.005

37. Kane JM, Schooler NR, Marcy P, et al. Effect of long-acting injectable antipsychotics vs usual care on time to first hospitalization in early-phase schizophrenia: a randomized clinical trial. JAMA Psychiatry. 2020;77(12):1217-1224.

38. Morrison AP, Pyle M, Maughan D, et al. Antipsychotic medication versus psychological intervention versus a combination of both in adolescents with first-episode psychosis (MAPS): a multicentre, three-arm, randomised controlled pilot and feasibility study. Lancet Psychiatry. 2020;7(9):788-800.

39. Chen YQ, Li XR, Zhang L, et al. Therapeutic response is associated with antipsychotic-induced weight gain in drug-naive first-episode patients with schizophrenia: an 8-week prospective study. J Clin Psychiatry. 2021;82(3):20m13469. doi: 10.4088/JCP.20m13469

40. Insel TR. RAISE-ing our expectations for first-episode psychosis. Am J Psychiatry. 2016;173(4):311-312.

41. Tandon R, Goldman M. Overview of neurobiology. In: Janicak PG, Marder SR, Tandon R, et al, eds. Schizophrenia: recent advances in diagnosis and treatment. Springer; 2014:27-33.

42. National Institutes of Health. Accelerating Medicines Partnership. Schizophrenia. Accessed August 30, 2021. https://www.nih.gov/research-training/accelerating-medicines-partnership-amp/schizophrenia

43. Guloksuz S, van Os J. The slow death of the concept of schizophrenia and the painful birth of the psychosis spectrum. Psychol Med. 2018;48(2):229-244.

44. Christ-Crain M, Bichet DG, Fenske WK, et al. Diabetes insipidus. Nat Rev Dis Primers. 2019;5(1):54.

45. Ahmadi L, Goldman MB. Primary polydipsia: update. Best Pract Res Clin Endocrinol Metab. 2020;34(5):101469. doi: 10.1016/j.beem.2020.101469

46. Early Assessment and Support Alliance. National Early Psychosis Directory. Accessed August 30, 2021. https://easacommunity.org/national-directory.php

Article PDF
Author and Disclosure Information

Morris B. Goldman, MD
Associate Professor of Psychiatry and Behavioral Sciences

Marko Mihailovic, MA, LCPC
Clinical Research Manager

Philip G. Janicak, MD
Adjunct Professor of Psychiatry and Behavioral Sciences

Northwestern University
Chicago, Illinois

Disclosures
Dr. Goldman has served as a speaker for PsychU/Otsuka, received research grants from the National Institute of Mental Health, and received income from Sunovion. Mr. Mihailovic reports no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products. Dr. Janicak has served as a consultant to TMS Solutions and Neuronetics and as a speaker for PsychU/Otsuka.

Issue
Current Psychiatry - 20(10)
Publications
Topics
Page Number
12-13,24-31
Sections
Author and Disclosure Information

Morris B. Goldman, MD
Associate Professor of Psychiatry and Behavioral Sciences

Marko Mihailovic, MA, LCPC
Clinical Research Manager

Philip G. Janicak, MD
Adjunct Professor of Psychiatry and Behavioral Sciences

Northwestern University
Chicago, Illinois

Disclosures
Dr. Goldman has served as a speaker for PsychU/Otsuka, received research grants from the National Institute of Mental Health, and received income from Sunovion. Mr. Mihailovic reports no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products. Dr. Janicak has served as a consultant to TMS Solutions and Neuronetics and as a speaker for PsychU/Otsuka.

Author and Disclosure Information

Morris B. Goldman, MD
Associate Professor of Psychiatry and Behavioral Sciences

Marko Mihailovic, MA, LCPC
Clinical Research Manager

Philip G. Janicak, MD
Adjunct Professor of Psychiatry and Behavioral Sciences

Northwestern University
Chicago, Illinois

Disclosures
Dr. Goldman has served as a speaker for PsychU/Otsuka, received research grants from the National Institute of Mental Health, and received income from Sunovion. Mr. Mihailovic reports no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products. Dr. Janicak has served as a consultant to TMS Solutions and Neuronetics and as a speaker for PsychU/Otsuka.

Article PDF
Article PDF

Neuroscience research over the past half century has failed to significantly advance the treatment of severe mental illness.1,2 Hence, evidence that a longer duration of untreated psychosis (DUP) aggravates—and early intervention with medication and social supports ameliorates—the long-term adverse consequences of psychotic disorders generated a great deal of interest.3,4 This knowledge led to the development of diverse early intervention services worldwide aimed at this putative “critical window.” It raised the possibility that appropriate interventions could prevent the long-term disability that makes chronic psychosis one of the most debilitating disorders.5,6 However, even beyond the varied cultural and economic confounds, it is difficult to assess, compare, and optimize program effectiveness.7 Obstacles include paucity of sufficiently powered, well-designed randomized controlled trials (RCTs), the absence of diagnostic biomarkers or other prognostic indicators to better account for the inherent heterogeneity in the population and associated outcomes, and the absence of modifiable risk factors that can guide interventions and provide intermediate outcomes.4,8-10

To better appreciate these issues, it is important to distinguish whether a program is designed to prevent psychosis, or to mitigate the effects of psychosis. Two models include the:

  • Prevention model, which focuses on young individuals who are not yet overtly psychotic but at high risk
  • First-episode recovery model, which focuses on those who have experienced a first episode of psychosis (FEP) but have not yet developed a chronic disorder.

Both models share long-term goals and are hampered by many of the same issues summarized above. They both deviate markedly from the standard medical model by including psychosocial services designed to promote restoration of a self-defined trajectory to greater independence.11-14 The 2 differ, however, in the challenges they must overcome to produce their sample populations and establish effective interventions.10,15,16

In this article, we provide a succinct overview of these issues and a set of recommendations based on a “strength-based” approach. This approach focuses on finding common ground between patients, their support system, and the treatment team in the service of empowering patients to resume responsibility for transition to adulthood.

The prevention model

While most prevention initiatives in medicine rely on the growing ability to target specific pathophysiologic pathways,3 preventing psychosis relies on clinical evidence showing that DUP and early interventions predict a better course of severe mental illness.17 In contrast, initiatives such as normalizing neonatal neuronal pathways are more consistent with the strategy utilized in other fields but have yet to yield a pathophysiologic target for psychosis.3,18

Initial efforts to identify ‘at-risk’ individuals

The prevention model of psychosis is based on the ability to identify young individuals at high risk for developing a psychotic disorder (Figure). The first screening measures were focused on prodromal psychosis (eg, significant loss of function, family history, and “intermittent” and “attenuated” psychotic symptoms). When applied to referred (ie, pre-screened) samples, 30% to 40% of this group who met criteria transitioned to psychosis over the next 1 to 3 years despite antidepressant and psychosocial interventions.19 Comprising 8 academic medical centers, the North American Prodrome Longitudinal Study (NAPLS) produced similar results using the Structured Interview for Prodromal Syndromes (SIPS).17 Thus, 30% to 50% of pre-screened individuals referred by school counselors and mental health professionals met SIPS criteria, and 35% of these individuals transitioned to psychosis over 30 months. The validity of this measure was further supported by the fact that higher baseline levels of unusual thought content, suspicion/paranoia, social impairment, and substance abuse successfully distinguished approximately 80% of those who transitioned to psychosis. The results of this first generation of screening studies were exciting because they seemed to demonstrate that highly concentrated samples of young persons at high risk of developing psychosis could be identified, and that fine-tuning the screening criteria could produce even more enriched samples (ie, positive predictive power).

CP02010012_f1.PNG

Initial interventions produced promising results

The development of effective screening measures led to reports of effective treatment interventions. These were largely applied in a clinical staging model that restricted antipsychotic medications to those who failed to improve after receiving potentially “less toxic” interventions (eg, omega-3 polyunsaturated fatty acids and other antioxidants; psychotherapy; cognitive-behavioral therapy [CBT]; family therapy).5 While study designs were typically quasi-experimental, the interventions appeared to dramatically diminish the transition to psychosis (ie, approximately 50%).

Continue to: The first generation...

 

 

The first generation of RCTs appeared to confirm these results, although sample sizes were small, and most study designs assessed only a single intervention. Initial meta-analyses of these data reported that both CBT and antipsychotics appeared to prevent approximately one-half of individuals from becoming psychotic at 12 months, and more than one-third at 2 to 4 years, compared with treatment as usual.20

While some researchers challenged the validity of these findings,21-23 the results generated tremendous international enthusiasm and calls for widespread implementation.6 The number of early intervention services (EIS) centers increased dramatically worldwide, and in 2014 the National Institute for Health and Care Excellence released standards for interventions to prevent transition to psychosis.24 These included close monitoring, CBT and family interventions, and avoiding antipsychotics when possible.24

Focusing on sensitivity over specificity

The first generation of studies generated by the prevention model relied on outreach programs or referrals, which produced small samples of carefully selected, pre-screened individuals (Figure, Pre-screened) who were then screened again to establish the high-risk sample.25 While approximately 33% of these individuals became psychotic, the screening process required a very efficient means of eliminating those not at high-risk (given the ultimate target population represented only approximately .5% of young people) (Figure). The pre-screening and screening processes in these first-generation studies were labor-intensive but could only identify approximately 5% of those individuals destined to become psychotic over the next 2 or 3 years. Thus, alternative methods to enhance sensitivity were needed to extend programming to the general population.

Second-generation pre-screening (Figure; Step 1). New pre-screening methods were identified that captured more individuals destined to become psychotic. For example, approximately 90% of this population were registered in health care organizations (eg, health maintenance organizations) and received a psychiatric diagnosis in the year prior to the onset of psychosis (true positives).8 These samples, however, contained a much higher percentage of persons not destined to become psychotic, and somehow the issue of specificity (decreasing false positives) was minimized.8,9 For example, pre-screened samples contained 20 to 50 individuals not destined to become psychotic for each one who did.26 Since screening measures could only eliminate approximately 20% of this group (Figure, Step 2, page 25), second-generation transition rates fell from 30% to 40% to 2% to 10%.27,28

Other pre-screening approaches were introduced, but they also focused on capturing more of those destined to become psychotic (sensitivity) than eliminating those who would not (specificity). For instance, Australia opened more than 100 “Headspace” community centers nationwide designed to promote engagement and self-esteem in youth experiencing anxiety; depression; stress; relationship, work, or school problems; or bullying.13 Most services were free and included mental health staff who screened for psychosis and provided a wide range of services in a destigmatized setting. These methods identified at least an additional 5% to 7% of individuals destined to become psychotic, but to our knowledge, no data have been published on whether they helped eliminate those who did not.

Continue to: Second-generation screening

 

 

Second-generation screening (Figure, Step 2). A second screening aims to retain those pre-screened individuals who will become psychotic (ie, minimizing false negatives) while further minimizing those who do not (ie, minimizing false positives). The addition of cognitive, neural (eg, structural MRI; neurophysiologic), and biochemical (eg, inflammatory immune and stress) markers to the risk calculators have produced a sensitivity close to 100%.8,9 Unfortunately, these studies downplayed specificity, which remained approximately 20%.8,9 Specificity is critical not just because of concerns about stigma (ie, labeling people as pre-psychotic when they are not) but also because of the adverse effects of antipsychotic medications and the effects on future program development (interventions are costly and labor-intensive). Also, diluting the pool with individuals not at risk makes it nearly impossible to identify effective interventions (ie, power).27,28

While some studies focused on increasing specificity (to approximately 75%), this leads to an unacceptable loss of sensitivity (from 90% to 60%),29 with 40% of pre-screened individuals who would become psychotic being eliminated from the study population. The addition of other biological markers (eg, salivary cortisol)30 and use of learning health systems may be able to enhance these numbers (initial reports of specificity = 87% and sensitivity = 85%).8,9 This is accomplished by integrating artificial and human intelligence measures of clinical (symptom and neurocognitive measures) and biological (eg, polygenetic risk scores; gray matter volume) variables.31 However, even if these results are replicated, more effective pre-screening measures will be required.

Identifying a suitable sample population for prevention program studies is clearly more complicated than for FEP studies, where one can usually identify many of those in the at-risk population by their first hospitalization for psychotic symptoms. The issues of false positives (eg, substance-induced psychosis) and negatives (eg, slow deterioration, prominent negative symptoms) are important concerns, but proportionately far less significant.

 

Prevention and FEP interventions

Once a study sample is constituted, 1 to 3 years of treatment interventions are initiated. Interventions for prevention programs typically include CBT directed at attenuated psychosis (eg, reframing or de-catastrophizing unusual thoughts and minimizing distress associated with unusual perceptions); case management to facilitate personal, educational, and vocational goals; and family therapy in single or multi-group formats to educate one’s support system about the risk state and to minimize adverse familial responses.14 Many programs also include supported education or employment services to promote reintegration in age-appropriate activities; group therapy focused on substance abuse and social skills training; cognitive remediation to ameliorate the cognitive dysfunction; and an array of pharmacologic interventions designed to delay or prevent transition to psychosis or to alleviate symptoms. While most interventions are similar, FEP programs have recently included peer support staff. This appears to instill hope in newly diagnosed patients, provide role models, and provide peer supporters an opportunity to use their experiences to help others and earn income.32

The breadth and depth of these services are critical because retention in the program is highly dependent on participant engagement, which in turn is highly dependent on whether the program can help individuals get what they want (eg, friends, employment, education, more autonomy, physical health). The setting and atmosphere of the treatment program and the willingness/ability of staff to meet participants in the community are also important elements.11,12 In this context, the Headspace community centers are having an impact far beyond Australia and may prove to be a particularly good model.13

Continue to: Assessing prevention and FEP interventions

 

 

Assessing prevention and FEP interventions

The second generation of studies of prevention programs has not confirmed, let alone extended, the earlier findings and meta-analyses. A 2020 report concluded CBT was still the most promising intervention; it was more effective than control treatments at 12 and 18 months, although not at 6, 24, or 48 months.33 This review included controlled, open-label, and naturalistic studies that assessed family therapy; omega-3 polyunsaturated fatty acids; integrated psychological therapy (a package of interventions that included family education, CBT, social skills training, and cognitive remediation); N-methyl-D-aspartate receptor modulators; mood stabilizers; and antipsychotics. In addition to the evidence supporting CBT, the results also indicated nonsignificant trends favoring family and integrated psychological therapy. Neither a 2019 Cochrane review34 nor a 2020 “umbrella” assessment of 42 meta-analyses9 found convincing evidence for the efficacy of any program components.

While these disappointing findings are at least partly attributable to the methodological challenges described above and in the Figure, other factors may hinder establishing effective interventions. In contrast to FEP studies, those focused on prevention had a very ambitious agenda (eliminating psychosis) and tended to downplay more modest intermediate outcomes. These studies also tended to assess new ideas with small samples rather than pursue promising findings with larger multi-site studies focused on a group of interventions. The authors of a Cochrane review observed “There is the impression that in this whole area there is a triumph of hope over adversity. There is the repeated hope invested in another—often unique—study question and then a study of fewer than 100 participants are completed. This results in the set of comparisons reported here, all 9 of which are too underpowered to really highlight clear differences.”34 To use a baseball analogy, it seems that investigators are “swinging for the fence” when a few singles are what’s really needed.

From the outset, the goals of FEP studies were more modest, largely ignoring the task of developing consensus definitions of recovery that require following patients for up to 5 to 10 years. Instead, they use intermediate endpoints based on adapting treatments that already appeared effective in patients with chronic mental disorders.35 As a consequence, researchers examining FEP demonstrated clear, albeit limited, salutary effects using large multi-site trials and previously established outcome measures.3,10,36 For instance, the Recovery After an Initial Schizophrenia Episode-Early Treatment Program (RAISE-ETP) study was a 2-year, multi-site RCT (N = 404) funded by the National Institute of Mental Health (NIMH). The investigators reported improved indices of social function (eg, quality of life; education and work participation) and total ratings of psychopathology and depression compared with treatment as usual. Furthermore, they established that DUP predicted treatment response.35 The latter finding was underscored by improvement being limited to the 50% with <74 weeks DUP. Annual costs of the program per 1 standard deviation improvement in quality of life were approximately $1,000 for patients with <74 weeks DUP and $40,000 for those with >74 weeks DUP. Concurrent meta-analyses confirmed and extended these findings,16 showing higher remission rates; diminished relapses and hospital admissions; greater engagement in programming; greater involvement in work and school; improved quality of life; and other steps toward recovery. These studies were also able to establish a clear benefit of antipsychotic medications, particularly a high acceptance of long-acting injectable antipsychotic formulations, which promoted adherence and decreased some adverse events37; and early use of clozapine therapy, which improved remission rates and longer-term outcomes.38 Other findings underscored the need to anticipate and address new problems associated with effective antipsychotic therapy (eg, antipsychotic response correlates with weight gain, a particularly intolerable adverse event for this age group).39 Providing pre-emptive strategies such as exercise groups and nutritional education may be necessary to maintain adherence.

 

Limitations of FEP studies

The effect sizes in these FEP studies were small to medium on outcome measures tracking recovery and associated indicators (eg, global functioning, school/work participation, treatment engagement); the number needed to treat for each of these was >10. There is no clear evidence that recovery programs such as RAISE-ETP actually reduce longer-term disability. Most studies showed disability payments increased while clinical benefits tended to fade over time. In addition, by grouping interventions together, the studies made it difficult to identify effective vs ineffective treatments, let alone determine how best to personalize therapy for participants in future studies.

The next generation of FEP studies

While limited in scope, the results of the recent FEP studies justify a next generation of recovery interventions designed to address these shortcomings and optimize program outcomes.39 Most previous FEP studies were conducted in community mental health center settings, thus eliminating the need to transition services developed in academia into the “real world.” The next generation of NIMH studies will be primarily conducted in analogous settings under the Early Psychosis Intervention Network (EPINET).40 EPINET’s study design echoes that responsible for the stepwise successes in the late 20th century that produced cures for the deadliest childhood cancer, acute lymphoblastic leukemia (ALL). This disease was successfully treated by modifying diverse evidence-based practices without relying on pharmacologic or other major treatment breakthroughs. Despite this, the effort yielded successful personalized interventions that were not obtainable for other severe childhood conditions.40 EPINET hopes to automate much of these stepwise advances with a learning health system. This program relies on data routinely collected in clinical practice to drive the process of scientific discovery. Specifically, it determines the relationships between clinical features, biologic measures, treatment characteristics, and symptomatic and functional outcomes. EPINET aims to accelerate our understanding of biomarkers of psychosis risk and onset, as well as factors associated with recovery and cure. Dashboard displays of outcomes will allow for real-time comparisons within and across early intervention clinics. This in turn identifies performance gaps and drives continuous quality improvement.

Continue to: Barriers to optimizing program efficacy for both models

 

 

Barriers to optimizing program efficacy for both models

Unfortunately, there are stark differences between ALL and severe mental disorders that potentially jeopardize the achievement of these aims, despite the advances in data analytic abilities that drive the learning health system. Specifically, the heterogeneity of psychotic illnesses and the absence of reliable prognostic and modifiable risk markers (responsible for failed efforts to enhance treatment of serious mental illness over the last half century1,2,41) are unlikely to be resolved by a learning health system. These measures are vital to determine whether specific interventions are effective, particularly given the absence of a randomized control group in the EPINET/learning health system design. Fortunately, however, the National Institutes for Health has recently initiated the Accelerating Medicines Partnership–Schizophrenia (AMP-SCZ). This approach seeks “promising biological markers that can help identify those at risk of developing schizophrenia as early as possible, track the progression of symptoms and other outcomes and ultimately define targets for treatment development.”42 The Box1,4,9,10,36,41,43-45 describes some of the challenges involved in identifying biomarkers of severe mental illness.

Box

Identifying biomarkers of severe mental illness

Biomarkers and modifiable risk factors4,9,10,41,43 are at the core of personalized medicine and its ultimate objective (ie, theragnostics). This is the ability to identify the correct intervention for a disorder based on a biomarker of the illness.10,36 The inability to identify biomarkers of severe mental illness is multifactorial but in part may be attributable to “looking in all the wrong places.”41 By focusing on neural processes that generate psychiatric symptomatology, investigators are assuming they can bridge the “mind gap”1 and specifically distinguish between pathological, compensatory, or collateral measures of poorly characterized limbic neural functions.41

It may be more productive to identify a pathological process within the limbic system that produces a medical condition as well as the mental disorder. If one can isolate the pathologic limbic circuit activity responsible for a medical condition, one may be able to reproduce this in animal models and determine whether analogous processes contribute to the core features of the mental illness. Characterization of the aberrant neural circuit in animal models also could yield targets for future therapies. For example, episodic water intoxication in a discrete subset of patients with schizophrenia44 appears to arise from a stress diathesis produced by anterior hippocampal pathology that disrupts regulation of antidiuretic hormone, oxytocin, and hypothalamic-pituitary-adrenal axis secretion. These patients also exhibit psychogenic polydipsia that may be a consequence of the same hippocampal pathology that disrupts ventral striatal and lateral hypothalamic circuits. These circuits, in turn, also modulate motivated behaviors and cognitive processes likely relevant to psychosis.45

A strength-based approach

The absence of sufficiently powered RCTs for prevention studies and the reliance on intermediate outcomes for FEP studies leaves unanswered whether such programs can effectively prevent chronic psychosis at a cost society is willing to pay. Still, substantial evidence indicates that outreach, long-acting injectable antipsychotics, early consideration of clozapine, family therapy, CBT for psychosis/attenuated psychosis, and services focused on competitive employment can preserve social and occupational functioning.16,34 Until these broader questions are more definitively addressed, it seems reasonable to apply what we have learned (Table11,12,35,37-39,46).

CP02010012_t1.PNG

Simply avoiding the most divisive aspects of the medical model that inadvertently promote stigma and undercut self-confidence may help maintain patients’ willingness to learn how best to apply their strengths and manage their limitations.11 The progression to enduring psychotic features (eg, fixed delusions) may reflect ongoing social isolation and alienation. A strength-based approach seeks first to establish common goals (eg, school, work, friends, family support, housing, leaving home) and then works to empower the patient to successfully reach those goals.35 This typically involves giving them the opportunity to fail, avoiding criticism when they do, and focusing on these experiences as learning opportunities from which success can ultimately result.

It is difficult to offer all these services in a typical private practice setting. Instead, it may make more sense to use one of the hundreds of early intervention services programs in the United States.46 If a psychiatric clinician is dedicated to working with this population, it may also be possible to establish ongoing relationships with primary care physicians, family and CBT therapists, family support services (eg, National Alliance on Mental Illness), caseworkers and employment counselors. In essence, a psychiatrist may be able re-create a multidisciplinary effort by taking advantage of the expertise of these various professionals. The challenge is to create a consistent message for patients and families in the absence of regular meetings with the clinical team, although the recent reliance on and improved sophistication of virtual meetings may help. Psychiatrists often play a critical role even when the patient is not prescribed medication, partly because they are most comfortable handling the risks and may have the most comprehensive understanding of the issues at play. When medications are appropriate and patients with FEP are willing to take them, early consideration of long-acting injectable antipsychotics and clozapine may provide better stabilization and diminish the risk of earlier and more frequent relapses.

Bottom Line

Early interventions for psychosis include the prevention model and the first-episode recovery model. It is difficult to assess, compare, and optimize the effectiveness of such programs. Current evidence supports a ‘strength-based’ approach focused on finding common ground between patients, their support system, and the treatment team.

Related Resources

  • Early Assessment and Support Alliance. National Early Psychosis Directory. https://easacommunity.org/nationaldirectory.php
  • Kane JM, Robinson DG, Schooler NR, et al. Comprehensive versus usual community care for first-episode psychosis: 2-year outcomes from the NIMH RAISE Early Treatment Program. Am J Psychiatry. 2016 ;173(4):362-372

Drug Brand Name

Clozapine • Clozaril

Neuroscience research over the past half century has failed to significantly advance the treatment of severe mental illness.1,2 Hence, evidence that a longer duration of untreated psychosis (DUP) aggravates—and early intervention with medication and social supports ameliorates—the long-term adverse consequences of psychotic disorders generated a great deal of interest.3,4 This knowledge led to the development of diverse early intervention services worldwide aimed at this putative “critical window.” It raised the possibility that appropriate interventions could prevent the long-term disability that makes chronic psychosis one of the most debilitating disorders.5,6 However, even beyond the varied cultural and economic confounds, it is difficult to assess, compare, and optimize program effectiveness.7 Obstacles include paucity of sufficiently powered, well-designed randomized controlled trials (RCTs), the absence of diagnostic biomarkers or other prognostic indicators to better account for the inherent heterogeneity in the population and associated outcomes, and the absence of modifiable risk factors that can guide interventions and provide intermediate outcomes.4,8-10

To better appreciate these issues, it is important to distinguish whether a program is designed to prevent psychosis, or to mitigate the effects of psychosis. Two models include the:

  • Prevention model, which focuses on young individuals who are not yet overtly psychotic but at high risk
  • First-episode recovery model, which focuses on those who have experienced a first episode of psychosis (FEP) but have not yet developed a chronic disorder.

Both models share long-term goals and are hampered by many of the same issues summarized above. They both deviate markedly from the standard medical model by including psychosocial services designed to promote restoration of a self-defined trajectory to greater independence.11-14 The 2 differ, however, in the challenges they must overcome to produce their sample populations and establish effective interventions.10,15,16

In this article, we provide a succinct overview of these issues and a set of recommendations based on a “strength-based” approach. This approach focuses on finding common ground between patients, their support system, and the treatment team in the service of empowering patients to resume responsibility for transition to adulthood.

The prevention model

While most prevention initiatives in medicine rely on the growing ability to target specific pathophysiologic pathways,3 preventing psychosis relies on clinical evidence showing that DUP and early interventions predict a better course of severe mental illness.17 In contrast, initiatives such as normalizing neonatal neuronal pathways are more consistent with the strategy utilized in other fields but have yet to yield a pathophysiologic target for psychosis.3,18

Initial efforts to identify ‘at-risk’ individuals

The prevention model of psychosis is based on the ability to identify young individuals at high risk for developing a psychotic disorder (Figure). The first screening measures were focused on prodromal psychosis (eg, significant loss of function, family history, and “intermittent” and “attenuated” psychotic symptoms). When applied to referred (ie, pre-screened) samples, 30% to 40% of this group who met criteria transitioned to psychosis over the next 1 to 3 years despite antidepressant and psychosocial interventions.19 Comprising 8 academic medical centers, the North American Prodrome Longitudinal Study (NAPLS) produced similar results using the Structured Interview for Prodromal Syndromes (SIPS).17 Thus, 30% to 50% of pre-screened individuals referred by school counselors and mental health professionals met SIPS criteria, and 35% of these individuals transitioned to psychosis over 30 months. The validity of this measure was further supported by the fact that higher baseline levels of unusual thought content, suspicion/paranoia, social impairment, and substance abuse successfully distinguished approximately 80% of those who transitioned to psychosis. The results of this first generation of screening studies were exciting because they seemed to demonstrate that highly concentrated samples of young persons at high risk of developing psychosis could be identified, and that fine-tuning the screening criteria could produce even more enriched samples (ie, positive predictive power).

CP02010012_f1.PNG

Initial interventions produced promising results

The development of effective screening measures led to reports of effective treatment interventions. These were largely applied in a clinical staging model that restricted antipsychotic medications to those who failed to improve after receiving potentially “less toxic” interventions (eg, omega-3 polyunsaturated fatty acids and other antioxidants; psychotherapy; cognitive-behavioral therapy [CBT]; family therapy).5 While study designs were typically quasi-experimental, the interventions appeared to dramatically diminish the transition to psychosis (ie, approximately 50%).

Continue to: The first generation...

 

 

The first generation of RCTs appeared to confirm these results, although sample sizes were small, and most study designs assessed only a single intervention. Initial meta-analyses of these data reported that both CBT and antipsychotics appeared to prevent approximately one-half of individuals from becoming psychotic at 12 months, and more than one-third at 2 to 4 years, compared with treatment as usual.20

While some researchers challenged the validity of these findings,21-23 the results generated tremendous international enthusiasm and calls for widespread implementation.6 The number of early intervention services (EIS) centers increased dramatically worldwide, and in 2014 the National Institute for Health and Care Excellence released standards for interventions to prevent transition to psychosis.24 These included close monitoring, CBT and family interventions, and avoiding antipsychotics when possible.24

Focusing on sensitivity over specificity

The first generation of studies generated by the prevention model relied on outreach programs or referrals, which produced small samples of carefully selected, pre-screened individuals (Figure, Pre-screened) who were then screened again to establish the high-risk sample.25 While approximately 33% of these individuals became psychotic, the screening process required a very efficient means of eliminating those not at high-risk (given the ultimate target population represented only approximately .5% of young people) (Figure). The pre-screening and screening processes in these first-generation studies were labor-intensive but could only identify approximately 5% of those individuals destined to become psychotic over the next 2 or 3 years. Thus, alternative methods to enhance sensitivity were needed to extend programming to the general population.

Second-generation pre-screening (Figure; Step 1). New pre-screening methods were identified that captured more individuals destined to become psychotic. For example, approximately 90% of this population were registered in health care organizations (eg, health maintenance organizations) and received a psychiatric diagnosis in the year prior to the onset of psychosis (true positives).8 These samples, however, contained a much higher percentage of persons not destined to become psychotic, and somehow the issue of specificity (decreasing false positives) was minimized.8,9 For example, pre-screened samples contained 20 to 50 individuals not destined to become psychotic for each one who did.26 Since screening measures could only eliminate approximately 20% of this group (Figure, Step 2, page 25), second-generation transition rates fell from 30% to 40% to 2% to 10%.27,28

Other pre-screening approaches were introduced, but they also focused on capturing more of those destined to become psychotic (sensitivity) than eliminating those who would not (specificity). For instance, Australia opened more than 100 “Headspace” community centers nationwide designed to promote engagement and self-esteem in youth experiencing anxiety; depression; stress; relationship, work, or school problems; or bullying.13 Most services were free and included mental health staff who screened for psychosis and provided a wide range of services in a destigmatized setting. These methods identified at least an additional 5% to 7% of individuals destined to become psychotic, but to our knowledge, no data have been published on whether they helped eliminate those who did not.

Continue to: Second-generation screening

 

 

Second-generation screening (Figure, Step 2). A second screening aims to retain those pre-screened individuals who will become psychotic (ie, minimizing false negatives) while further minimizing those who do not (ie, minimizing false positives). The addition of cognitive, neural (eg, structural MRI; neurophysiologic), and biochemical (eg, inflammatory immune and stress) markers to the risk calculators have produced a sensitivity close to 100%.8,9 Unfortunately, these studies downplayed specificity, which remained approximately 20%.8,9 Specificity is critical not just because of concerns about stigma (ie, labeling people as pre-psychotic when they are not) but also because of the adverse effects of antipsychotic medications and the effects on future program development (interventions are costly and labor-intensive). Also, diluting the pool with individuals not at risk makes it nearly impossible to identify effective interventions (ie, power).27,28

While some studies focused on increasing specificity (to approximately 75%), this leads to an unacceptable loss of sensitivity (from 90% to 60%),29 with 40% of pre-screened individuals who would become psychotic being eliminated from the study population. The addition of other biological markers (eg, salivary cortisol)30 and use of learning health systems may be able to enhance these numbers (initial reports of specificity = 87% and sensitivity = 85%).8,9 This is accomplished by integrating artificial and human intelligence measures of clinical (symptom and neurocognitive measures) and biological (eg, polygenetic risk scores; gray matter volume) variables.31 However, even if these results are replicated, more effective pre-screening measures will be required.

Identifying a suitable sample population for prevention program studies is clearly more complicated than for FEP studies, where one can usually identify many of those in the at-risk population by their first hospitalization for psychotic symptoms. The issues of false positives (eg, substance-induced psychosis) and negatives (eg, slow deterioration, prominent negative symptoms) are important concerns, but proportionately far less significant.

 

Prevention and FEP interventions

Once a study sample is constituted, 1 to 3 years of treatment interventions are initiated. Interventions for prevention programs typically include CBT directed at attenuated psychosis (eg, reframing or de-catastrophizing unusual thoughts and minimizing distress associated with unusual perceptions); case management to facilitate personal, educational, and vocational goals; and family therapy in single or multi-group formats to educate one’s support system about the risk state and to minimize adverse familial responses.14 Many programs also include supported education or employment services to promote reintegration in age-appropriate activities; group therapy focused on substance abuse and social skills training; cognitive remediation to ameliorate the cognitive dysfunction; and an array of pharmacologic interventions designed to delay or prevent transition to psychosis or to alleviate symptoms. While most interventions are similar, FEP programs have recently included peer support staff. This appears to instill hope in newly diagnosed patients, provide role models, and provide peer supporters an opportunity to use their experiences to help others and earn income.32

The breadth and depth of these services are critical because retention in the program is highly dependent on participant engagement, which in turn is highly dependent on whether the program can help individuals get what they want (eg, friends, employment, education, more autonomy, physical health). The setting and atmosphere of the treatment program and the willingness/ability of staff to meet participants in the community are also important elements.11,12 In this context, the Headspace community centers are having an impact far beyond Australia and may prove to be a particularly good model.13

Continue to: Assessing prevention and FEP interventions

 

 

Assessing prevention and FEP interventions

The second generation of studies of prevention programs has not confirmed, let alone extended, the earlier findings and meta-analyses. A 2020 report concluded CBT was still the most promising intervention; it was more effective than control treatments at 12 and 18 months, although not at 6, 24, or 48 months.33 This review included controlled, open-label, and naturalistic studies that assessed family therapy; omega-3 polyunsaturated fatty acids; integrated psychological therapy (a package of interventions that included family education, CBT, social skills training, and cognitive remediation); N-methyl-D-aspartate receptor modulators; mood stabilizers; and antipsychotics. In addition to the evidence supporting CBT, the results also indicated nonsignificant trends favoring family and integrated psychological therapy. Neither a 2019 Cochrane review34 nor a 2020 “umbrella” assessment of 42 meta-analyses9 found convincing evidence for the efficacy of any program components.

While these disappointing findings are at least partly attributable to the methodological challenges described above and in the Figure, other factors may hinder establishing effective interventions. In contrast to FEP studies, those focused on prevention had a very ambitious agenda (eliminating psychosis) and tended to downplay more modest intermediate outcomes. These studies also tended to assess new ideas with small samples rather than pursue promising findings with larger multi-site studies focused on a group of interventions. The authors of a Cochrane review observed “There is the impression that in this whole area there is a triumph of hope over adversity. There is the repeated hope invested in another—often unique—study question and then a study of fewer than 100 participants are completed. This results in the set of comparisons reported here, all 9 of which are too underpowered to really highlight clear differences.”34 To use a baseball analogy, it seems that investigators are “swinging for the fence” when a few singles are what’s really needed.

From the outset, the goals of FEP studies were more modest, largely ignoring the task of developing consensus definitions of recovery that require following patients for up to 5 to 10 years. Instead, they use intermediate endpoints based on adapting treatments that already appeared effective in patients with chronic mental disorders.35 As a consequence, researchers examining FEP demonstrated clear, albeit limited, salutary effects using large multi-site trials and previously established outcome measures.3,10,36 For instance, the Recovery After an Initial Schizophrenia Episode-Early Treatment Program (RAISE-ETP) study was a 2-year, multi-site RCT (N = 404) funded by the National Institute of Mental Health (NIMH). The investigators reported improved indices of social function (eg, quality of life; education and work participation) and total ratings of psychopathology and depression compared with treatment as usual. Furthermore, they established that DUP predicted treatment response.35 The latter finding was underscored by improvement being limited to the 50% with <74 weeks DUP. Annual costs of the program per 1 standard deviation improvement in quality of life were approximately $1,000 for patients with <74 weeks DUP and $40,000 for those with >74 weeks DUP. Concurrent meta-analyses confirmed and extended these findings,16 showing higher remission rates; diminished relapses and hospital admissions; greater engagement in programming; greater involvement in work and school; improved quality of life; and other steps toward recovery. These studies were also able to establish a clear benefit of antipsychotic medications, particularly a high acceptance of long-acting injectable antipsychotic formulations, which promoted adherence and decreased some adverse events37; and early use of clozapine therapy, which improved remission rates and longer-term outcomes.38 Other findings underscored the need to anticipate and address new problems associated with effective antipsychotic therapy (eg, antipsychotic response correlates with weight gain, a particularly intolerable adverse event for this age group).39 Providing pre-emptive strategies such as exercise groups and nutritional education may be necessary to maintain adherence.

 

Limitations of FEP studies

The effect sizes in these FEP studies were small to medium on outcome measures tracking recovery and associated indicators (eg, global functioning, school/work participation, treatment engagement); the number needed to treat for each of these was >10. There is no clear evidence that recovery programs such as RAISE-ETP actually reduce longer-term disability. Most studies showed disability payments increased while clinical benefits tended to fade over time. In addition, by grouping interventions together, the studies made it difficult to identify effective vs ineffective treatments, let alone determine how best to personalize therapy for participants in future studies.

The next generation of FEP studies

While limited in scope, the results of the recent FEP studies justify a next generation of recovery interventions designed to address these shortcomings and optimize program outcomes.39 Most previous FEP studies were conducted in community mental health center settings, thus eliminating the need to transition services developed in academia into the “real world.” The next generation of NIMH studies will be primarily conducted in analogous settings under the Early Psychosis Intervention Network (EPINET).40 EPINET’s study design echoes that responsible for the stepwise successes in the late 20th century that produced cures for the deadliest childhood cancer, acute lymphoblastic leukemia (ALL). This disease was successfully treated by modifying diverse evidence-based practices without relying on pharmacologic or other major treatment breakthroughs. Despite this, the effort yielded successful personalized interventions that were not obtainable for other severe childhood conditions.40 EPINET hopes to automate much of these stepwise advances with a learning health system. This program relies on data routinely collected in clinical practice to drive the process of scientific discovery. Specifically, it determines the relationships between clinical features, biologic measures, treatment characteristics, and symptomatic and functional outcomes. EPINET aims to accelerate our understanding of biomarkers of psychosis risk and onset, as well as factors associated with recovery and cure. Dashboard displays of outcomes will allow for real-time comparisons within and across early intervention clinics. This in turn identifies performance gaps and drives continuous quality improvement.

Continue to: Barriers to optimizing program efficacy for both models

 

 

Barriers to optimizing program efficacy for both models

Unfortunately, there are stark differences between ALL and severe mental disorders that potentially jeopardize the achievement of these aims, despite the advances in data analytic abilities that drive the learning health system. Specifically, the heterogeneity of psychotic illnesses and the absence of reliable prognostic and modifiable risk markers (responsible for failed efforts to enhance treatment of serious mental illness over the last half century1,2,41) are unlikely to be resolved by a learning health system. These measures are vital to determine whether specific interventions are effective, particularly given the absence of a randomized control group in the EPINET/learning health system design. Fortunately, however, the National Institutes for Health has recently initiated the Accelerating Medicines Partnership–Schizophrenia (AMP-SCZ). This approach seeks “promising biological markers that can help identify those at risk of developing schizophrenia as early as possible, track the progression of symptoms and other outcomes and ultimately define targets for treatment development.”42 The Box1,4,9,10,36,41,43-45 describes some of the challenges involved in identifying biomarkers of severe mental illness.

Box

Identifying biomarkers of severe mental illness

Biomarkers and modifiable risk factors4,9,10,41,43 are at the core of personalized medicine and its ultimate objective (ie, theragnostics). This is the ability to identify the correct intervention for a disorder based on a biomarker of the illness.10,36 The inability to identify biomarkers of severe mental illness is multifactorial but in part may be attributable to “looking in all the wrong places.”41 By focusing on neural processes that generate psychiatric symptomatology, investigators are assuming they can bridge the “mind gap”1 and specifically distinguish between pathological, compensatory, or collateral measures of poorly characterized limbic neural functions.41

It may be more productive to identify a pathological process within the limbic system that produces a medical condition as well as the mental disorder. If one can isolate the pathologic limbic circuit activity responsible for a medical condition, one may be able to reproduce this in animal models and determine whether analogous processes contribute to the core features of the mental illness. Characterization of the aberrant neural circuit in animal models also could yield targets for future therapies. For example, episodic water intoxication in a discrete subset of patients with schizophrenia44 appears to arise from a stress diathesis produced by anterior hippocampal pathology that disrupts regulation of antidiuretic hormone, oxytocin, and hypothalamic-pituitary-adrenal axis secretion. These patients also exhibit psychogenic polydipsia that may be a consequence of the same hippocampal pathology that disrupts ventral striatal and lateral hypothalamic circuits. These circuits, in turn, also modulate motivated behaviors and cognitive processes likely relevant to psychosis.45

A strength-based approach

The absence of sufficiently powered RCTs for prevention studies and the reliance on intermediate outcomes for FEP studies leaves unanswered whether such programs can effectively prevent chronic psychosis at a cost society is willing to pay. Still, substantial evidence indicates that outreach, long-acting injectable antipsychotics, early consideration of clozapine, family therapy, CBT for psychosis/attenuated psychosis, and services focused on competitive employment can preserve social and occupational functioning.16,34 Until these broader questions are more definitively addressed, it seems reasonable to apply what we have learned (Table11,12,35,37-39,46).

CP02010012_t1.PNG

Simply avoiding the most divisive aspects of the medical model that inadvertently promote stigma and undercut self-confidence may help maintain patients’ willingness to learn how best to apply their strengths and manage their limitations.11 The progression to enduring psychotic features (eg, fixed delusions) may reflect ongoing social isolation and alienation. A strength-based approach seeks first to establish common goals (eg, school, work, friends, family support, housing, leaving home) and then works to empower the patient to successfully reach those goals.35 This typically involves giving them the opportunity to fail, avoiding criticism when they do, and focusing on these experiences as learning opportunities from which success can ultimately result.

It is difficult to offer all these services in a typical private practice setting. Instead, it may make more sense to use one of the hundreds of early intervention services programs in the United States.46 If a psychiatric clinician is dedicated to working with this population, it may also be possible to establish ongoing relationships with primary care physicians, family and CBT therapists, family support services (eg, National Alliance on Mental Illness), caseworkers and employment counselors. In essence, a psychiatrist may be able re-create a multidisciplinary effort by taking advantage of the expertise of these various professionals. The challenge is to create a consistent message for patients and families in the absence of regular meetings with the clinical team, although the recent reliance on and improved sophistication of virtual meetings may help. Psychiatrists often play a critical role even when the patient is not prescribed medication, partly because they are most comfortable handling the risks and may have the most comprehensive understanding of the issues at play. When medications are appropriate and patients with FEP are willing to take them, early consideration of long-acting injectable antipsychotics and clozapine may provide better stabilization and diminish the risk of earlier and more frequent relapses.

Bottom Line

Early interventions for psychosis include the prevention model and the first-episode recovery model. It is difficult to assess, compare, and optimize the effectiveness of such programs. Current evidence supports a ‘strength-based’ approach focused on finding common ground between patients, their support system, and the treatment team.

Related Resources

  • Early Assessment and Support Alliance. National Early Psychosis Directory. https://easacommunity.org/nationaldirectory.php
  • Kane JM, Robinson DG, Schooler NR, et al. Comprehensive versus usual community care for first-episode psychosis: 2-year outcomes from the NIMH RAISE Early Treatment Program. Am J Psychiatry. 2016 ;173(4):362-372

Drug Brand Name

Clozapine • Clozaril

References

1. Hyman SE. Revolution stalled. Sci Transl Med. 2012;4(155):155cm11. doi: 10.1126/scitranslmed.3003142

2. Harrington A. Mind fixers: psychiatry’s troubled search for the biology of mental illness. W.W. Norton & Company; 2019.

3. Millan MJ, Andrieux A, Bartzokis G, et al. Altering the course of schizophrenia: progress and perspectives. Nat Rev Drug Discov. 2016;15(7):485-515.

4. Lieberman JA, Small SA, Girgis RR. Early detection and preventive intervention in schizophrenia: from fantasy to reality. Am J Psychiatry. 2019;176(10):794-810.

5. McGorry PD, Nelson B, Nordentoft M, et al. Intervention in individuals at ultra-high risk for psychosis: a review and future directions. J Clin Psychiatry. 2009;70(9):1206-1212.

6. Csillag C, Nordentoft M, Mizuno M, et al. Early intervention in psychosis: From clinical intervention to health system implementation. Early Interv Psychiatry. 2018;12(4):757-764.

7. McGorry PD, Ratheesh A, O’Donoghue B. Early intervention—an implementation challenge for 21st century mental health care. JAMA Psychiatry. 2018;75(6):545-546.

8. Rosenheck R. Toward dissemination of secondary prevention for psychosis. Am J Psychiatry. 2018;175(5):393-394.

9. Fusar-Poli P, Salazar de Pablo G, Correll CU, et al. Prevention of psychosis: advances in detection, prognosis, and intervention. JAMA Psychiatry. 2020;77(7):755-765.

10. Oliver D, Reilly TJ, Baccaredda Boy O, et al. What causes the onset of psychosis in individuals at clinical high risk? A meta-analysis of risk and protective factors. Schizophr Bull. 2020;46(1):110-120.

11. Tindall R, Simmons M, Allott K, et al. Disengagement processes within an early intervention service for first-episode psychosis: a longitudinal, qualitative, multi-perspective study. Front Psychiatry. 2020;11:565-565.

12. Dixon LB, Holoshitz Y, Nossel I. Treatment engagement of individuals experiencing mental illness: review and update. World Psychiatry. 2016;15(1):13-20.

13. Rickwood D, Paraskakis M, Quin D, et al. Australia’s innovation in youth mental health care: The headspace centre model. Early Interv Psychiatry. 2019;13(1):159-166.

14. Woodberry KA, Shapiro DI, Bryant C, et al. Progress and future directions in research on the psychosis prodrome: a review for clinicians. Harv Rev Psychiatry. 2016;24(2):87-103.

15. Gupta T, Mittal VA. Advances in clinical staging, early intervention, and the prevention of psychosis. F1000Res. 2019;8:F1000 Faculty Rev-2027. doi: 10.12688/f1000research.20346.1

16. Correll CU, Galling B, Pawar A, et al. Comparison of early intervention services vs treatment as usual for early-phase psychosis: a systematic review, meta-analysis, and meta-regression. JAMA Psychiatry. 2018;75(6):555-565.

17. Cannon TD, Cadenhead K, Cornblatt B, et al. Prediction of psychosis in youth at high clinical risk: a multisite longitudinal study in North America. Arch Gen Psychiatry. 2008;65(1):28-37.

18. Sommer IE, Bearden CE, van Dellen E, et al. Early interventions in risk groups for schizophrenia: what are we waiting for? NPJ Schizophr. 2016;2(1):16003-16003.

19. McGorry PD, Nelson B. Clinical high risk for psychosis—not seeing the trees for the wood. JAMA Psychiatry. 2020;77(7):559-560.

20. van der Gaag M, Smit F, Bechdolf A, et al. Preventing a first episode of psychosis: meta-analysis of randomized controlled prevention trials of 12 month and longer-term follow-ups. Schizophr Res. 2013;149(1):56-62.

21. Marshall M, Rathbone J. Early intervention for psychosis. Cochrane Database Syst Rev. 2011;(6):CD004718. doi: 10.1002/14651858.CD004718.pub3

22. Heinssen RK, Insel TR. Preventing the onset of psychosis: not quite there yet. Schizophr Bull. 2015;41(1):28-29.

23. Amos AJ. Evidence that treatment prevents transition to psychosis in ultra-high-risk patients remains questionable. Schizophr Res. 2014;153(1):240.

24. National Institute for Health and Care Excellence. Psychosis and schizophrenia in adults: prevention and management. Clinical guideline [CG178]. 1.3.7 How to deliver psychological interventions. Published February 12, 2014. Updated March 1, 2014. Accessed August 30, 2021. https://www.nice.org.uk/guidance/cg178/chapter/recommendations#how-to-deliver-psychological-interventions

25. Fusar-Poli P, Werbeloff N, Rutigliano G, et al. Transdiagnostic risk calculator for the automatic detection of individuals at risk and the prediction of psychosis: second replication in an independent National Health Service Trust. Schizophr Bull. 2019;45(3):562-570.

26. Fusar-Poli P, Oliver D, Spada G, et al. The case for improved transdiagnostic detection of first-episode psychosis: electronic health record cohort study. Schizophr Res. 2021;228:547-554.

27. Fusar-Poli P. Negative psychosis prevention trials. JAMA Psychiatry. 2017;74(6):651.

28. Cuijpers P, Smit F, Furukawa TA. Most at‐risk individuals will not develop a mental disorder: the limited predictive strength of risk factors. World Psychiatry. 2021;20(2):224-225.

29. Carrión RE, Cornblatt BA, Burton CZ, et al. Personalized prediction of psychosis: external validation of the NAPLS-2 psychosis risk calculator with the EDIPPP Project. Am J Psychiatry. 2016;173(10):989-996.

30. Worthington MA, Walker EF, Addington J, et al. Incorporating cortisol into the NAPLS2 individualized risk calculator for prediction of psychosis. Schizophr Res. 2021;227:95-100.

31. Koutsouleris N, Dwyer DB, Degenhardt F, et al. Multimodal machine learning workflows for prediction of psychosis in patients with clinical high-risk syndromes and recent-onset depression. JAMA Psychiatry. 2021;78(2):195-209.

32. Simmons MB, Grace D, Fava NJ, et al. The experiences of youth mental health peer workers over time: a qualitative study with longitudinal analysis. Community Ment Health J. 2020;56(5):906-914.

33. Devoe DJ, Farris MS, Townes P, et al. Interventions and transition in youth at risk of psychosis: a systematic review and meta-analyses. J Clin Psychiatry. 2020;81(3):17r12053. doi: 10.4088/JCP.17r12053

34. Bosnjak Kuharic D, Kekin I, Hew J, et al. Interventions for prodromal stage of psychosis. Cochrane Database Syst Rev. 2019;2019(11):CD012236

35. Dixon LB, Goldman HH, Srihari VH, et al. Transforming the treatment of schizophrenia in the United States: The RAISE Initiative. Annu Rev Clin Psychol. 2018;14:237-258.

36. Friedman-Yakoobian MS, Parrish EM, Eack SM, et al. Neurocognitive and social cognitive training for youth at clinical high risk (CHR) for psychosis: a randomized controlled feasibility trial. Schizophr Res. 2020;S0920-9964(20)30461-8. doi: 10.1016/j.schres.2020.09.005

37. Kane JM, Schooler NR, Marcy P, et al. Effect of long-acting injectable antipsychotics vs usual care on time to first hospitalization in early-phase schizophrenia: a randomized clinical trial. JAMA Psychiatry. 2020;77(12):1217-1224.

38. Morrison AP, Pyle M, Maughan D, et al. Antipsychotic medication versus psychological intervention versus a combination of both in adolescents with first-episode psychosis (MAPS): a multicentre, three-arm, randomised controlled pilot and feasibility study. Lancet Psychiatry. 2020;7(9):788-800.

39. Chen YQ, Li XR, Zhang L, et al. Therapeutic response is associated with antipsychotic-induced weight gain in drug-naive first-episode patients with schizophrenia: an 8-week prospective study. J Clin Psychiatry. 2021;82(3):20m13469. doi: 10.4088/JCP.20m13469

40. Insel TR. RAISE-ing our expectations for first-episode psychosis. Am J Psychiatry. 2016;173(4):311-312.

41. Tandon R, Goldman M. Overview of neurobiology. In: Janicak PG, Marder SR, Tandon R, et al, eds. Schizophrenia: recent advances in diagnosis and treatment. Springer; 2014:27-33.

42. National Institutes of Health. Accelerating Medicines Partnership. Schizophrenia. Accessed August 30, 2021. https://www.nih.gov/research-training/accelerating-medicines-partnership-amp/schizophrenia

43. Guloksuz S, van Os J. The slow death of the concept of schizophrenia and the painful birth of the psychosis spectrum. Psychol Med. 2018;48(2):229-244.

44. Christ-Crain M, Bichet DG, Fenske WK, et al. Diabetes insipidus. Nat Rev Dis Primers. 2019;5(1):54.

45. Ahmadi L, Goldman MB. Primary polydipsia: update. Best Pract Res Clin Endocrinol Metab. 2020;34(5):101469. doi: 10.1016/j.beem.2020.101469

46. Early Assessment and Support Alliance. National Early Psychosis Directory. Accessed August 30, 2021. https://easacommunity.org/national-directory.php

References

1. Hyman SE. Revolution stalled. Sci Transl Med. 2012;4(155):155cm11. doi: 10.1126/scitranslmed.3003142

2. Harrington A. Mind fixers: psychiatry’s troubled search for the biology of mental illness. W.W. Norton & Company; 2019.

3. Millan MJ, Andrieux A, Bartzokis G, et al. Altering the course of schizophrenia: progress and perspectives. Nat Rev Drug Discov. 2016;15(7):485-515.

4. Lieberman JA, Small SA, Girgis RR. Early detection and preventive intervention in schizophrenia: from fantasy to reality. Am J Psychiatry. 2019;176(10):794-810.

5. McGorry PD, Nelson B, Nordentoft M, et al. Intervention in individuals at ultra-high risk for psychosis: a review and future directions. J Clin Psychiatry. 2009;70(9):1206-1212.

6. Csillag C, Nordentoft M, Mizuno M, et al. Early intervention in psychosis: From clinical intervention to health system implementation. Early Interv Psychiatry. 2018;12(4):757-764.

7. McGorry PD, Ratheesh A, O’Donoghue B. Early intervention—an implementation challenge for 21st century mental health care. JAMA Psychiatry. 2018;75(6):545-546.

8. Rosenheck R. Toward dissemination of secondary prevention for psychosis. Am J Psychiatry. 2018;175(5):393-394.

9. Fusar-Poli P, Salazar de Pablo G, Correll CU, et al. Prevention of psychosis: advances in detection, prognosis, and intervention. JAMA Psychiatry. 2020;77(7):755-765.

10. Oliver D, Reilly TJ, Baccaredda Boy O, et al. What causes the onset of psychosis in individuals at clinical high risk? A meta-analysis of risk and protective factors. Schizophr Bull. 2020;46(1):110-120.

11. Tindall R, Simmons M, Allott K, et al. Disengagement processes within an early intervention service for first-episode psychosis: a longitudinal, qualitative, multi-perspective study. Front Psychiatry. 2020;11:565-565.

12. Dixon LB, Holoshitz Y, Nossel I. Treatment engagement of individuals experiencing mental illness: review and update. World Psychiatry. 2016;15(1):13-20.

13. Rickwood D, Paraskakis M, Quin D, et al. Australia’s innovation in youth mental health care: The headspace centre model. Early Interv Psychiatry. 2019;13(1):159-166.

14. Woodberry KA, Shapiro DI, Bryant C, et al. Progress and future directions in research on the psychosis prodrome: a review for clinicians. Harv Rev Psychiatry. 2016;24(2):87-103.

15. Gupta T, Mittal VA. Advances in clinical staging, early intervention, and the prevention of psychosis. F1000Res. 2019;8:F1000 Faculty Rev-2027. doi: 10.12688/f1000research.20346.1

16. Correll CU, Galling B, Pawar A, et al. Comparison of early intervention services vs treatment as usual for early-phase psychosis: a systematic review, meta-analysis, and meta-regression. JAMA Psychiatry. 2018;75(6):555-565.

17. Cannon TD, Cadenhead K, Cornblatt B, et al. Prediction of psychosis in youth at high clinical risk: a multisite longitudinal study in North America. Arch Gen Psychiatry. 2008;65(1):28-37.

18. Sommer IE, Bearden CE, van Dellen E, et al. Early interventions in risk groups for schizophrenia: what are we waiting for? NPJ Schizophr. 2016;2(1):16003-16003.

19. McGorry PD, Nelson B. Clinical high risk for psychosis—not seeing the trees for the wood. JAMA Psychiatry. 2020;77(7):559-560.

20. van der Gaag M, Smit F, Bechdolf A, et al. Preventing a first episode of psychosis: meta-analysis of randomized controlled prevention trials of 12 month and longer-term follow-ups. Schizophr Res. 2013;149(1):56-62.

21. Marshall M, Rathbone J. Early intervention for psychosis. Cochrane Database Syst Rev. 2011;(6):CD004718. doi: 10.1002/14651858.CD004718.pub3

22. Heinssen RK, Insel TR. Preventing the onset of psychosis: not quite there yet. Schizophr Bull. 2015;41(1):28-29.

23. Amos AJ. Evidence that treatment prevents transition to psychosis in ultra-high-risk patients remains questionable. Schizophr Res. 2014;153(1):240.

24. National Institute for Health and Care Excellence. Psychosis and schizophrenia in adults: prevention and management. Clinical guideline [CG178]. 1.3.7 How to deliver psychological interventions. Published February 12, 2014. Updated March 1, 2014. Accessed August 30, 2021. https://www.nice.org.uk/guidance/cg178/chapter/recommendations#how-to-deliver-psychological-interventions

25. Fusar-Poli P, Werbeloff N, Rutigliano G, et al. Transdiagnostic risk calculator for the automatic detection of individuals at risk and the prediction of psychosis: second replication in an independent National Health Service Trust. Schizophr Bull. 2019;45(3):562-570.

26. Fusar-Poli P, Oliver D, Spada G, et al. The case for improved transdiagnostic detection of first-episode psychosis: electronic health record cohort study. Schizophr Res. 2021;228:547-554.

27. Fusar-Poli P. Negative psychosis prevention trials. JAMA Psychiatry. 2017;74(6):651.

28. Cuijpers P, Smit F, Furukawa TA. Most at‐risk individuals will not develop a mental disorder: the limited predictive strength of risk factors. World Psychiatry. 2021;20(2):224-225.

29. Carrión RE, Cornblatt BA, Burton CZ, et al. Personalized prediction of psychosis: external validation of the NAPLS-2 psychosis risk calculator with the EDIPPP Project. Am J Psychiatry. 2016;173(10):989-996.

30. Worthington MA, Walker EF, Addington J, et al. Incorporating cortisol into the NAPLS2 individualized risk calculator for prediction of psychosis. Schizophr Res. 2021;227:95-100.

31. Koutsouleris N, Dwyer DB, Degenhardt F, et al. Multimodal machine learning workflows for prediction of psychosis in patients with clinical high-risk syndromes and recent-onset depression. JAMA Psychiatry. 2021;78(2):195-209.

32. Simmons MB, Grace D, Fava NJ, et al. The experiences of youth mental health peer workers over time: a qualitative study with longitudinal analysis. Community Ment Health J. 2020;56(5):906-914.

33. Devoe DJ, Farris MS, Townes P, et al. Interventions and transition in youth at risk of psychosis: a systematic review and meta-analyses. J Clin Psychiatry. 2020;81(3):17r12053. doi: 10.4088/JCP.17r12053

34. Bosnjak Kuharic D, Kekin I, Hew J, et al. Interventions for prodromal stage of psychosis. Cochrane Database Syst Rev. 2019;2019(11):CD012236

35. Dixon LB, Goldman HH, Srihari VH, et al. Transforming the treatment of schizophrenia in the United States: The RAISE Initiative. Annu Rev Clin Psychol. 2018;14:237-258.

36. Friedman-Yakoobian MS, Parrish EM, Eack SM, et al. Neurocognitive and social cognitive training for youth at clinical high risk (CHR) for psychosis: a randomized controlled feasibility trial. Schizophr Res. 2020;S0920-9964(20)30461-8. doi: 10.1016/j.schres.2020.09.005

37. Kane JM, Schooler NR, Marcy P, et al. Effect of long-acting injectable antipsychotics vs usual care on time to first hospitalization in early-phase schizophrenia: a randomized clinical trial. JAMA Psychiatry. 2020;77(12):1217-1224.

38. Morrison AP, Pyle M, Maughan D, et al. Antipsychotic medication versus psychological intervention versus a combination of both in adolescents with first-episode psychosis (MAPS): a multicentre, three-arm, randomised controlled pilot and feasibility study. Lancet Psychiatry. 2020;7(9):788-800.

39. Chen YQ, Li XR, Zhang L, et al. Therapeutic response is associated with antipsychotic-induced weight gain in drug-naive first-episode patients with schizophrenia: an 8-week prospective study. J Clin Psychiatry. 2021;82(3):20m13469. doi: 10.4088/JCP.20m13469

40. Insel TR. RAISE-ing our expectations for first-episode psychosis. Am J Psychiatry. 2016;173(4):311-312.

41. Tandon R, Goldman M. Overview of neurobiology. In: Janicak PG, Marder SR, Tandon R, et al, eds. Schizophrenia: recent advances in diagnosis and treatment. Springer; 2014:27-33.

42. National Institutes of Health. Accelerating Medicines Partnership. Schizophrenia. Accessed August 30, 2021. https://www.nih.gov/research-training/accelerating-medicines-partnership-amp/schizophrenia

43. Guloksuz S, van Os J. The slow death of the concept of schizophrenia and the painful birth of the psychosis spectrum. Psychol Med. 2018;48(2):229-244.

44. Christ-Crain M, Bichet DG, Fenske WK, et al. Diabetes insipidus. Nat Rev Dis Primers. 2019;5(1):54.

45. Ahmadi L, Goldman MB. Primary polydipsia: update. Best Pract Res Clin Endocrinol Metab. 2020;34(5):101469. doi: 10.1016/j.beem.2020.101469

46. Early Assessment and Support Alliance. National Early Psychosis Directory. Accessed August 30, 2021. https://easacommunity.org/national-directory.php

Issue
Current Psychiatry - 20(10)
Issue
Current Psychiatry - 20(10)
Page Number
12-13,24-31
Page Number
12-13,24-31
Publications
Publications
Topics
Article Type
Display Headline
Early interventions for psychosis
Display Headline
Early interventions for psychosis
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
18001E4D.SIG
Disable zoom
Off

Nontraditional therapies for treatment-resistant depression: Part 2

Article Type
Changed
Fri, 10/01/2021 - 14:53
Display Headline
Nontraditional therapies for treatment-resistant depression: Part 2

When patients with major depressive disorder (MDD) do not achieve optimal outcomes after FDA-approved first-line treatments and standard adjunctive strategies, clinicians look for additional approaches to alleviate their patients’ symptoms. Recent research suggests that several “nontraditional” treatments used primarily as adjuncts to standard antidepressants have promise for treatment-resistant depression.

In Part 1 of this article (Current Psychiatry, September 2021), we examined off-label medications. In Part 2, we will review other nontraditional approaches to treatment-resistant depression, including herbal/nutraceutical agents, anti-inflammatory/immune system therapies, device-based treatments, and other alternative approaches. Importantly, some treatments also demonstrate adverse effects (Table1-32). With a careful consideration of the risk/benefit balance, this article reviews some of the better-studied nontraditional treatment options for patients with treatment-resistant depression.

CP02010032_t1.PNG

Herbal/nutraceutical agents

This category encompasses a variety of commonly available “natural” options patients often ask about and at times self-prescribe. Examples evaluated in clinical trials include:

  • vitamin D
  • essential fatty acids (omega-3, omega-6)
  • S-adenosyl-L-methionine (SAMe)
  • hypericum perforatum (St. John’s Wort)
  • probiotics.

Vitamin D deficiency has been linked to depression, possibly by lowering serotonin, norepinephrine, and dopamine concentrations.1-3

A meta-analysis of 3 prospective, observational studies (N = 8,815) found an elevated risk of affective disorders in patients with low vitamin D levels.4 In addition, a systematic review and meta-analysis supported a potential role for vitamin D supplementation for patients with treatment-resistant depresssion.5

Toxicity can occur at levels >100 ng/mL, and resulting adverse effects may include weakness, fatigue, sleepiness, headache, loss of appetite, dry mouth, metallic taste, nausea, and vomiting. This vitamin can be considered as an adjunct to standard antidepressants, particularly in patients with treatment-resistant depression who have low vitamin D levels, but regular monitoring is necessary to avoid toxicity.

Essential fatty acids. Protein receptors embedded in lipid membranes and their binding affinities are influenced by omega-3 and omega-6 polyunsaturated fatty acids. Thus, essential fatty acids may benefit depression by maintaining membrane integrity and fluidity, as well as via their anti-inflammatory activity.

Continue to: Although results from...

 

 

Although results from controlled trials are mixed, a systematic review and meta-analysis of adjunctive nutraceuticals supported a potential role for essential fatty acids, primarily eicosapentaenoic acid (EPA), by itself or in combination with docosahexaenoic acid (DHA), with total EPA >60%.5 A second meta-analysis of 26 studies (N = 2,160) that considered only essential fatty acids concluded that EPA ≥60% at ≤1 g/d could benefit depression.6 Furthermore, omega-3 fatty acids may be helpful as an add-on agent for postpartum depression.7

Be aware that a diet rich in omega-6 greatly increases oxidized low-density lipoprotein levels in adipose tissue, potentially posing a cardiac risk factor. Clinicians need to be aware that self-prescribed use of essential fatty acids is common, and to ask about and monitor their patients’ use of these agents.

S-adenosyl-L-methionine (SAMe) is an intracellular amino acid and methyl donor. Among other actions, it is involved in the biosynthesis of hormones and neurotransmitters. There is promising but limited preliminary evidence of its efficacy and safety as a monotherapy or for antidepressant augmentation.8 For example, when compared with placebo for depressive symptoms in 19 randomized controlled trials (RCTs) (N = 878) 8:

  • Five out of 6 earlier controlled studies reported SAMe IV (200 to 400 mg/d) or IM (45 to 50 mg/d) was more effective than placebo
  • When the above studies were added to 14 subsequent studies for a meta-analysis, 12 of 19 RCTs reported that parenteral or oral SAMe was significantly more effective than placebo for depression (P < .05).

Overall, the safety and tolerability of SAMe are good. Common adverse effects include nausea, mild insomnia, dizziness, irritability, and anxiety. This is another compound widely available without a prescription and at times self-prescribed. It carries an acceptable risk/benefit balance, with decades of experience.

Hypericum perforatum (St. John’s Wort) is widely prescribed for depression in China and Europe, typically in doses ranging from 500 to 900 mg/d. Its mechanism of action in depression may relate to inhibition of serotonin, dopamine, and norepinephrine uptake from the synaptic cleft of these interconnecting neurotransmitter systems.

Continue to: A meta-analysis of 7 clinical trials...

 

 

A meta-analysis of 7 clinical trials (N = 3,808) comparing St. John’s Wort with various selective serotonin reuptake inhibitors (SSRIs) reported comparable rates of response (pooled relative risk .983, 95% CI .924 to 1.042; P < .001) and remission (pooled relative risk 1.013, 95% CI .892 to 1.134; P < .001).9 Further, there were significantly lower discontinuation/dropout rates (pooled odds ratio .587, 95% CI .478 to 0.697; P < .001) for St. John’s Wort compared with the SSRIs.

Existing evidence on the long-term efficacy and safety is limited (studies ranged from 4 to 12 weeks), as is evidence for patients with more severe depression or high suicidality.

Serious drug interactions include the potential for serotonin syndrome when St. John’s Wort is combined with certain antidepressants, compromised efficacy of benzodiazepines and standard antidepressants, and severe skin reactions to sun exposure. In addition, St. John’s Wort may not be safe to use during pregnancy or while breastfeeding. Because potential drug interactions can be serious and individuals often self-prescribe this agent, it is important to ask patients about their use of St. John’s Wort, and to be vigilant for such potential adverse interactions.

Probiotics. These agents produce neuroactive substances that act on the brain/gut axis. Preliminary evidence suggests that these “psychobiotics” confer mental health benefits.10-12 Relative to other approaches, their low-risk profile make them an attractive option for some patients.

Anti-inflammatory/immune system therapies

Inflammation is linked to various medical and brain disorders. For example, patients with depression often demonstrate increased levels of peripheral blood inflammatory biomarkers (such as C-reactive protein and interleukin-6 and -17) that are known to alter norepinephrine, neuroendocrine (eg, the hypothalamic-pituitary-adrenal axis), and microglia function in addition to neuro­plasticity. Thus, targeting inflammation may facilitate the development of novel antidepressants. In addition, these agents may benefit depression associated with comorbid autoimmune disorders, such as psoriasis or rheumatoid arthritis. A systematic review and meta-analysis of 36 RCTs (N = 10,000) found 5 out of 6 anti-inflammatory agents improved depression.13,14 In general, reported disadvantages of anti-inflammatories/immunosuppressants include the potential to block the antidepressant effect of some agents, the risk of opportunistic infections, and an increased risk of suicide.

Continue to: Statins

 

 

Statins

In a meta-analysis of 3 randomized, double-blind trials, 3 statins (lovastatin, atorvastatin, and simvastatin) significantly improved depression scores when used as an adjunctive therapy to fluoxetine and citalopram, compared with adjunctive placebo (N = 165, P < .001).15

Specific adverse effects of statins include headaches, muscle pain (rarely rhabdomyolysis), dizziness, rash, and liver damage. Statins also have the potential for adverse interactions with other medications. Given the limited efficacy literature on statins for depression and the potential for serious adverse effects, these agents probably should be limited to patients with treatment-resistant depression for whom a statin is indicated for a comorbid medical disorder, such as hypercholesteremia.

Neurosteroids

Brexanolone is FDA-approved for the treatment of postpartum depression. It is an IV formulation of the neuroactive steroid hormone allopregnanolone (a metabolite of progesterone), which acts as a positive allosteric modulator of the GABA-A receptor. Unfortunately, the infusion needs to occur over a 60-hour period.

Ganaxolone is an oral analog formulation of allopregnanolone. In an uncontrolled, open-label pilot study, this medication was administered for 8 weeks as an adjunct to an adequately dosed antidepressant to 10 postmenopausal women with persistent MDD.16 Of the 9 women who completed the study, 4 (44%) improved significantly (P < .019) and the benefit was sustained for 2 additional weeks.16 Adverse effects of ganaxolone included dizziness in 60% of participants, and sleepiness and fatigue in all of them with twice-daily dosing. If the FDA approves ganaxolone, it would become an easier-to-administer option to brexanolone.

Zuranolone is an investigational agent being studied as a treatment for postpartum depression. In a double-blind RCT that evaluated 151 women with postpartum depression, those who took oral zuranolone, 30 mg daily at bedtime for 2 weeks, experienced significant reductions in Hamilton Depression Rating Scale-17 (HDRS-17) scores compared with placebo (P < .003).17 Improvement in core depression symptom ratings was seen as early as Day 3 and persisted through Day 45.

Continue to: The most common...

 

 

The most common (≥5%) treatment-emergent adverse effects were somnolence (15%), headache (9%), dizziness (8%), upper respiratory tract infection (8%), diarrhea (6%), and sedation (5%). Two patients experienced a serious adverse event: one who received zuranolone (confusional state) and one who received placebo (pancreatitis). One patient discontinued zuranolone due to adverse effects vs no discontinuations among those who received placebo. The risk of taking zuranolone while breastfeeding is not known.

Device-based strategies

In addition to FDA-cleared approaches (eg, electroconvulsive therapy [ECT], vagus nerve stimulation [VNS], transcranial magnetic stimulation [TMS]), other devices have also demonstrated promising results.

Transcranial direct current stimulation (tDCS) involves delivering weak electrical current to the cerebral cortex through small scalp electrodes to produce the following effects:

  • anodal tDCS enhances cortical excitability
  • cathodal tDCS reduces cortical excitability.

A typical protocol consists of delivering 1 to 2 mA over 20 minutes with scalp electrodes placed in different configurations based on the targeted symptom(s).

While tDCS has been evaluated as a treatment for various neuropsychiatric disorders, including bipolar depression, Parkinson’s disease, and schizophrenia, most trials have looked at its use for treating depression. Results have been promising but mixed. For example, 1 meta-analysis of 6 RCTs (comprising 96 active and 80 sham tDCS courses) reported that active tDCS was superior to a sham procedure (Hedges’ g = 0.743) for symptoms of depression.18 By contrast, another meta-analysis of 6 RCTs (N = 200) did not find a significant difference between active and sham tDCS for response and remission rates.19 More recently, a group of experts created an evidence-based guideline using a systematic review of the controlled trial literature. These authors concluded there is “probable efficacy for anodal tDCS of the left dorsolateral prefrontal cortex (DLPFC) (with right orbitofrontal cathode) in major depressive episodes without drug resistance but probable inefficacy for drug-resistant major depressive episodes.”20

Continue to: Adverse effects of tDCS...

 

 

Adverse effects of tDCS are typically mild but may include persistent skin lesions similar to burns; mania or hypomania; and one reported seizure in a pediatric patient.

Because various over-the-counter direct current stimulation devices are available for purchase at modest cost, clinicians should ask patients if they have been self-administering this treatment.

Chronotherapy strategies

Agomelatine combines serotonergic (5-HT2B and 5-HT2C antagonist) and melatonergic (MT1-MT2 agonist in the suprachiasmatic nucleus) actions that contribute to stabilization of circadian rhythms and subsequent improvement in sleep patterns. Agomelatine (n = 1,274) significantly lowered depression symptoms compared with placebo (n = 689) (standardized mean difference −0.26; P < 3.48×10-11), but the clinical relevance was questionable.21 A recent review of the literature and expert opinion suggest this agent may also have efficacy for anhedonia; however, in placebo-controlled, relapse prevention studies, its long-term efficacy was not consistent.22

Common adverse effects include anxiety; nausea, vomiting, and stomach pain; abnormal dreams and insomnia; dizziness; drowsiness and fatigue; and weight gain. Some reviewers have expressed concerns about agomelatine’s potential for hepatotoxicity and the need for repeated clinical laboratory tests. Although agomelatine is approved outside of the United States, limited efficacy data and the potential for serious adverse effects have precluded FDA approval of this agent.

Sleep deprivation as a treatment technique for depression has been developed over the past 50 years. With total sleep deprivation (TSD) over 1 cycle, patients stay awake for approximately 36 hours, from daytime until the next day’s evening. While 1 to 6 cycles can produce acute antidepressant effects, prompt relapse after sleep recovery is common.

Continue to: In a systematic review...

 

 

In a systematic review and meta-analysis of 7 studies that included a total of 311 patients with bipolar depression23:

  • TSD plus medications resulted in a significant decrease in depressive symptoms at 1 week compared with medications alone
  • higher response rates were maintained after 3 months with lithium.

Adverse effects commonly include general fatigue and headaches; possible switch into mania with bipolar depression; and rarely, seizures or other unexpected medical conditions (eg, acute coronary syndrome). Presently, this approach is limited to research laboratories with the appropriate sophistication to safely conduct such trials.

Other nontraditional strategies

Cardiovascular exercise, resistance training, mindfulness, and yoga have been shown to decrease severe depressive symptoms when used as adjuncts for patients with treatment-resistant depression, or as monotherapy to treat patients with milder depression.

Exercise. The significant benefits of exercise in various forms as treatment for mild to moderate depression are well described in the literature, but it is less clear if it is effective for treatment-resistant depression. A 2013 Cochrane report24 (39 studies with 2,326 participants total) and 2 meta-analyses undertaken in 2015 (Kvam et al25 included 23 studies with 977 participants, and Schuh et al26 included 25 trials with 1,487 participants) reported that various types of exercise ameliorate depression of differing subtypes and severity, with effect sizes ranging from small to large. Schuh et al26 found that publication bias underestimated effect size. Also, not surprisingly, separate analysis of only higher-quality trials decreased effect size.24-26 A meta-analysis that included tai chi and yoga in addition to aerobic exercise and strength training (25 trials with 2,083 participants) found low to moderate benefit for exercise and yoga.27 Finally, a meta-analysis by Cramer et al28 that included 12 RCTs (N = 619) supported the use of yoga plus controlled breathing techniques as an ancillary treatment for depression.

Two small exercise trials specifically evaluated patients with treatment-resistant depression.29,30 Mota-Pereira et al29 compared 22 participants who walked for 30 to 45 minutes, 5 days a week for 12 weeks in addition to pharmacotherapy with 11 patients who received pharmacotherapy only. Exercise improved all outcomes, including HDRS score (both compared to baseline and to the control group). Moreover, 26% of the exercise group went into remission. Pilu et al30 evaluated strength training as an adjunctive treatment. Participants received 1 hour of strength training twice weekly for 8 months (n = 10), or pharmacotherapy only (n = 20). The adjunct strength training group had a statistically significant (P < .0001) improvement in HDRS scores at the end of the 8 months, whereas the control group did not (P < .28).

Continue to: Adverse effects...

 

 

Adverse effects of exercise are typically limited to sprains or strains; rarely, participants experience serious injuries.

Mindfulness-based interventions involve purposely paying attention in the present moment to enhance self-understanding and decrease anxiety about the future and regrets about the past, both of which complicate depression. A meta-analysis of 12 RCTs (N = 578) found this approach significantly reduced depression severity when used as an adjunctive therapy.31 There may be risks if mindfulness-based interventions are practiced incorrectly. For example, some reports have linked mindfulness-based interventions to psychotic episodes, meditation addiction, and antisocial or asocial behavior.32

 

Bottom Line

Nonpharmacologic options for patients with treatment-resistant depression include herbal/nutraceuticals, anti-inflammatory/immune system therapies, and devices. While research suggests some of these approaches are promising, clinicians need to carefully consider potential adverse effects, some of which may be serious.

Related Resources

Drug Brand Names

Atorvastatin • Lipitor
Brexanolone • Zulresso
Citalopram • Celexa
Fluoxetine • Prozac
Lithium • Eskalith, Lithobid
Lovastatin • Altoprev, Mevacor
Minocycline • Dynacin, Minocin
Simvastatin • Flolipid, Zocor

[embed:render:related:node:244798]

[embed:render:related:node:195221]

References

1. Pittampalli S, Mekala HM, Upadhyayula, S, et al. Does vitamin D deficiency cause depression? Prim Care Companion CNS Disord. 2018;20(5):17l02263.

2. Parker GB, Brotchie H, Graham RK. Vitamin D and depression. J Affect Disord. 2017;208:56-61.

3. Berridge MJ. Vitamin D and depression: cellular and regulatory mechanisms. Pharmacol Rev. 2017;69(2):80-92.

4. Anglin RE, Samaan Z, Walter SD, et al. Vitamin D deficiency and depression in adults: systematic review and meta-analysis. Br J Psychiatry. 2013;202:100-107.

5. Sarris J, Murphy J, Mischoulon D, et al. Adjunctive nutraceuticals for depression: a systematic review and meta-analyses. Am J Psychiatry 2016;173(6);575-587.

6. Liao Y, Xie B, Zhang H, et al. Efficacy of omega-3 PUFAs in depression: a meta-analysis. Transl Psychiatry. 2019;9(1):190.

7. Mocking RJT, Steijn K, Roos C, et al. Omega-3 fatty acid supplementation for perinatal depression: a meta-analysis. J Clin Psychiatry. 2020;81(5):19r13106.

8. Sharma A, Gerbarg P, Bottiglieri T, et al; Work Group of the American Psychiatric Association Council on Research. S-Adenosylmethionine (SAMe) for neuropsychiatric disorders: a clinician-oriented review of research. J Clin Psychiatry. 2017;78(6):e656-e667.

9. Ng QX, Venkatanarayanan N, Ho CY. Clinical use of hypericum perforatum (St John’s wort) in depression: a meta-analysis. J Affect Disord 2017;210:211-221.

10. Huang R, Wang K, Hu J. Effect of probiotics on depression: a systematic review and meta-analysis of randomized controlled trials. Nutrients. 2016;8(8):483.

11. Liu RT, Walsh RFL, Sheehan AE. Prebiotics and probiotics for depression and anxiety: a systematic review and meta-analysis of controlled clinical trials. Neurosci Biobehav Rev. 2019;102:13-23.

12. Wallace CJK, Milev RV. The efficacy, safety, and tolerability of probiotics on depression: clinical results from an open-label pilot study. Front Psychiatry. 2021;12(132):618279.

13. Köhler-Forsberg O, N Lyndholm C, Hjorthøj C, et al. Efficacy of anti-inflammatory treatment on major depressive disorder or depressive symptoms: meta-analysis of clinical trials. Acta Psychiatr Scand. 2019;139(5):404-419.

14. Jha MK. Anti-inflammatory treatments for major depressive disorder: what’s on the horizon? J Clin Psychiatry. 2019;80(6)18ac12630.

15. Salagre E, Fernandes BS, Dodd S, et al. Statins for the treatment of depression: a meta-analysis of randomized, double-blind, placebo-controlled trials. J Affect Disord. 2016;200:235-242.

16. Dichtel LE, Nyer M, Dording C, et al. Effects of open-label, adjunctive ganaxolone on persistent depression despite adequate antidepressant treatment in postmenopausal women: a pilot study. J Clin Psychiatry. 2020;81(4):19m12887.

17. Deligiannidis KM, Meltzer-Brody S, Gunduz-Bruce H, et al. Effect of zuranolone vs placebo in postpartum depression: a randomized clinical trial. JAMA Psychiatry. 2021;78(9):951-959.

18. Kalu UG, Sexton CE, Loo CK, et al. Transcranial direct current stimulation in the treatment of major depression: a meta-analysis. Psychol Med. 2012;42(9):1791-800.

19. Berlim MT, Van den Eynde F, Daskalakis ZJ. Clinical utility of transcranial direct current stimulation (tDCS) for treating major depression: a systematic review and meta-analysis of randomized, double-blind and sham-controlled trials. J Psychiatr Res. 2013;47(1):1-7.

20. Lefaucheur JP, Antal A, Ayache SS, et al. Evidence-based guidelines on the therapeutic use of transcranial direct current stimulation (tDCS). Clin Neurophysiol. 2017;128(1):56-92.

21. Singh SP, Singh V, Kar N. Efficacy of agomelatine in major depressive disorder: meta-analysis and appraisal. Int J Neuropsychopharmacol. 2012;15(3):417-428.

22. Norman TR, Olver JS. Agomelatine for depression: expanding the horizons? Expert Opin Pharmacother. 2019;20(6):647-656.

23. Ramirez-Mahaluf JP, Rozas-Serri E, Ivanovic-Zuvic F, et al. Effectiveness of sleep deprivation in treating acute bipolar depression as augmentation strategy: a systematic review and meta-analysis. Front Psychiatry. 2020;11:70.

24. Cooney GM, Dwan K, Greig CA, et al. Exercise for depression. Cochrane Database Syst Rev. 2013;(9):CD004366.

25. Kvam S, Kleppe CL, Nordhus IH, et al. Exercise as a treatment for depression: a meta-analysis. J Affect Disord. 2016;202:67-86.

26. Schuch FB, Vancampfort D, Richards J, et al. Exercise as a treatment for depression: a meta-analysis adjusting for publication bias. J Psychiatr Res. 2016;77:42-51.

27. Seshadri A, Adaji A, Orth SS, et al. Exercise, yoga, and tai chi for treatment of major depressive disorder in outpatient settings: a systematic review and meta-analysis. Prim Care Companion CNS Disord. 2020;23(1):20r02722.

28. Cramer H, Lauche R, Langhorst J, et al. Yoga for depression: a systematic review and meta-analysis. Depress Anxiety. 2013;30(11):1068-1083.

29. Mota-Pereira J, Silverio J, Carvalho S, et al. Moderate exercise improves depression parameters in treatment-resistant patients with major depressive disorder. J Psychiatr Res. 2011;45(8):1005-1011.

30. Pilu A, Sorba M, Hardoy MC, et al. Efficacy of physical activity in the adjunctive treatment of major depressive disorders: preliminary results. Clin Pract Epidemiol Ment Health. 2007;3:8.

31. Strauss C, Cavanagh K, Oliver A, et al. Mindfulness-based interventions for people diagnosed with a current episode of an anxiety or depressive disorder: a meta-analysis of randomised controlled trials. PLoS One. 2014;9(4):e96110.

32. Shonin E, Van Gordon W, Griffiths MD. Are there risks associated with using mindfulness for the treatment of psychopathology? Clinical Practice. 2014;11(4):389-392.

Article PDF
Author and Disclosure Information

Mehmet E. Dokucu, MD, PhD
Associate Professor
Department of Psychiatry and Behavioral Sciences
Northwestern University Feinberg School of Medicine
Chicago, Illinois

Philip G. Janicak, MD
Adjunct Professor
Department of Psychiatry and Behavioral Sciences
Northwestern University Feinberg School of Medicine
Chicago, Illinois

Disclosures
Dr. Dokucu reports no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products. Dr. Janicak is a speaker for Otsuka PsychU program and TMS Health Solutions.

Issue
Current Psychiatry - 20(10)
Publications
Topics
Page Number
32-38
Sections
Author and Disclosure Information

Mehmet E. Dokucu, MD, PhD
Associate Professor
Department of Psychiatry and Behavioral Sciences
Northwestern University Feinberg School of Medicine
Chicago, Illinois

Philip G. Janicak, MD
Adjunct Professor
Department of Psychiatry and Behavioral Sciences
Northwestern University Feinberg School of Medicine
Chicago, Illinois

Disclosures
Dr. Dokucu reports no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products. Dr. Janicak is a speaker for Otsuka PsychU program and TMS Health Solutions.

Author and Disclosure Information

Mehmet E. Dokucu, MD, PhD
Associate Professor
Department of Psychiatry and Behavioral Sciences
Northwestern University Feinberg School of Medicine
Chicago, Illinois

Philip G. Janicak, MD
Adjunct Professor
Department of Psychiatry and Behavioral Sciences
Northwestern University Feinberg School of Medicine
Chicago, Illinois

Disclosures
Dr. Dokucu reports no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products. Dr. Janicak is a speaker for Otsuka PsychU program and TMS Health Solutions.

Article PDF
Article PDF

When patients with major depressive disorder (MDD) do not achieve optimal outcomes after FDA-approved first-line treatments and standard adjunctive strategies, clinicians look for additional approaches to alleviate their patients’ symptoms. Recent research suggests that several “nontraditional” treatments used primarily as adjuncts to standard antidepressants have promise for treatment-resistant depression.

In Part 1 of this article (Current Psychiatry, September 2021), we examined off-label medications. In Part 2, we will review other nontraditional approaches to treatment-resistant depression, including herbal/nutraceutical agents, anti-inflammatory/immune system therapies, device-based treatments, and other alternative approaches. Importantly, some treatments also demonstrate adverse effects (Table1-32). With a careful consideration of the risk/benefit balance, this article reviews some of the better-studied nontraditional treatment options for patients with treatment-resistant depression.

CP02010032_t1.PNG

Herbal/nutraceutical agents

This category encompasses a variety of commonly available “natural” options patients often ask about and at times self-prescribe. Examples evaluated in clinical trials include:

  • vitamin D
  • essential fatty acids (omega-3, omega-6)
  • S-adenosyl-L-methionine (SAMe)
  • hypericum perforatum (St. John’s Wort)
  • probiotics.

Vitamin D deficiency has been linked to depression, possibly by lowering serotonin, norepinephrine, and dopamine concentrations.1-3

A meta-analysis of 3 prospective, observational studies (N = 8,815) found an elevated risk of affective disorders in patients with low vitamin D levels.4 In addition, a systematic review and meta-analysis supported a potential role for vitamin D supplementation for patients with treatment-resistant depresssion.5

Toxicity can occur at levels >100 ng/mL, and resulting adverse effects may include weakness, fatigue, sleepiness, headache, loss of appetite, dry mouth, metallic taste, nausea, and vomiting. This vitamin can be considered as an adjunct to standard antidepressants, particularly in patients with treatment-resistant depression who have low vitamin D levels, but regular monitoring is necessary to avoid toxicity.

Essential fatty acids. Protein receptors embedded in lipid membranes and their binding affinities are influenced by omega-3 and omega-6 polyunsaturated fatty acids. Thus, essential fatty acids may benefit depression by maintaining membrane integrity and fluidity, as well as via their anti-inflammatory activity.

Continue to: Although results from...

 

 

Although results from controlled trials are mixed, a systematic review and meta-analysis of adjunctive nutraceuticals supported a potential role for essential fatty acids, primarily eicosapentaenoic acid (EPA), by itself or in combination with docosahexaenoic acid (DHA), with total EPA >60%.5 A second meta-analysis of 26 studies (N = 2,160) that considered only essential fatty acids concluded that EPA ≥60% at ≤1 g/d could benefit depression.6 Furthermore, omega-3 fatty acids may be helpful as an add-on agent for postpartum depression.7

Be aware that a diet rich in omega-6 greatly increases oxidized low-density lipoprotein levels in adipose tissue, potentially posing a cardiac risk factor. Clinicians need to be aware that self-prescribed use of essential fatty acids is common, and to ask about and monitor their patients’ use of these agents.

S-adenosyl-L-methionine (SAMe) is an intracellular amino acid and methyl donor. Among other actions, it is involved in the biosynthesis of hormones and neurotransmitters. There is promising but limited preliminary evidence of its efficacy and safety as a monotherapy or for antidepressant augmentation.8 For example, when compared with placebo for depressive symptoms in 19 randomized controlled trials (RCTs) (N = 878) 8:

  • Five out of 6 earlier controlled studies reported SAMe IV (200 to 400 mg/d) or IM (45 to 50 mg/d) was more effective than placebo
  • When the above studies were added to 14 subsequent studies for a meta-analysis, 12 of 19 RCTs reported that parenteral or oral SAMe was significantly more effective than placebo for depression (P < .05).

Overall, the safety and tolerability of SAMe are good. Common adverse effects include nausea, mild insomnia, dizziness, irritability, and anxiety. This is another compound widely available without a prescription and at times self-prescribed. It carries an acceptable risk/benefit balance, with decades of experience.

Hypericum perforatum (St. John’s Wort) is widely prescribed for depression in China and Europe, typically in doses ranging from 500 to 900 mg/d. Its mechanism of action in depression may relate to inhibition of serotonin, dopamine, and norepinephrine uptake from the synaptic cleft of these interconnecting neurotransmitter systems.

Continue to: A meta-analysis of 7 clinical trials...

 

 

A meta-analysis of 7 clinical trials (N = 3,808) comparing St. John’s Wort with various selective serotonin reuptake inhibitors (SSRIs) reported comparable rates of response (pooled relative risk .983, 95% CI .924 to 1.042; P < .001) and remission (pooled relative risk 1.013, 95% CI .892 to 1.134; P < .001).9 Further, there were significantly lower discontinuation/dropout rates (pooled odds ratio .587, 95% CI .478 to 0.697; P < .001) for St. John’s Wort compared with the SSRIs.

Existing evidence on the long-term efficacy and safety is limited (studies ranged from 4 to 12 weeks), as is evidence for patients with more severe depression or high suicidality.

Serious drug interactions include the potential for serotonin syndrome when St. John’s Wort is combined with certain antidepressants, compromised efficacy of benzodiazepines and standard antidepressants, and severe skin reactions to sun exposure. In addition, St. John’s Wort may not be safe to use during pregnancy or while breastfeeding. Because potential drug interactions can be serious and individuals often self-prescribe this agent, it is important to ask patients about their use of St. John’s Wort, and to be vigilant for such potential adverse interactions.

Probiotics. These agents produce neuroactive substances that act on the brain/gut axis. Preliminary evidence suggests that these “psychobiotics” confer mental health benefits.10-12 Relative to other approaches, their low-risk profile make them an attractive option for some patients.

Anti-inflammatory/immune system therapies

Inflammation is linked to various medical and brain disorders. For example, patients with depression often demonstrate increased levels of peripheral blood inflammatory biomarkers (such as C-reactive protein and interleukin-6 and -17) that are known to alter norepinephrine, neuroendocrine (eg, the hypothalamic-pituitary-adrenal axis), and microglia function in addition to neuro­plasticity. Thus, targeting inflammation may facilitate the development of novel antidepressants. In addition, these agents may benefit depression associated with comorbid autoimmune disorders, such as psoriasis or rheumatoid arthritis. A systematic review and meta-analysis of 36 RCTs (N = 10,000) found 5 out of 6 anti-inflammatory agents improved depression.13,14 In general, reported disadvantages of anti-inflammatories/immunosuppressants include the potential to block the antidepressant effect of some agents, the risk of opportunistic infections, and an increased risk of suicide.

Continue to: Statins

 

 

Statins

In a meta-analysis of 3 randomized, double-blind trials, 3 statins (lovastatin, atorvastatin, and simvastatin) significantly improved depression scores when used as an adjunctive therapy to fluoxetine and citalopram, compared with adjunctive placebo (N = 165, P < .001).15

Specific adverse effects of statins include headaches, muscle pain (rarely rhabdomyolysis), dizziness, rash, and liver damage. Statins also have the potential for adverse interactions with other medications. Given the limited efficacy literature on statins for depression and the potential for serious adverse effects, these agents probably should be limited to patients with treatment-resistant depression for whom a statin is indicated for a comorbid medical disorder, such as hypercholesteremia.

Neurosteroids

Brexanolone is FDA-approved for the treatment of postpartum depression. It is an IV formulation of the neuroactive steroid hormone allopregnanolone (a metabolite of progesterone), which acts as a positive allosteric modulator of the GABA-A receptor. Unfortunately, the infusion needs to occur over a 60-hour period.

Ganaxolone is an oral analog formulation of allopregnanolone. In an uncontrolled, open-label pilot study, this medication was administered for 8 weeks as an adjunct to an adequately dosed antidepressant to 10 postmenopausal women with persistent MDD.16 Of the 9 women who completed the study, 4 (44%) improved significantly (P < .019) and the benefit was sustained for 2 additional weeks.16 Adverse effects of ganaxolone included dizziness in 60% of participants, and sleepiness and fatigue in all of them with twice-daily dosing. If the FDA approves ganaxolone, it would become an easier-to-administer option to brexanolone.

Zuranolone is an investigational agent being studied as a treatment for postpartum depression. In a double-blind RCT that evaluated 151 women with postpartum depression, those who took oral zuranolone, 30 mg daily at bedtime for 2 weeks, experienced significant reductions in Hamilton Depression Rating Scale-17 (HDRS-17) scores compared with placebo (P < .003).17 Improvement in core depression symptom ratings was seen as early as Day 3 and persisted through Day 45.

Continue to: The most common...

 

 

The most common (≥5%) treatment-emergent adverse effects were somnolence (15%), headache (9%), dizziness (8%), upper respiratory tract infection (8%), diarrhea (6%), and sedation (5%). Two patients experienced a serious adverse event: one who received zuranolone (confusional state) and one who received placebo (pancreatitis). One patient discontinued zuranolone due to adverse effects vs no discontinuations among those who received placebo. The risk of taking zuranolone while breastfeeding is not known.

Device-based strategies

In addition to FDA-cleared approaches (eg, electroconvulsive therapy [ECT], vagus nerve stimulation [VNS], transcranial magnetic stimulation [TMS]), other devices have also demonstrated promising results.

Transcranial direct current stimulation (tDCS) involves delivering weak electrical current to the cerebral cortex through small scalp electrodes to produce the following effects:

  • anodal tDCS enhances cortical excitability
  • cathodal tDCS reduces cortical excitability.

A typical protocol consists of delivering 1 to 2 mA over 20 minutes with scalp electrodes placed in different configurations based on the targeted symptom(s).

While tDCS has been evaluated as a treatment for various neuropsychiatric disorders, including bipolar depression, Parkinson’s disease, and schizophrenia, most trials have looked at its use for treating depression. Results have been promising but mixed. For example, 1 meta-analysis of 6 RCTs (comprising 96 active and 80 sham tDCS courses) reported that active tDCS was superior to a sham procedure (Hedges’ g = 0.743) for symptoms of depression.18 By contrast, another meta-analysis of 6 RCTs (N = 200) did not find a significant difference between active and sham tDCS for response and remission rates.19 More recently, a group of experts created an evidence-based guideline using a systematic review of the controlled trial literature. These authors concluded there is “probable efficacy for anodal tDCS of the left dorsolateral prefrontal cortex (DLPFC) (with right orbitofrontal cathode) in major depressive episodes without drug resistance but probable inefficacy for drug-resistant major depressive episodes.”20

Continue to: Adverse effects of tDCS...

 

 

Adverse effects of tDCS are typically mild but may include persistent skin lesions similar to burns; mania or hypomania; and one reported seizure in a pediatric patient.

Because various over-the-counter direct current stimulation devices are available for purchase at modest cost, clinicians should ask patients if they have been self-administering this treatment.

Chronotherapy strategies

Agomelatine combines serotonergic (5-HT2B and 5-HT2C antagonist) and melatonergic (MT1-MT2 agonist in the suprachiasmatic nucleus) actions that contribute to stabilization of circadian rhythms and subsequent improvement in sleep patterns. Agomelatine (n = 1,274) significantly lowered depression symptoms compared with placebo (n = 689) (standardized mean difference −0.26; P < 3.48×10-11), but the clinical relevance was questionable.21 A recent review of the literature and expert opinion suggest this agent may also have efficacy for anhedonia; however, in placebo-controlled, relapse prevention studies, its long-term efficacy was not consistent.22

Common adverse effects include anxiety; nausea, vomiting, and stomach pain; abnormal dreams and insomnia; dizziness; drowsiness and fatigue; and weight gain. Some reviewers have expressed concerns about agomelatine’s potential for hepatotoxicity and the need for repeated clinical laboratory tests. Although agomelatine is approved outside of the United States, limited efficacy data and the potential for serious adverse effects have precluded FDA approval of this agent.

Sleep deprivation as a treatment technique for depression has been developed over the past 50 years. With total sleep deprivation (TSD) over 1 cycle, patients stay awake for approximately 36 hours, from daytime until the next day’s evening. While 1 to 6 cycles can produce acute antidepressant effects, prompt relapse after sleep recovery is common.

Continue to: In a systematic review...

 

 

In a systematic review and meta-analysis of 7 studies that included a total of 311 patients with bipolar depression23:

  • TSD plus medications resulted in a significant decrease in depressive symptoms at 1 week compared with medications alone
  • higher response rates were maintained after 3 months with lithium.

Adverse effects commonly include general fatigue and headaches; possible switch into mania with bipolar depression; and rarely, seizures or other unexpected medical conditions (eg, acute coronary syndrome). Presently, this approach is limited to research laboratories with the appropriate sophistication to safely conduct such trials.

Other nontraditional strategies

Cardiovascular exercise, resistance training, mindfulness, and yoga have been shown to decrease severe depressive symptoms when used as adjuncts for patients with treatment-resistant depression, or as monotherapy to treat patients with milder depression.

Exercise. The significant benefits of exercise in various forms as treatment for mild to moderate depression are well described in the literature, but it is less clear if it is effective for treatment-resistant depression. A 2013 Cochrane report24 (39 studies with 2,326 participants total) and 2 meta-analyses undertaken in 2015 (Kvam et al25 included 23 studies with 977 participants, and Schuh et al26 included 25 trials with 1,487 participants) reported that various types of exercise ameliorate depression of differing subtypes and severity, with effect sizes ranging from small to large. Schuh et al26 found that publication bias underestimated effect size. Also, not surprisingly, separate analysis of only higher-quality trials decreased effect size.24-26 A meta-analysis that included tai chi and yoga in addition to aerobic exercise and strength training (25 trials with 2,083 participants) found low to moderate benefit for exercise and yoga.27 Finally, a meta-analysis by Cramer et al28 that included 12 RCTs (N = 619) supported the use of yoga plus controlled breathing techniques as an ancillary treatment for depression.

Two small exercise trials specifically evaluated patients with treatment-resistant depression.29,30 Mota-Pereira et al29 compared 22 participants who walked for 30 to 45 minutes, 5 days a week for 12 weeks in addition to pharmacotherapy with 11 patients who received pharmacotherapy only. Exercise improved all outcomes, including HDRS score (both compared to baseline and to the control group). Moreover, 26% of the exercise group went into remission. Pilu et al30 evaluated strength training as an adjunctive treatment. Participants received 1 hour of strength training twice weekly for 8 months (n = 10), or pharmacotherapy only (n = 20). The adjunct strength training group had a statistically significant (P < .0001) improvement in HDRS scores at the end of the 8 months, whereas the control group did not (P < .28).

Continue to: Adverse effects...

 

 

Adverse effects of exercise are typically limited to sprains or strains; rarely, participants experience serious injuries.

Mindfulness-based interventions involve purposely paying attention in the present moment to enhance self-understanding and decrease anxiety about the future and regrets about the past, both of which complicate depression. A meta-analysis of 12 RCTs (N = 578) found this approach significantly reduced depression severity when used as an adjunctive therapy.31 There may be risks if mindfulness-based interventions are practiced incorrectly. For example, some reports have linked mindfulness-based interventions to psychotic episodes, meditation addiction, and antisocial or asocial behavior.32

 

Bottom Line

Nonpharmacologic options for patients with treatment-resistant depression include herbal/nutraceuticals, anti-inflammatory/immune system therapies, and devices. While research suggests some of these approaches are promising, clinicians need to carefully consider potential adverse effects, some of which may be serious.

Related Resources

Drug Brand Names

Atorvastatin • Lipitor
Brexanolone • Zulresso
Citalopram • Celexa
Fluoxetine • Prozac
Lithium • Eskalith, Lithobid
Lovastatin • Altoprev, Mevacor
Minocycline • Dynacin, Minocin
Simvastatin • Flolipid, Zocor

[embed:render:related:node:244798]

[embed:render:related:node:195221]

When patients with major depressive disorder (MDD) do not achieve optimal outcomes after FDA-approved first-line treatments and standard adjunctive strategies, clinicians look for additional approaches to alleviate their patients’ symptoms. Recent research suggests that several “nontraditional” treatments used primarily as adjuncts to standard antidepressants have promise for treatment-resistant depression.

In Part 1 of this article (Current Psychiatry, September 2021), we examined off-label medications. In Part 2, we will review other nontraditional approaches to treatment-resistant depression, including herbal/nutraceutical agents, anti-inflammatory/immune system therapies, device-based treatments, and other alternative approaches. Importantly, some treatments also demonstrate adverse effects (Table1-32). With a careful consideration of the risk/benefit balance, this article reviews some of the better-studied nontraditional treatment options for patients with treatment-resistant depression.

CP02010032_t1.PNG

Herbal/nutraceutical agents

This category encompasses a variety of commonly available “natural” options patients often ask about and at times self-prescribe. Examples evaluated in clinical trials include:

  • vitamin D
  • essential fatty acids (omega-3, omega-6)
  • S-adenosyl-L-methionine (SAMe)
  • hypericum perforatum (St. John’s Wort)
  • probiotics.

Vitamin D deficiency has been linked to depression, possibly by lowering serotonin, norepinephrine, and dopamine concentrations.1-3

A meta-analysis of 3 prospective, observational studies (N = 8,815) found an elevated risk of affective disorders in patients with low vitamin D levels.4 In addition, a systematic review and meta-analysis supported a potential role for vitamin D supplementation for patients with treatment-resistant depresssion.5

Toxicity can occur at levels >100 ng/mL, and resulting adverse effects may include weakness, fatigue, sleepiness, headache, loss of appetite, dry mouth, metallic taste, nausea, and vomiting. This vitamin can be considered as an adjunct to standard antidepressants, particularly in patients with treatment-resistant depression who have low vitamin D levels, but regular monitoring is necessary to avoid toxicity.

Essential fatty acids. Protein receptors embedded in lipid membranes and their binding affinities are influenced by omega-3 and omega-6 polyunsaturated fatty acids. Thus, essential fatty acids may benefit depression by maintaining membrane integrity and fluidity, as well as via their anti-inflammatory activity.

Continue to: Although results from...

 

 

Although results from controlled trials are mixed, a systematic review and meta-analysis of adjunctive nutraceuticals supported a potential role for essential fatty acids, primarily eicosapentaenoic acid (EPA), by itself or in combination with docosahexaenoic acid (DHA), with total EPA >60%.5 A second meta-analysis of 26 studies (N = 2,160) that considered only essential fatty acids concluded that EPA ≥60% at ≤1 g/d could benefit depression.6 Furthermore, omega-3 fatty acids may be helpful as an add-on agent for postpartum depression.7

Be aware that a diet rich in omega-6 greatly increases oxidized low-density lipoprotein levels in adipose tissue, potentially posing a cardiac risk factor. Clinicians need to be aware that self-prescribed use of essential fatty acids is common, and to ask about and monitor their patients’ use of these agents.

S-adenosyl-L-methionine (SAMe) is an intracellular amino acid and methyl donor. Among other actions, it is involved in the biosynthesis of hormones and neurotransmitters. There is promising but limited preliminary evidence of its efficacy and safety as a monotherapy or for antidepressant augmentation.8 For example, when compared with placebo for depressive symptoms in 19 randomized controlled trials (RCTs) (N = 878) 8:

  • Five out of 6 earlier controlled studies reported SAMe IV (200 to 400 mg/d) or IM (45 to 50 mg/d) was more effective than placebo
  • When the above studies were added to 14 subsequent studies for a meta-analysis, 12 of 19 RCTs reported that parenteral or oral SAMe was significantly more effective than placebo for depression (P < .05).

Overall, the safety and tolerability of SAMe are good. Common adverse effects include nausea, mild insomnia, dizziness, irritability, and anxiety. This is another compound widely available without a prescription and at times self-prescribed. It carries an acceptable risk/benefit balance, with decades of experience.

Hypericum perforatum (St. John’s Wort) is widely prescribed for depression in China and Europe, typically in doses ranging from 500 to 900 mg/d. Its mechanism of action in depression may relate to inhibition of serotonin, dopamine, and norepinephrine uptake from the synaptic cleft of these interconnecting neurotransmitter systems.

Continue to: A meta-analysis of 7 clinical trials...

 

 

A meta-analysis of 7 clinical trials (N = 3,808) comparing St. John’s Wort with various selective serotonin reuptake inhibitors (SSRIs) reported comparable rates of response (pooled relative risk .983, 95% CI .924 to 1.042; P < .001) and remission (pooled relative risk 1.013, 95% CI .892 to 1.134; P < .001).9 Further, there were significantly lower discontinuation/dropout rates (pooled odds ratio .587, 95% CI .478 to 0.697; P < .001) for St. John’s Wort compared with the SSRIs.

Existing evidence on the long-term efficacy and safety is limited (studies ranged from 4 to 12 weeks), as is evidence for patients with more severe depression or high suicidality.

Serious drug interactions include the potential for serotonin syndrome when St. John’s Wort is combined with certain antidepressants, compromised efficacy of benzodiazepines and standard antidepressants, and severe skin reactions to sun exposure. In addition, St. John’s Wort may not be safe to use during pregnancy or while breastfeeding. Because potential drug interactions can be serious and individuals often self-prescribe this agent, it is important to ask patients about their use of St. John’s Wort, and to be vigilant for such potential adverse interactions.

Probiotics. These agents produce neuroactive substances that act on the brain/gut axis. Preliminary evidence suggests that these “psychobiotics” confer mental health benefits.10-12 Relative to other approaches, their low-risk profile make them an attractive option for some patients.

Anti-inflammatory/immune system therapies

Inflammation is linked to various medical and brain disorders. For example, patients with depression often demonstrate increased levels of peripheral blood inflammatory biomarkers (such as C-reactive protein and interleukin-6 and -17) that are known to alter norepinephrine, neuroendocrine (eg, the hypothalamic-pituitary-adrenal axis), and microglia function in addition to neuro­plasticity. Thus, targeting inflammation may facilitate the development of novel antidepressants. In addition, these agents may benefit depression associated with comorbid autoimmune disorders, such as psoriasis or rheumatoid arthritis. A systematic review and meta-analysis of 36 RCTs (N = 10,000) found 5 out of 6 anti-inflammatory agents improved depression.13,14 In general, reported disadvantages of anti-inflammatories/immunosuppressants include the potential to block the antidepressant effect of some agents, the risk of opportunistic infections, and an increased risk of suicide.

Continue to: Statins

 

 

Statins

In a meta-analysis of 3 randomized, double-blind trials, 3 statins (lovastatin, atorvastatin, and simvastatin) significantly improved depression scores when used as an adjunctive therapy to fluoxetine and citalopram, compared with adjunctive placebo (N = 165, P < .001).15

Specific adverse effects of statins include headaches, muscle pain (rarely rhabdomyolysis), dizziness, rash, and liver damage. Statins also have the potential for adverse interactions with other medications. Given the limited efficacy literature on statins for depression and the potential for serious adverse effects, these agents probably should be limited to patients with treatment-resistant depression for whom a statin is indicated for a comorbid medical disorder, such as hypercholesteremia.

Neurosteroids

Brexanolone is FDA-approved for the treatment of postpartum depression. It is an IV formulation of the neuroactive steroid hormone allopregnanolone (a metabolite of progesterone), which acts as a positive allosteric modulator of the GABA-A receptor. Unfortunately, the infusion needs to occur over a 60-hour period.

Ganaxolone is an oral analog formulation of allopregnanolone. In an uncontrolled, open-label pilot study, this medication was administered for 8 weeks as an adjunct to an adequately dosed antidepressant to 10 postmenopausal women with persistent MDD.16 Of the 9 women who completed the study, 4 (44%) improved significantly (P < .019) and the benefit was sustained for 2 additional weeks.16 Adverse effects of ganaxolone included dizziness in 60% of participants, and sleepiness and fatigue in all of them with twice-daily dosing. If the FDA approves ganaxolone, it would become an easier-to-administer option to brexanolone.

Zuranolone is an investigational agent being studied as a treatment for postpartum depression. In a double-blind RCT that evaluated 151 women with postpartum depression, those who took oral zuranolone, 30 mg daily at bedtime for 2 weeks, experienced significant reductions in Hamilton Depression Rating Scale-17 (HDRS-17) scores compared with placebo (P < .003).17 Improvement in core depression symptom ratings was seen as early as Day 3 and persisted through Day 45.

Continue to: The most common...

 

 

The most common (≥5%) treatment-emergent adverse effects were somnolence (15%), headache (9%), dizziness (8%), upper respiratory tract infection (8%), diarrhea (6%), and sedation (5%). Two patients experienced a serious adverse event: one who received zuranolone (confusional state) and one who received placebo (pancreatitis). One patient discontinued zuranolone due to adverse effects vs no discontinuations among those who received placebo. The risk of taking zuranolone while breastfeeding is not known.

Device-based strategies

In addition to FDA-cleared approaches (eg, electroconvulsive therapy [ECT], vagus nerve stimulation [VNS], transcranial magnetic stimulation [TMS]), other devices have also demonstrated promising results.

Transcranial direct current stimulation (tDCS) involves delivering weak electrical current to the cerebral cortex through small scalp electrodes to produce the following effects:

  • anodal tDCS enhances cortical excitability
  • cathodal tDCS reduces cortical excitability.

A typical protocol consists of delivering 1 to 2 mA over 20 minutes with scalp electrodes placed in different configurations based on the targeted symptom(s).

While tDCS has been evaluated as a treatment for various neuropsychiatric disorders, including bipolar depression, Parkinson’s disease, and schizophrenia, most trials have looked at its use for treating depression. Results have been promising but mixed. For example, 1 meta-analysis of 6 RCTs (comprising 96 active and 80 sham tDCS courses) reported that active tDCS was superior to a sham procedure (Hedges’ g = 0.743) for symptoms of depression.18 By contrast, another meta-analysis of 6 RCTs (N = 200) did not find a significant difference between active and sham tDCS for response and remission rates.19 More recently, a group of experts created an evidence-based guideline using a systematic review of the controlled trial literature. These authors concluded there is “probable efficacy for anodal tDCS of the left dorsolateral prefrontal cortex (DLPFC) (with right orbitofrontal cathode) in major depressive episodes without drug resistance but probable inefficacy for drug-resistant major depressive episodes.”20

Continue to: Adverse effects of tDCS...

 

 

Adverse effects of tDCS are typically mild but may include persistent skin lesions similar to burns; mania or hypomania; and one reported seizure in a pediatric patient.

Because various over-the-counter direct current stimulation devices are available for purchase at modest cost, clinicians should ask patients if they have been self-administering this treatment.

Chronotherapy strategies

Agomelatine combines serotonergic (5-HT2B and 5-HT2C antagonist) and melatonergic (MT1-MT2 agonist in the suprachiasmatic nucleus) actions that contribute to stabilization of circadian rhythms and subsequent improvement in sleep patterns. Agomelatine (n = 1,274) significantly lowered depression symptoms compared with placebo (n = 689) (standardized mean difference −0.26; P < 3.48×10-11), but the clinical relevance was questionable.21 A recent review of the literature and expert opinion suggest this agent may also have efficacy for anhedonia; however, in placebo-controlled, relapse prevention studies, its long-term efficacy was not consistent.22

Common adverse effects include anxiety; nausea, vomiting, and stomach pain; abnormal dreams and insomnia; dizziness; drowsiness and fatigue; and weight gain. Some reviewers have expressed concerns about agomelatine’s potential for hepatotoxicity and the need for repeated clinical laboratory tests. Although agomelatine is approved outside of the United States, limited efficacy data and the potential for serious adverse effects have precluded FDA approval of this agent.

Sleep deprivation as a treatment technique for depression has been developed over the past 50 years. With total sleep deprivation (TSD) over 1 cycle, patients stay awake for approximately 36 hours, from daytime until the next day’s evening. While 1 to 6 cycles can produce acute antidepressant effects, prompt relapse after sleep recovery is common.

Continue to: In a systematic review...

 

 

In a systematic review and meta-analysis of 7 studies that included a total of 311 patients with bipolar depression23:

  • TSD plus medications resulted in a significant decrease in depressive symptoms at 1 week compared with medications alone
  • higher response rates were maintained after 3 months with lithium.

Adverse effects commonly include general fatigue and headaches; possible switch into mania with bipolar depression; and rarely, seizures or other unexpected medical conditions (eg, acute coronary syndrome). Presently, this approach is limited to research laboratories with the appropriate sophistication to safely conduct such trials.

Other nontraditional strategies

Cardiovascular exercise, resistance training, mindfulness, and yoga have been shown to decrease severe depressive symptoms when used as adjuncts for patients with treatment-resistant depression, or as monotherapy to treat patients with milder depression.

Exercise. The significant benefits of exercise in various forms as treatment for mild to moderate depression are well described in the literature, but it is less clear if it is effective for treatment-resistant depression. A 2013 Cochrane report24 (39 studies with 2,326 participants total) and 2 meta-analyses undertaken in 2015 (Kvam et al25 included 23 studies with 977 participants, and Schuh et al26 included 25 trials with 1,487 participants) reported that various types of exercise ameliorate depression of differing subtypes and severity, with effect sizes ranging from small to large. Schuh et al26 found that publication bias underestimated effect size. Also, not surprisingly, separate analysis of only higher-quality trials decreased effect size.24-26 A meta-analysis that included tai chi and yoga in addition to aerobic exercise and strength training (25 trials with 2,083 participants) found low to moderate benefit for exercise and yoga.27 Finally, a meta-analysis by Cramer et al28 that included 12 RCTs (N = 619) supported the use of yoga plus controlled breathing techniques as an ancillary treatment for depression.

Two small exercise trials specifically evaluated patients with treatment-resistant depression.29,30 Mota-Pereira et al29 compared 22 participants who walked for 30 to 45 minutes, 5 days a week for 12 weeks in addition to pharmacotherapy with 11 patients who received pharmacotherapy only. Exercise improved all outcomes, including HDRS score (both compared to baseline and to the control group). Moreover, 26% of the exercise group went into remission. Pilu et al30 evaluated strength training as an adjunctive treatment. Participants received 1 hour of strength training twice weekly for 8 months (n = 10), or pharmacotherapy only (n = 20). The adjunct strength training group had a statistically significant (P < .0001) improvement in HDRS scores at the end of the 8 months, whereas the control group did not (P < .28).

Continue to: Adverse effects...

 

 

Adverse effects of exercise are typically limited to sprains or strains; rarely, participants experience serious injuries.

Mindfulness-based interventions involve purposely paying attention in the present moment to enhance self-understanding and decrease anxiety about the future and regrets about the past, both of which complicate depression. A meta-analysis of 12 RCTs (N = 578) found this approach significantly reduced depression severity when used as an adjunctive therapy.31 There may be risks if mindfulness-based interventions are practiced incorrectly. For example, some reports have linked mindfulness-based interventions to psychotic episodes, meditation addiction, and antisocial or asocial behavior.32

 

Bottom Line

Nonpharmacologic options for patients with treatment-resistant depression include herbal/nutraceuticals, anti-inflammatory/immune system therapies, and devices. While research suggests some of these approaches are promising, clinicians need to carefully consider potential adverse effects, some of which may be serious.

Related Resources

Drug Brand Names

Atorvastatin • Lipitor
Brexanolone • Zulresso
Citalopram • Celexa
Fluoxetine • Prozac
Lithium • Eskalith, Lithobid
Lovastatin • Altoprev, Mevacor
Minocycline • Dynacin, Minocin
Simvastatin • Flolipid, Zocor

[embed:render:related:node:244798]

[embed:render:related:node:195221]

References

1. Pittampalli S, Mekala HM, Upadhyayula, S, et al. Does vitamin D deficiency cause depression? Prim Care Companion CNS Disord. 2018;20(5):17l02263.

2. Parker GB, Brotchie H, Graham RK. Vitamin D and depression. J Affect Disord. 2017;208:56-61.

3. Berridge MJ. Vitamin D and depression: cellular and regulatory mechanisms. Pharmacol Rev. 2017;69(2):80-92.

4. Anglin RE, Samaan Z, Walter SD, et al. Vitamin D deficiency and depression in adults: systematic review and meta-analysis. Br J Psychiatry. 2013;202:100-107.

5. Sarris J, Murphy J, Mischoulon D, et al. Adjunctive nutraceuticals for depression: a systematic review and meta-analyses. Am J Psychiatry 2016;173(6);575-587.

6. Liao Y, Xie B, Zhang H, et al. Efficacy of omega-3 PUFAs in depression: a meta-analysis. Transl Psychiatry. 2019;9(1):190.

7. Mocking RJT, Steijn K, Roos C, et al. Omega-3 fatty acid supplementation for perinatal depression: a meta-analysis. J Clin Psychiatry. 2020;81(5):19r13106.

8. Sharma A, Gerbarg P, Bottiglieri T, et al; Work Group of the American Psychiatric Association Council on Research. S-Adenosylmethionine (SAMe) for neuropsychiatric disorders: a clinician-oriented review of research. J Clin Psychiatry. 2017;78(6):e656-e667.

9. Ng QX, Venkatanarayanan N, Ho CY. Clinical use of hypericum perforatum (St John’s wort) in depression: a meta-analysis. J Affect Disord 2017;210:211-221.

10. Huang R, Wang K, Hu J. Effect of probiotics on depression: a systematic review and meta-analysis of randomized controlled trials. Nutrients. 2016;8(8):483.

11. Liu RT, Walsh RFL, Sheehan AE. Prebiotics and probiotics for depression and anxiety: a systematic review and meta-analysis of controlled clinical trials. Neurosci Biobehav Rev. 2019;102:13-23.

12. Wallace CJK, Milev RV. The efficacy, safety, and tolerability of probiotics on depression: clinical results from an open-label pilot study. Front Psychiatry. 2021;12(132):618279.

13. Köhler-Forsberg O, N Lyndholm C, Hjorthøj C, et al. Efficacy of anti-inflammatory treatment on major depressive disorder or depressive symptoms: meta-analysis of clinical trials. Acta Psychiatr Scand. 2019;139(5):404-419.

14. Jha MK. Anti-inflammatory treatments for major depressive disorder: what’s on the horizon? J Clin Psychiatry. 2019;80(6)18ac12630.

15. Salagre E, Fernandes BS, Dodd S, et al. Statins for the treatment of depression: a meta-analysis of randomized, double-blind, placebo-controlled trials. J Affect Disord. 2016;200:235-242.

16. Dichtel LE, Nyer M, Dording C, et al. Effects of open-label, adjunctive ganaxolone on persistent depression despite adequate antidepressant treatment in postmenopausal women: a pilot study. J Clin Psychiatry. 2020;81(4):19m12887.

17. Deligiannidis KM, Meltzer-Brody S, Gunduz-Bruce H, et al. Effect of zuranolone vs placebo in postpartum depression: a randomized clinical trial. JAMA Psychiatry. 2021;78(9):951-959.

18. Kalu UG, Sexton CE, Loo CK, et al. Transcranial direct current stimulation in the treatment of major depression: a meta-analysis. Psychol Med. 2012;42(9):1791-800.

19. Berlim MT, Van den Eynde F, Daskalakis ZJ. Clinical utility of transcranial direct current stimulation (tDCS) for treating major depression: a systematic review and meta-analysis of randomized, double-blind and sham-controlled trials. J Psychiatr Res. 2013;47(1):1-7.

20. Lefaucheur JP, Antal A, Ayache SS, et al. Evidence-based guidelines on the therapeutic use of transcranial direct current stimulation (tDCS). Clin Neurophysiol. 2017;128(1):56-92.

21. Singh SP, Singh V, Kar N. Efficacy of agomelatine in major depressive disorder: meta-analysis and appraisal. Int J Neuropsychopharmacol. 2012;15(3):417-428.

22. Norman TR, Olver JS. Agomelatine for depression: expanding the horizons? Expert Opin Pharmacother. 2019;20(6):647-656.

23. Ramirez-Mahaluf JP, Rozas-Serri E, Ivanovic-Zuvic F, et al. Effectiveness of sleep deprivation in treating acute bipolar depression as augmentation strategy: a systematic review and meta-analysis. Front Psychiatry. 2020;11:70.

24. Cooney GM, Dwan K, Greig CA, et al. Exercise for depression. Cochrane Database Syst Rev. 2013;(9):CD004366.

25. Kvam S, Kleppe CL, Nordhus IH, et al. Exercise as a treatment for depression: a meta-analysis. J Affect Disord. 2016;202:67-86.

26. Schuch FB, Vancampfort D, Richards J, et al. Exercise as a treatment for depression: a meta-analysis adjusting for publication bias. J Psychiatr Res. 2016;77:42-51.

27. Seshadri A, Adaji A, Orth SS, et al. Exercise, yoga, and tai chi for treatment of major depressive disorder in outpatient settings: a systematic review and meta-analysis. Prim Care Companion CNS Disord. 2020;23(1):20r02722.

28. Cramer H, Lauche R, Langhorst J, et al. Yoga for depression: a systematic review and meta-analysis. Depress Anxiety. 2013;30(11):1068-1083.

29. Mota-Pereira J, Silverio J, Carvalho S, et al. Moderate exercise improves depression parameters in treatment-resistant patients with major depressive disorder. J Psychiatr Res. 2011;45(8):1005-1011.

30. Pilu A, Sorba M, Hardoy MC, et al. Efficacy of physical activity in the adjunctive treatment of major depressive disorders: preliminary results. Clin Pract Epidemiol Ment Health. 2007;3:8.

31. Strauss C, Cavanagh K, Oliver A, et al. Mindfulness-based interventions for people diagnosed with a current episode of an anxiety or depressive disorder: a meta-analysis of randomised controlled trials. PLoS One. 2014;9(4):e96110.

32. Shonin E, Van Gordon W, Griffiths MD. Are there risks associated with using mindfulness for the treatment of psychopathology? Clinical Practice. 2014;11(4):389-392.

References

1. Pittampalli S, Mekala HM, Upadhyayula, S, et al. Does vitamin D deficiency cause depression? Prim Care Companion CNS Disord. 2018;20(5):17l02263.

2. Parker GB, Brotchie H, Graham RK. Vitamin D and depression. J Affect Disord. 2017;208:56-61.

3. Berridge MJ. Vitamin D and depression: cellular and regulatory mechanisms. Pharmacol Rev. 2017;69(2):80-92.

4. Anglin RE, Samaan Z, Walter SD, et al. Vitamin D deficiency and depression in adults: systematic review and meta-analysis. Br J Psychiatry. 2013;202:100-107.

5. Sarris J, Murphy J, Mischoulon D, et al. Adjunctive nutraceuticals for depression: a systematic review and meta-analyses. Am J Psychiatry 2016;173(6);575-587.

6. Liao Y, Xie B, Zhang H, et al. Efficacy of omega-3 PUFAs in depression: a meta-analysis. Transl Psychiatry. 2019;9(1):190.

7. Mocking RJT, Steijn K, Roos C, et al. Omega-3 fatty acid supplementation for perinatal depression: a meta-analysis. J Clin Psychiatry. 2020;81(5):19r13106.

8. Sharma A, Gerbarg P, Bottiglieri T, et al; Work Group of the American Psychiatric Association Council on Research. S-Adenosylmethionine (SAMe) for neuropsychiatric disorders: a clinician-oriented review of research. J Clin Psychiatry. 2017;78(6):e656-e667.

9. Ng QX, Venkatanarayanan N, Ho CY. Clinical use of hypericum perforatum (St John’s wort) in depression: a meta-analysis. J Affect Disord 2017;210:211-221.

10. Huang R, Wang K, Hu J. Effect of probiotics on depression: a systematic review and meta-analysis of randomized controlled trials. Nutrients. 2016;8(8):483.

11. Liu RT, Walsh RFL, Sheehan AE. Prebiotics and probiotics for depression and anxiety: a systematic review and meta-analysis of controlled clinical trials. Neurosci Biobehav Rev. 2019;102:13-23.

12. Wallace CJK, Milev RV. The efficacy, safety, and tolerability of probiotics on depression: clinical results from an open-label pilot study. Front Psychiatry. 2021;12(132):618279.

13. Köhler-Forsberg O, N Lyndholm C, Hjorthøj C, et al. Efficacy of anti-inflammatory treatment on major depressive disorder or depressive symptoms: meta-analysis of clinical trials. Acta Psychiatr Scand. 2019;139(5):404-419.

14. Jha MK. Anti-inflammatory treatments for major depressive disorder: what’s on the horizon? J Clin Psychiatry. 2019;80(6)18ac12630.

15. Salagre E, Fernandes BS, Dodd S, et al. Statins for the treatment of depression: a meta-analysis of randomized, double-blind, placebo-controlled trials. J Affect Disord. 2016;200:235-242.

16. Dichtel LE, Nyer M, Dording C, et al. Effects of open-label, adjunctive ganaxolone on persistent depression despite adequate antidepressant treatment in postmenopausal women: a pilot study. J Clin Psychiatry. 2020;81(4):19m12887.

17. Deligiannidis KM, Meltzer-Brody S, Gunduz-Bruce H, et al. Effect of zuranolone vs placebo in postpartum depression: a randomized clinical trial. JAMA Psychiatry. 2021;78(9):951-959.

18. Kalu UG, Sexton CE, Loo CK, et al. Transcranial direct current stimulation in the treatment of major depression: a meta-analysis. Psychol Med. 2012;42(9):1791-800.

19. Berlim MT, Van den Eynde F, Daskalakis ZJ. Clinical utility of transcranial direct current stimulation (tDCS) for treating major depression: a systematic review and meta-analysis of randomized, double-blind and sham-controlled trials. J Psychiatr Res. 2013;47(1):1-7.

20. Lefaucheur JP, Antal A, Ayache SS, et al. Evidence-based guidelines on the therapeutic use of transcranial direct current stimulation (tDCS). Clin Neurophysiol. 2017;128(1):56-92.

21. Singh SP, Singh V, Kar N. Efficacy of agomelatine in major depressive disorder: meta-analysis and appraisal. Int J Neuropsychopharmacol. 2012;15(3):417-428.

22. Norman TR, Olver JS. Agomelatine for depression: expanding the horizons? Expert Opin Pharmacother. 2019;20(6):647-656.

23. Ramirez-Mahaluf JP, Rozas-Serri E, Ivanovic-Zuvic F, et al. Effectiveness of sleep deprivation in treating acute bipolar depression as augmentation strategy: a systematic review and meta-analysis. Front Psychiatry. 2020;11:70.

24. Cooney GM, Dwan K, Greig CA, et al. Exercise for depression. Cochrane Database Syst Rev. 2013;(9):CD004366.

25. Kvam S, Kleppe CL, Nordhus IH, et al. Exercise as a treatment for depression: a meta-analysis. J Affect Disord. 2016;202:67-86.

26. Schuch FB, Vancampfort D, Richards J, et al. Exercise as a treatment for depression: a meta-analysis adjusting for publication bias. J Psychiatr Res. 2016;77:42-51.

27. Seshadri A, Adaji A, Orth SS, et al. Exercise, yoga, and tai chi for treatment of major depressive disorder in outpatient settings: a systematic review and meta-analysis. Prim Care Companion CNS Disord. 2020;23(1):20r02722.

28. Cramer H, Lauche R, Langhorst J, et al. Yoga for depression: a systematic review and meta-analysis. Depress Anxiety. 2013;30(11):1068-1083.

29. Mota-Pereira J, Silverio J, Carvalho S, et al. Moderate exercise improves depression parameters in treatment-resistant patients with major depressive disorder. J Psychiatr Res. 2011;45(8):1005-1011.

30. Pilu A, Sorba M, Hardoy MC, et al. Efficacy of physical activity in the adjunctive treatment of major depressive disorders: preliminary results. Clin Pract Epidemiol Ment Health. 2007;3:8.

31. Strauss C, Cavanagh K, Oliver A, et al. Mindfulness-based interventions for people diagnosed with a current episode of an anxiety or depressive disorder: a meta-analysis of randomised controlled trials. PLoS One. 2014;9(4):e96110.

32. Shonin E, Van Gordon W, Griffiths MD. Are there risks associated with using mindfulness for the treatment of psychopathology? Clinical Practice. 2014;11(4):389-392.

Issue
Current Psychiatry - 20(10)
Issue
Current Psychiatry - 20(10)
Page Number
32-38
Page Number
32-38
Publications
Publications
Topics
Article Type
Display Headline
Nontraditional therapies for treatment-resistant depression: Part 2
Display Headline
Nontraditional therapies for treatment-resistant depression: Part 2
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
18001E4E.SIG
Disable zoom
Off

Nontraditional therapies for treatment-resistant depression

Article Type
Changed
Thu, 09/02/2021 - 07:57
Display Headline
Nontraditional therapies for treatment-resistant depression

Presently, FDA-approved first-line treatments and standard adjunctive strategies (eg, lithium, thyroid supplementation, stimulants, second-generation antipsychotics) for major depressive disorder (MDD) often produce less-than-desired outcomes while carrying a potentially substantial safety and tolerability burden. The lack of clinically useful and individual-based biomarkers (eg, genetic, neurophysiological, imaging) is a major obstacle to enhancing treatment efficacy and/or decreasing associated adverse effects (AEs). While the discovery of such tools is being aggressively pursued and ultimately will facilitate a more precision-based choice of therapy, empirical strategies remain our primary approach.

In controlled trials, several nontraditional treatments used primarily as adjuncts to standard antidepressants have shown promise. These include “repurposed” (off-label) medications, herbal/nutraceuticals, anti-inflammatory/immune system therapies, device-based treatments, and other alternative approaches.

Importantly, some nontraditional treatments also demonstrate AEs (Table1-16). With a careful consideration of the risk/benefit balance, this article reviews some of the better-studied treatment options for patients with treatment-resistant depression (TRD). In Part 1, we will examine off-label medications. In Part 2, we will review other nontraditional approaches to TRD, including herbal/nutraceuticals, anti-inflammatory/immune system therapies, device-based treatments, and other alternative approaches.

CP02009038_t1.PNG

We believe this review will help clinicians who need to formulate a different approach after their patient with depression is not helped by traditional first-, second-, and third-line treatments. The potential options discussed in Part 1 of this article are categorized based on their putative mechanism of action (MOA) for depression.

Serotonergic and noradrenergic strategies

Pimavanserin is FDA-approved for treatment of Parkinson’s psychosis. Its potential MOA as an adjunctive strategy for MDD may involve 5-HT2A antagonist and inverse agonist receptor activity, as well as lesser effects at the 5-HT2Creceptor.

A 2-stage, 5-week randomized controlled trial (RCT) (CLARITY; N = 207) found adjunctive pimavanserin (34 mg/d) produced a robust antidepressant effect vs placebo in patients whose depression did not respond to selective serotonin reuptake inhibitors (SSRIs) or serotonin-norepinephrine reuptake inhibitors (SNRIs).1 Furthermore, a secondary analysis of the data suggested that pimavanserin also improved sleepiness (P < .0003) and daily functioning (P < .014) at Week 5.2

Unfortunately, two 6-week, Phase III RCTs (CLARITY-2 and -3; N = 298) did not find a statistically significant difference between active treatment and placebo. This was based on change in the primary outcome measure (Hamilton Depression Rating Scale-17 score) when adjunctive pimavanserin (34 mg/d) was added to an SSRI or SNRI in patients with TRD.3 There was, however, a significant difference favoring active treatment over placebo based on the Clinical Global Impression–Severity score.

Continue to: In these trials...

 

 

In these trials, pimavanserin was generally well-tolerated. The most common AEs were dry mouth, nausea, and headache. Pimavanserin has minimal activity at norepinephrine, dopamine, histamine, or acetylcholine receptors, thus avoiding AEs associated with these receptor interactions.

Given the mixed efficacy results of existing trials, further studies are needed to clarify this agent’s overall risk/benefit in the context of TRD.

Antihypertensive medications

Emerging data suggest that some beta-adrenergic blockers, angiotensin-inhibiting agents, and calcium antagonists are associated with a decreased incidence of depression. A large 2020 study (N = 3,747,190) used population-based Danish registries (2005 to 2015) to evaluate if any of the 41 most commonly prescribed antihypertensive medications were associated with the diagnosis of depressive disorder or use of antidepressants.4 These researchers found that enalapril, ramipril, amlodipine, propranolol, atenolol, bisoprolol, carvedilol (P < .001), and verapamil (P < .004) were strongly associated with a decreased risk of depression.4

Adverse effects across these different classes of antihypertensives are well characterized, can be substantial, and commonly are related to their impact on cardiovascular function (eg, hypotension). Clinically, these agents may be potential adjuncts for patients with TRD who need antihypertensive therapy. Their use and the choice of specific agent should only be determined in consultation with the patient’s primary care physician (PCP) or appropriate specialist.

Glutamatergic strategies

Ketamine is a dissociative anesthetic and analgesic. Its MOA for treating depression appears to occur primarily through antagonist activity at the N-methyl-D-aspartate ionotropic receptor of the glutamatergic system. There is preliminary evidence that its opioid receptor actions also may contribute to its antidepressant effect.5

Continue to: Many published studies...

 

 

Many published studies and reviews have described ketamine’s role for treating MDD. Several studies have reported that low-dose (0.5 mg/kg) IV ketamine infusions can rapidly attenuate severe episodes of MDD as well as associated suicidality. For example, a meta-analysis of 9 RCTs (N = 368) comparing ketamine to placebo for acute treatment of unipolar and bipolar depression reported superior therapeutic effects with active treatment at 24 hours, 72 hours, and 7 days.6 The response and remission rates for ketamine were 52% and 21% at 24 hours; 48% and 24% at 72 hours; and 40% and 26% at 7 days, respectively.6

The most commonly reported AEs during the 4 hours after ketamine infusion included7:

  • drowsiness, dizziness, poor coordination
  • blurred vision, feeling strange or unreal
  • hemodynamic changes (approximately 33%)
  • small but significant (P < .05) increases in psychotomimetic and dissociative symptoms.

Because some individuals use ketamine recreationally, this agent also carries the risk of abuse.

Research is ongoing on strategies for long-term maintenance ketamine treatment, and the results of both short- and long-term trials will require careful scrutiny to better assess this agent’s safety and tolerability. Clinicians should first consider esketamine—the S-enantiomer of ketamine—because an intranasal formulation of this agent is FDA-approved for treating patients with TRD or MDD with suicidality when administered in a Risk Evaluation and Mitigation Strategy–certified setting.

Cholinergic strategies

Scopolamine is a potent muscarinic receptor antagonist used to prevent nausea and vomiting caused by motion sickness or medications used during surgery. Its use for MDD is based on the theory that muscarinic receptors may be hypersensitive in mood disorders.

Continue to: Several double-blind RCTs...

 

 

Several double-blind RCTs of patients with unipolar or bipolar depression that used 3 pulsed IV infusions (4.0 mcg/kg) over 15 minutes found a rapid, robust antidepressant effect with scopolamine vs placebo.8,9 The oral formulation might also be effective, but would not have a rapid onset.

Common adverse effects of scopolamine include agitation, dry mouth, urinary retention, and cognitive clouding. Given scopolamine’s substantial AE profile, it should be considered only for patients with TRD who could also benefit from the oral formulation for the medical indications noted above, should generally be avoided in older patients, and should be prescribed in consultation with the patient’s PCP.

Botulinum toxin. This neurotoxin inhibits acetylcholine release. It is used to treat disorders characterized by abnormal muscular contraction, such as strabismus, blepharospasm, and chronic pain syndromes. Its MOA for depression may involve its paralytic effects after injection into the glabella forehead muscle (based on the facial feedback hypothesis), as well as modulation of neurotransmitters implicated in the pathophysiology of depression.

In several small trials, injectable botulinum toxin type A (BTA) (29 units) demonstrated antidepressant effects. A recent review that considered 6 trials (N = 235; 4 of the 6 studies were RCTs, 3 of which were rated as high quality) concluded that BTA may be a promising treatment for MDD.10 Limitations of this review included lack of a priori hypotheses, small sample sizes, gender bias, and difficulty in blinding.

In clinical trials, the most common AEs included local irritation at the injection site and transient headache. This agent’s relatively mild AE profile and possible overlap when used for some of the medical indications noted above opens its potential use as an adjunct in patients with comorbid TRD.

Continue to: Endocrine strategies

 

 

Endocrine strategies

Mifepristone (RU486). This anti-glucocorticoid receptor antagonist is used as an abortifacient. Based on the theory that hyperactivity of the hypothalamic-pituitary-adrenal axis is implicated in the pathophysiology of MDD with psychotic features (psychotic depression), this agent has been studied as a treatment for this indication.

An analysis of 5 double-blind RCTs (N = 1,460) found that 7 days of mifepristone, 1,200 mg/d, was superior to placebo (P < .004) in reducing psychotic symptoms of depression.11 Plasma concentrations ≥1,600 ng/mL may be required to maximize benefit.11

Overall, this agent demonstrated a good safety profile in clinical trials, with treatment-emergent AEs reported in 556 (66.7%) patients who received mifepristone vs 386 (61.6%) patients who received placebo.11 Common AEs included gastrointestinal (GI) symptoms, headache, and dizziness. However, 3 deaths occurred: 2 patients who received mifepristone and 1 patient who received placebo. Given this potential for a fatal outcome, clinicians should first consider prescribing an adjunctive antipsychotic agent or electroconvulsive therapy.

Estrogens. These hormones are important for sexual and reproductive development and are used to treat various sexual/reproductive disorders, primarily in women. Their role in treating depression is based on the observation that perimenopause is accompanied by an increased risk of new and recurrent depression coincident with declining ovarian function.

Evidence supports the antidepressant efficacy of transdermal estradiol plus progesterone for perimenopausal depression, but not for postmenopausal depression.12-14 However, estrogens carry significant risks that must be carefully considered in relationship to their potential benefits. These risks include:

  • vaginal bleeding, dysmenorrhea
  • fibroid enlargement
  • galactorrhea
  • ovarian cancer, endometrial cancer, breast cancer
  • deep vein thrombosis, pulmonary embolism
  • hypertension, chest pain, myocardial infarction, stroke.

Continue to: The use of estrogens...

 

 

The use of estrogens as an adjunctive therapy for women with treatment-resistant perimenopausal depression should only be undertaken when standard strategies have failed, and in consultation with an endocrine specialist who can monitor for potentially serious AEs.

Opioid medications

Buprenorphine is used to treat opioid use disorder (OUD) as well as acute and chronic pain. The opioid system is involved in the regulation of mood and may be an appropriate target for novel antidepressants. The use of buprenorphine in combination with samidorphan (a preferential mu-opioid receptor antagonist) has shown initial promise for TRD while minimizing abuse potential.

Although earlier results were mixed, a pooled analysis of 2 recent large RCTs (N = 760) of patients with MDD who had not responded to antidepressants reported greater reduction in Montgomery-Åsberg Depression Rating Scale scores from baseline for active treatment (buprenorphine/samidorphan; 2 mg/2 mg) vs placebo at multiple timepoints, including end of treatment (-1.8; P < .010).15

The most common AEs included nausea, constipation, dizziness, vomiting, somnolence, fatigue, and sedation. There was minimal evidence of abuse, dependence, or opioid withdrawal. Due to the opioid crisis in the United States, the resulting relaxation of regulations regarding prescribing buprenorphine, and the high rates of depression among patients with OUD, buprenorphine/samidorphan, which is an investigational agent that is not FDA-approved, may be particularly helpful for patients with OUD who also experience comorbid TRD.

Antioxidant agents

N-acetylcysteine (NAC) is an amino acid that can treat acetaminophen toxicity and moderate hepatic damage by increasing glutathione levels. Glutathione is also the primary antioxidant in the CNS. NAC may protect against oxidative stress, chelate heavy metals, reduce inflammation, protect against mitochondrial dysfunction, inhibit apoptosis, and enhance neurogenesis, all potential pathophysiological processes that may contribute to depression.16

Continue to: A systematic review...

 

 

A systematic review and meta-analysis of 5 RCTs (N = 574) considered patients with various depression diagnoses who were randomized to adjunctive NAC, 1,000 mg twice a day, or placebo. Over 12 to 24 weeks, there was a significantly greater improvement in mood symptoms and functionality with NAC vs placebo.16

Overall, NAC was well-tolerated. The most common AEs were GI symptoms, musculoskeletal complaints, decreased energy, and headache. While NAC has been touted as a potential adjunct therapy for several psychiatric disorders, including TRD, the evidence for benefit remains limited. Given its favorable AE profile, however, and over-the-counter availability, it remains an option for select patients. It is important to ask patients if they are already taking NAC.

Options beyond off-label medications

There are a multitude of options available for addressing TRD. Many FDA-approved medications are repurposed and prescribed off-label for other indications when the risk/benefit balance is favorable. In Part 1 of this article, we reviewed several off-label medications that have supportive controlled data for treating TRD. In Part 2, we will review other nontraditional therapies for TRD, including herbal/nutraceuticals, anti-inflammatory/immune system therapies, device-based treatments, and other alternative approaches.

Bottom Line

Off-label medications that may offer benefit for patients with treatment-resistant depression (TRD) include pimavanserin, antihypertensive agents, ketamine, scopolamine, botulinum toxin, mifepristone, estrogens, buprenorphine, and N-acetylcysteine. Although some evidence supports use of these agents as adjuncts for TRD, an individualized risk/benefit analysis is required.

Related Resource

Drug Brand Names

Amlodipine • Katerzia, Norvasc
Atenolol • Tenormin
Bisoprolol • Zebeta
Buprenorphine • Sublocade, Subutex
Carvedilol • Coreg
Enalapril • Vasotec
Esketamine • Spravato
Estradiol transdermal • Estraderm
Ketamine • Ketalar
Mifepristone • Mifeprex
Pimavanserin • Nuplazid
Progesterone • Prometrium
Propranolol • Inderal
Ramipril • Altace
Verapamil • Calan, Verelan

[embed:render:related:node:227598]

References

1. Fava M, Dirks B, Freeman M, et al. A phase 2, randomized, double-blind, placebo-controlled study of adjunctive pimavanserin in patients with major depressive disorder and an inadequate response to therapy (CLARITY). J Clin Psychiatry. 2019;80(6):19m12928.

2. Jha MK, Fava M, Freeman MP, et al. Effect of adjunctive pimavanserin on sleep/wakefulness in patients with major depressive disorder: secondary analysis from CLARITY. J Clin Psychiatry. 2020;82(1):20m13425.

3. ACADIA Pharmaceuticals announces top-line results from the Phase 3 CLARITY study evaluating pimavanserin for the adjunctive treatment of major depressive disorder. News release. Acadia Pharmaceuticals Inc. Published July 20, 2020. https://ir.acadia-pharm.com/news-releases/news-release-details/acadia-pharmaceuticals-announces-top-line-results-phase-3-0

4. Kessing LV, Rytgaard HC, Ekstrom CT, et al. Antihypertensive drugs and risk of depression: a nationwide population-based study. Hypertension. 2020;76(4):1263-1279.

5. Williams NR, Heifets BD, Blasey C, et al. Attenuation of antidepressant effects of ketamine by opioid receptor antagonism. Am J Psychiatry. 2018;175(12):1205-1215.

6. Han Y, Chen J, Zou D, et al. Efficacy of ketamine in the rapid treatment of major depressive disorder: a meta-analysis of randomized, double-blind, placebo-controlled studies. Neuropsychiatr Dis Treat. 2016;12:2859-2867.

7. Wan LB, Levitch CF, Perez AM, et al. Ketamine safety and tolerability in clinical trials for treatment-resistant depression. J Clin Psychiatry. 2015;76(3):247-252.

8. Hasselmann, H. Scopolamine and depression: a role for muscarinic antagonism? CNS Neurol Disord Drug Targets. 2014;13(4):673-683.

9. Drevets WC, Zarate CA Jr, Furey ML. Antidepressant effects of the muscarinic cholinergic receptor antagonist scopolamine: a review. Biol Psychiatry. 2013;73(12):1156-1163.

10. Stearns TP, Shad MU, Guzman GC. Glabellar botulinum toxin injections in major depressive disorder: a critical review. Prim Care Companion CNS Disord. 2018;20(5): 18r02298.

11. Block TS, Kushner H, Kalin N, et al. Combined analysis of mifepristone for psychotic depression: plasma levels associated with clinical response. Biol Psychiatry. 2018;84(1):46-54.

12. Rubinow DR, Johnson SL, Schmidt PJ, et al. Efficacy of estradiol in perimenopausal depression: so much promise and so few answers. Depress Anxiety. 2015;32(8):539-549.

13. Schmidt PJ, Ben Dor R, Martinez PE, et al. Effects of estradiol withdrawal on mood in women with past perimenopausal depression: a randomized clinical trial. JAMA Psychiatry. 2015;72(7):714-726.

14. Gordon JL, Rubinow DR, Eisenlohr-Moul TA, et al. Efficacy of transdermal estradiol and micronized progesterone in the prevention of depressive symptoms in the menopause transition: a randomized clinical trial. JAMA Psychiatry. 2018;75(2):149-157.

15. Fava M, Thase ME, Trivedi MH, et al. Opioid system modulation with buprenorphine/samidorphan combination for major depressive disorder: two randomized controlled studies. Mol Psychiatry. 2020;25(7):1580-1591.

16. Fernandes BS, Dean OM, Dodd S, et al. N-Acetylcysteine in depressive symptoms and functionality: a systematic review and meta-analysis. J Clin Psychiatry. 2016;77(4):e457-466.

Article PDF
Author and Disclosure Information

Mehmet E. Dokucu, MD, PhD
Associate Professor
Department of Psychiatry and Behavioral Sciences
Northwestern University Feinberg School of Medicine
Chicago, Illinois

Philip G. Janicak, MD
Adjunct Professor
Department of Psychiatry and Behavioral Sciences
Northwestern University Feinberg School of Medicine
Chicago, Illinois

Disclosures
Dr. Dokucu reports no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products. Dr. Janicak is a speaker for Otsuka PsychU program and TMS Health Solutions.

Issue
Current Psychiatry - 20(9)
Publications
Topics
Page Number
38-43,49
Sections
Author and Disclosure Information

Mehmet E. Dokucu, MD, PhD
Associate Professor
Department of Psychiatry and Behavioral Sciences
Northwestern University Feinberg School of Medicine
Chicago, Illinois

Philip G. Janicak, MD
Adjunct Professor
Department of Psychiatry and Behavioral Sciences
Northwestern University Feinberg School of Medicine
Chicago, Illinois

Disclosures
Dr. Dokucu reports no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products. Dr. Janicak is a speaker for Otsuka PsychU program and TMS Health Solutions.

Author and Disclosure Information

Mehmet E. Dokucu, MD, PhD
Associate Professor
Department of Psychiatry and Behavioral Sciences
Northwestern University Feinberg School of Medicine
Chicago, Illinois

Philip G. Janicak, MD
Adjunct Professor
Department of Psychiatry and Behavioral Sciences
Northwestern University Feinberg School of Medicine
Chicago, Illinois

Disclosures
Dr. Dokucu reports no financial relationships with any companies whose products are mentioned in this article, or with manufacturers of competing products. Dr. Janicak is a speaker for Otsuka PsychU program and TMS Health Solutions.

Article PDF
Article PDF

Presently, FDA-approved first-line treatments and standard adjunctive strategies (eg, lithium, thyroid supplementation, stimulants, second-generation antipsychotics) for major depressive disorder (MDD) often produce less-than-desired outcomes while carrying a potentially substantial safety and tolerability burden. The lack of clinically useful and individual-based biomarkers (eg, genetic, neurophysiological, imaging) is a major obstacle to enhancing treatment efficacy and/or decreasing associated adverse effects (AEs). While the discovery of such tools is being aggressively pursued and ultimately will facilitate a more precision-based choice of therapy, empirical strategies remain our primary approach.

In controlled trials, several nontraditional treatments used primarily as adjuncts to standard antidepressants have shown promise. These include “repurposed” (off-label) medications, herbal/nutraceuticals, anti-inflammatory/immune system therapies, device-based treatments, and other alternative approaches.

Importantly, some nontraditional treatments also demonstrate AEs (Table1-16). With a careful consideration of the risk/benefit balance, this article reviews some of the better-studied treatment options for patients with treatment-resistant depression (TRD). In Part 1, we will examine off-label medications. In Part 2, we will review other nontraditional approaches to TRD, including herbal/nutraceuticals, anti-inflammatory/immune system therapies, device-based treatments, and other alternative approaches.

CP02009038_t1.PNG

We believe this review will help clinicians who need to formulate a different approach after their patient with depression is not helped by traditional first-, second-, and third-line treatments. The potential options discussed in Part 1 of this article are categorized based on their putative mechanism of action (MOA) for depression.

Serotonergic and noradrenergic strategies

Pimavanserin is FDA-approved for treatment of Parkinson’s psychosis. Its potential MOA as an adjunctive strategy for MDD may involve 5-HT2A antagonist and inverse agonist receptor activity, as well as lesser effects at the 5-HT2Creceptor.

A 2-stage, 5-week randomized controlled trial (RCT) (CLARITY; N = 207) found adjunctive pimavanserin (34 mg/d) produced a robust antidepressant effect vs placebo in patients whose depression did not respond to selective serotonin reuptake inhibitors (SSRIs) or serotonin-norepinephrine reuptake inhibitors (SNRIs).1 Furthermore, a secondary analysis of the data suggested that pimavanserin also improved sleepiness (P < .0003) and daily functioning (P < .014) at Week 5.2

Unfortunately, two 6-week, Phase III RCTs (CLARITY-2 and -3; N = 298) did not find a statistically significant difference between active treatment and placebo. This was based on change in the primary outcome measure (Hamilton Depression Rating Scale-17 score) when adjunctive pimavanserin (34 mg/d) was added to an SSRI or SNRI in patients with TRD.3 There was, however, a significant difference favoring active treatment over placebo based on the Clinical Global Impression–Severity score.

Continue to: In these trials...

 

 

In these trials, pimavanserin was generally well-tolerated. The most common AEs were dry mouth, nausea, and headache. Pimavanserin has minimal activity at norepinephrine, dopamine, histamine, or acetylcholine receptors, thus avoiding AEs associated with these receptor interactions.

Given the mixed efficacy results of existing trials, further studies are needed to clarify this agent’s overall risk/benefit in the context of TRD.

Antihypertensive medications

Emerging data suggest that some beta-adrenergic blockers, angiotensin-inhibiting agents, and calcium antagonists are associated with a decreased incidence of depression. A large 2020 study (N = 3,747,190) used population-based Danish registries (2005 to 2015) to evaluate if any of the 41 most commonly prescribed antihypertensive medications were associated with the diagnosis of depressive disorder or use of antidepressants.4 These researchers found that enalapril, ramipril, amlodipine, propranolol, atenolol, bisoprolol, carvedilol (P < .001), and verapamil (P < .004) were strongly associated with a decreased risk of depression.4

Adverse effects across these different classes of antihypertensives are well characterized, can be substantial, and commonly are related to their impact on cardiovascular function (eg, hypotension). Clinically, these agents may be potential adjuncts for patients with TRD who need antihypertensive therapy. Their use and the choice of specific agent should only be determined in consultation with the patient’s primary care physician (PCP) or appropriate specialist.

Glutamatergic strategies

Ketamine is a dissociative anesthetic and analgesic. Its MOA for treating depression appears to occur primarily through antagonist activity at the N-methyl-D-aspartate ionotropic receptor of the glutamatergic system. There is preliminary evidence that its opioid receptor actions also may contribute to its antidepressant effect.5

Continue to: Many published studies...

 

 

Many published studies and reviews have described ketamine’s role for treating MDD. Several studies have reported that low-dose (0.5 mg/kg) IV ketamine infusions can rapidly attenuate severe episodes of MDD as well as associated suicidality. For example, a meta-analysis of 9 RCTs (N = 368) comparing ketamine to placebo for acute treatment of unipolar and bipolar depression reported superior therapeutic effects with active treatment at 24 hours, 72 hours, and 7 days.6 The response and remission rates for ketamine were 52% and 21% at 24 hours; 48% and 24% at 72 hours; and 40% and 26% at 7 days, respectively.6

The most commonly reported AEs during the 4 hours after ketamine infusion included7:

  • drowsiness, dizziness, poor coordination
  • blurred vision, feeling strange or unreal
  • hemodynamic changes (approximately 33%)
  • small but significant (P < .05) increases in psychotomimetic and dissociative symptoms.

Because some individuals use ketamine recreationally, this agent also carries the risk of abuse.

Research is ongoing on strategies for long-term maintenance ketamine treatment, and the results of both short- and long-term trials will require careful scrutiny to better assess this agent’s safety and tolerability. Clinicians should first consider esketamine—the S-enantiomer of ketamine—because an intranasal formulation of this agent is FDA-approved for treating patients with TRD or MDD with suicidality when administered in a Risk Evaluation and Mitigation Strategy–certified setting.

Cholinergic strategies

Scopolamine is a potent muscarinic receptor antagonist used to prevent nausea and vomiting caused by motion sickness or medications used during surgery. Its use for MDD is based on the theory that muscarinic receptors may be hypersensitive in mood disorders.

Continue to: Several double-blind RCTs...

 

 

Several double-blind RCTs of patients with unipolar or bipolar depression that used 3 pulsed IV infusions (4.0 mcg/kg) over 15 minutes found a rapid, robust antidepressant effect with scopolamine vs placebo.8,9 The oral formulation might also be effective, but would not have a rapid onset.

Common adverse effects of scopolamine include agitation, dry mouth, urinary retention, and cognitive clouding. Given scopolamine’s substantial AE profile, it should be considered only for patients with TRD who could also benefit from the oral formulation for the medical indications noted above, should generally be avoided in older patients, and should be prescribed in consultation with the patient’s PCP.

Botulinum toxin. This neurotoxin inhibits acetylcholine release. It is used to treat disorders characterized by abnormal muscular contraction, such as strabismus, blepharospasm, and chronic pain syndromes. Its MOA for depression may involve its paralytic effects after injection into the glabella forehead muscle (based on the facial feedback hypothesis), as well as modulation of neurotransmitters implicated in the pathophysiology of depression.

In several small trials, injectable botulinum toxin type A (BTA) (29 units) demonstrated antidepressant effects. A recent review that considered 6 trials (N = 235; 4 of the 6 studies were RCTs, 3 of which were rated as high quality) concluded that BTA may be a promising treatment for MDD.10 Limitations of this review included lack of a priori hypotheses, small sample sizes, gender bias, and difficulty in blinding.

In clinical trials, the most common AEs included local irritation at the injection site and transient headache. This agent’s relatively mild AE profile and possible overlap when used for some of the medical indications noted above opens its potential use as an adjunct in patients with comorbid TRD.

Continue to: Endocrine strategies

 

 

Endocrine strategies

Mifepristone (RU486). This anti-glucocorticoid receptor antagonist is used as an abortifacient. Based on the theory that hyperactivity of the hypothalamic-pituitary-adrenal axis is implicated in the pathophysiology of MDD with psychotic features (psychotic depression), this agent has been studied as a treatment for this indication.

An analysis of 5 double-blind RCTs (N = 1,460) found that 7 days of mifepristone, 1,200 mg/d, was superior to placebo (P < .004) in reducing psychotic symptoms of depression.11 Plasma concentrations ≥1,600 ng/mL may be required to maximize benefit.11

Overall, this agent demonstrated a good safety profile in clinical trials, with treatment-emergent AEs reported in 556 (66.7%) patients who received mifepristone vs 386 (61.6%) patients who received placebo.11 Common AEs included gastrointestinal (GI) symptoms, headache, and dizziness. However, 3 deaths occurred: 2 patients who received mifepristone and 1 patient who received placebo. Given this potential for a fatal outcome, clinicians should first consider prescribing an adjunctive antipsychotic agent or electroconvulsive therapy.

Estrogens. These hormones are important for sexual and reproductive development and are used to treat various sexual/reproductive disorders, primarily in women. Their role in treating depression is based on the observation that perimenopause is accompanied by an increased risk of new and recurrent depression coincident with declining ovarian function.

Evidence supports the antidepressant efficacy of transdermal estradiol plus progesterone for perimenopausal depression, but not for postmenopausal depression.12-14 However, estrogens carry significant risks that must be carefully considered in relationship to their potential benefits. These risks include:

  • vaginal bleeding, dysmenorrhea
  • fibroid enlargement
  • galactorrhea
  • ovarian cancer, endometrial cancer, breast cancer
  • deep vein thrombosis, pulmonary embolism
  • hypertension, chest pain, myocardial infarction, stroke.

Continue to: The use of estrogens...

 

 

The use of estrogens as an adjunctive therapy for women with treatment-resistant perimenopausal depression should only be undertaken when standard strategies have failed, and in consultation with an endocrine specialist who can monitor for potentially serious AEs.

Opioid medications

Buprenorphine is used to treat opioid use disorder (OUD) as well as acute and chronic pain. The opioid system is involved in the regulation of mood and may be an appropriate target for novel antidepressants. The use of buprenorphine in combination with samidorphan (a preferential mu-opioid receptor antagonist) has shown initial promise for TRD while minimizing abuse potential.

Although earlier results were mixed, a pooled analysis of 2 recent large RCTs (N = 760) of patients with MDD who had not responded to antidepressants reported greater reduction in Montgomery-Åsberg Depression Rating Scale scores from baseline for active treatment (buprenorphine/samidorphan; 2 mg/2 mg) vs placebo at multiple timepoints, including end of treatment (-1.8; P < .010).15

The most common AEs included nausea, constipation, dizziness, vomiting, somnolence, fatigue, and sedation. There was minimal evidence of abuse, dependence, or opioid withdrawal. Due to the opioid crisis in the United States, the resulting relaxation of regulations regarding prescribing buprenorphine, and the high rates of depression among patients with OUD, buprenorphine/samidorphan, which is an investigational agent that is not FDA-approved, may be particularly helpful for patients with OUD who also experience comorbid TRD.

Antioxidant agents

N-acetylcysteine (NAC) is an amino acid that can treat acetaminophen toxicity and moderate hepatic damage by increasing glutathione levels. Glutathione is also the primary antioxidant in the CNS. NAC may protect against oxidative stress, chelate heavy metals, reduce inflammation, protect against mitochondrial dysfunction, inhibit apoptosis, and enhance neurogenesis, all potential pathophysiological processes that may contribute to depression.16

Continue to: A systematic review...

 

 

A systematic review and meta-analysis of 5 RCTs (N = 574) considered patients with various depression diagnoses who were randomized to adjunctive NAC, 1,000 mg twice a day, or placebo. Over 12 to 24 weeks, there was a significantly greater improvement in mood symptoms and functionality with NAC vs placebo.16

Overall, NAC was well-tolerated. The most common AEs were GI symptoms, musculoskeletal complaints, decreased energy, and headache. While NAC has been touted as a potential adjunct therapy for several psychiatric disorders, including TRD, the evidence for benefit remains limited. Given its favorable AE profile, however, and over-the-counter availability, it remains an option for select patients. It is important to ask patients if they are already taking NAC.

Options beyond off-label medications

There are a multitude of options available for addressing TRD. Many FDA-approved medications are repurposed and prescribed off-label for other indications when the risk/benefit balance is favorable. In Part 1 of this article, we reviewed several off-label medications that have supportive controlled data for treating TRD. In Part 2, we will review other nontraditional therapies for TRD, including herbal/nutraceuticals, anti-inflammatory/immune system therapies, device-based treatments, and other alternative approaches.

Bottom Line

Off-label medications that may offer benefit for patients with treatment-resistant depression (TRD) include pimavanserin, antihypertensive agents, ketamine, scopolamine, botulinum toxin, mifepristone, estrogens, buprenorphine, and N-acetylcysteine. Although some evidence supports use of these agents as adjuncts for TRD, an individualized risk/benefit analysis is required.

Related Resource

Drug Brand Names

Amlodipine • Katerzia, Norvasc
Atenolol • Tenormin
Bisoprolol • Zebeta
Buprenorphine • Sublocade, Subutex
Carvedilol • Coreg
Enalapril • Vasotec
Esketamine • Spravato
Estradiol transdermal • Estraderm
Ketamine • Ketalar
Mifepristone • Mifeprex
Pimavanserin • Nuplazid
Progesterone • Prometrium
Propranolol • Inderal
Ramipril • Altace
Verapamil • Calan, Verelan

[embed:render:related:node:227598]

Presently, FDA-approved first-line treatments and standard adjunctive strategies (eg, lithium, thyroid supplementation, stimulants, second-generation antipsychotics) for major depressive disorder (MDD) often produce less-than-desired outcomes while carrying a potentially substantial safety and tolerability burden. The lack of clinically useful and individual-based biomarkers (eg, genetic, neurophysiological, imaging) is a major obstacle to enhancing treatment efficacy and/or decreasing associated adverse effects (AEs). While the discovery of such tools is being aggressively pursued and ultimately will facilitate a more precision-based choice of therapy, empirical strategies remain our primary approach.

In controlled trials, several nontraditional treatments used primarily as adjuncts to standard antidepressants have shown promise. These include “repurposed” (off-label) medications, herbal/nutraceuticals, anti-inflammatory/immune system therapies, device-based treatments, and other alternative approaches.

Importantly, some nontraditional treatments also demonstrate AEs (Table1-16). With a careful consideration of the risk/benefit balance, this article reviews some of the better-studied treatment options for patients with treatment-resistant depression (TRD). In Part 1, we will examine off-label medications. In Part 2, we will review other nontraditional approaches to TRD, including herbal/nutraceuticals, anti-inflammatory/immune system therapies, device-based treatments, and other alternative approaches.

CP02009038_t1.PNG

We believe this review will help clinicians who need to formulate a different approach after their patient with depression is not helped by traditional first-, second-, and third-line treatments. The potential options discussed in Part 1 of this article are categorized based on their putative mechanism of action (MOA) for depression.

Serotonergic and noradrenergic strategies

Pimavanserin is FDA-approved for treatment of Parkinson’s psychosis. Its potential MOA as an adjunctive strategy for MDD may involve 5-HT2A antagonist and inverse agonist receptor activity, as well as lesser effects at the 5-HT2Creceptor.

A 2-stage, 5-week randomized controlled trial (RCT) (CLARITY; N = 207) found adjunctive pimavanserin (34 mg/d) produced a robust antidepressant effect vs placebo in patients whose depression did not respond to selective serotonin reuptake inhibitors (SSRIs) or serotonin-norepinephrine reuptake inhibitors (SNRIs).1 Furthermore, a secondary analysis of the data suggested that pimavanserin also improved sleepiness (P < .0003) and daily functioning (P < .014) at Week 5.2

Unfortunately, two 6-week, Phase III RCTs (CLARITY-2 and -3; N = 298) did not find a statistically significant difference between active treatment and placebo. This was based on change in the primary outcome measure (Hamilton Depression Rating Scale-17 score) when adjunctive pimavanserin (34 mg/d) was added to an SSRI or SNRI in patients with TRD.3 There was, however, a significant difference favoring active treatment over placebo based on the Clinical Global Impression–Severity score.

Continue to: In these trials...

 

 

In these trials, pimavanserin was generally well-tolerated. The most common AEs were dry mouth, nausea, and headache. Pimavanserin has minimal activity at norepinephrine, dopamine, histamine, or acetylcholine receptors, thus avoiding AEs associated with these receptor interactions.

Given the mixed efficacy results of existing trials, further studies are needed to clarify this agent’s overall risk/benefit in the context of TRD.

Antihypertensive medications

Emerging data suggest that some beta-adrenergic blockers, angiotensin-inhibiting agents, and calcium antagonists are associated with a decreased incidence of depression. A large 2020 study (N = 3,747,190) used population-based Danish registries (2005 to 2015) to evaluate if any of the 41 most commonly prescribed antihypertensive medications were associated with the diagnosis of depressive disorder or use of antidepressants.4 These researchers found that enalapril, ramipril, amlodipine, propranolol, atenolol, bisoprolol, carvedilol (P < .001), and verapamil (P < .004) were strongly associated with a decreased risk of depression.4

Adverse effects across these different classes of antihypertensives are well characterized, can be substantial, and commonly are related to their impact on cardiovascular function (eg, hypotension). Clinically, these agents may be potential adjuncts for patients with TRD who need antihypertensive therapy. Their use and the choice of specific agent should only be determined in consultation with the patient’s primary care physician (PCP) or appropriate specialist.

Glutamatergic strategies

Ketamine is a dissociative anesthetic and analgesic. Its MOA for treating depression appears to occur primarily through antagonist activity at the N-methyl-D-aspartate ionotropic receptor of the glutamatergic system. There is preliminary evidence that its opioid receptor actions also may contribute to its antidepressant effect.5

Continue to: Many published studies...

 

 

Many published studies and reviews have described ketamine’s role for treating MDD. Several studies have reported that low-dose (0.5 mg/kg) IV ketamine infusions can rapidly attenuate severe episodes of MDD as well as associated suicidality. For example, a meta-analysis of 9 RCTs (N = 368) comparing ketamine to placebo for acute treatment of unipolar and bipolar depression reported superior therapeutic effects with active treatment at 24 hours, 72 hours, and 7 days.6 The response and remission rates for ketamine were 52% and 21% at 24 hours; 48% and 24% at 72 hours; and 40% and 26% at 7 days, respectively.6

The most commonly reported AEs during the 4 hours after ketamine infusion included7:

  • drowsiness, dizziness, poor coordination
  • blurred vision, feeling strange or unreal
  • hemodynamic changes (approximately 33%)
  • small but significant (P < .05) increases in psychotomimetic and dissociative symptoms.

Because some individuals use ketamine recreationally, this agent also carries the risk of abuse.

Research is ongoing on strategies for long-term maintenance ketamine treatment, and the results of both short- and long-term trials will require careful scrutiny to better assess this agent’s safety and tolerability. Clinicians should first consider esketamine—the S-enantiomer of ketamine—because an intranasal formulation of this agent is FDA-approved for treating patients with TRD or MDD with suicidality when administered in a Risk Evaluation and Mitigation Strategy–certified setting.

Cholinergic strategies

Scopolamine is a potent muscarinic receptor antagonist used to prevent nausea and vomiting caused by motion sickness or medications used during surgery. Its use for MDD is based on the theory that muscarinic receptors may be hypersensitive in mood disorders.

Continue to: Several double-blind RCTs...

 

 

Several double-blind RCTs of patients with unipolar or bipolar depression that used 3 pulsed IV infusions (4.0 mcg/kg) over 15 minutes found a rapid, robust antidepressant effect with scopolamine vs placebo.8,9 The oral formulation might also be effective, but would not have a rapid onset.

Common adverse effects of scopolamine include agitation, dry mouth, urinary retention, and cognitive clouding. Given scopolamine’s substantial AE profile, it should be considered only for patients with TRD who could also benefit from the oral formulation for the medical indications noted above, should generally be avoided in older patients, and should be prescribed in consultation with the patient’s PCP.

Botulinum toxin. This neurotoxin inhibits acetylcholine release. It is used to treat disorders characterized by abnormal muscular contraction, such as strabismus, blepharospasm, and chronic pain syndromes. Its MOA for depression may involve its paralytic effects after injection into the glabella forehead muscle (based on the facial feedback hypothesis), as well as modulation of neurotransmitters implicated in the pathophysiology of depression.

In several small trials, injectable botulinum toxin type A (BTA) (29 units) demonstrated antidepressant effects. A recent review that considered 6 trials (N = 235; 4 of the 6 studies were RCTs, 3 of which were rated as high quality) concluded that BTA may be a promising treatment for MDD.10 Limitations of this review included lack of a priori hypotheses, small sample sizes, gender bias, and difficulty in blinding.

In clinical trials, the most common AEs included local irritation at the injection site and transient headache. This agent’s relatively mild AE profile and possible overlap when used for some of the medical indications noted above opens its potential use as an adjunct in patients with comorbid TRD.

Continue to: Endocrine strategies

 

 

Endocrine strategies

Mifepristone (RU486). This anti-glucocorticoid receptor antagonist is used as an abortifacient. Based on the theory that hyperactivity of the hypothalamic-pituitary-adrenal axis is implicated in the pathophysiology of MDD with psychotic features (psychotic depression), this agent has been studied as a treatment for this indication.

An analysis of 5 double-blind RCTs (N = 1,460) found that 7 days of mifepristone, 1,200 mg/d, was superior to placebo (P < .004) in reducing psychotic symptoms of depression.11 Plasma concentrations ≥1,600 ng/mL may be required to maximize benefit.11

Overall, this agent demonstrated a good safety profile in clinical trials, with treatment-emergent AEs reported in 556 (66.7%) patients who received mifepristone vs 386 (61.6%) patients who received placebo.11 Common AEs included gastrointestinal (GI) symptoms, headache, and dizziness. However, 3 deaths occurred: 2 patients who received mifepristone and 1 patient who received placebo. Given this potential for a fatal outcome, clinicians should first consider prescribing an adjunctive antipsychotic agent or electroconvulsive therapy.

Estrogens. These hormones are important for sexual and reproductive development and are used to treat various sexual/reproductive disorders, primarily in women. Their role in treating depression is based on the observation that perimenopause is accompanied by an increased risk of new and recurrent depression coincident with declining ovarian function.

Evidence supports the antidepressant efficacy of transdermal estradiol plus progesterone for perimenopausal depression, but not for postmenopausal depression.12-14 However, estrogens carry significant risks that must be carefully considered in relationship to their potential benefits. These risks include:

  • vaginal bleeding, dysmenorrhea
  • fibroid enlargement
  • galactorrhea
  • ovarian cancer, endometrial cancer, breast cancer
  • deep vein thrombosis, pulmonary embolism
  • hypertension, chest pain, myocardial infarction, stroke.

Continue to: The use of estrogens...

 

 

The use of estrogens as an adjunctive therapy for women with treatment-resistant perimenopausal depression should only be undertaken when standard strategies have failed, and in consultation with an endocrine specialist who can monitor for potentially serious AEs.

Opioid medications

Buprenorphine is used to treat opioid use disorder (OUD) as well as acute and chronic pain. The opioid system is involved in the regulation of mood and may be an appropriate target for novel antidepressants. The use of buprenorphine in combination with samidorphan (a preferential mu-opioid receptor antagonist) has shown initial promise for TRD while minimizing abuse potential.

Although earlier results were mixed, a pooled analysis of 2 recent large RCTs (N = 760) of patients with MDD who had not responded to antidepressants reported greater reduction in Montgomery-Åsberg Depression Rating Scale scores from baseline for active treatment (buprenorphine/samidorphan; 2 mg/2 mg) vs placebo at multiple timepoints, including end of treatment (-1.8; P < .010).15

The most common AEs included nausea, constipation, dizziness, vomiting, somnolence, fatigue, and sedation. There was minimal evidence of abuse, dependence, or opioid withdrawal. Due to the opioid crisis in the United States, the resulting relaxation of regulations regarding prescribing buprenorphine, and the high rates of depression among patients with OUD, buprenorphine/samidorphan, which is an investigational agent that is not FDA-approved, may be particularly helpful for patients with OUD who also experience comorbid TRD.

Antioxidant agents

N-acetylcysteine (NAC) is an amino acid that can treat acetaminophen toxicity and moderate hepatic damage by increasing glutathione levels. Glutathione is also the primary antioxidant in the CNS. NAC may protect against oxidative stress, chelate heavy metals, reduce inflammation, protect against mitochondrial dysfunction, inhibit apoptosis, and enhance neurogenesis, all potential pathophysiological processes that may contribute to depression.16

Continue to: A systematic review...

 

 

A systematic review and meta-analysis of 5 RCTs (N = 574) considered patients with various depression diagnoses who were randomized to adjunctive NAC, 1,000 mg twice a day, or placebo. Over 12 to 24 weeks, there was a significantly greater improvement in mood symptoms and functionality with NAC vs placebo.16

Overall, NAC was well-tolerated. The most common AEs were GI symptoms, musculoskeletal complaints, decreased energy, and headache. While NAC has been touted as a potential adjunct therapy for several psychiatric disorders, including TRD, the evidence for benefit remains limited. Given its favorable AE profile, however, and over-the-counter availability, it remains an option for select patients. It is important to ask patients if they are already taking NAC.

Options beyond off-label medications

There are a multitude of options available for addressing TRD. Many FDA-approved medications are repurposed and prescribed off-label for other indications when the risk/benefit balance is favorable. In Part 1 of this article, we reviewed several off-label medications that have supportive controlled data for treating TRD. In Part 2, we will review other nontraditional therapies for TRD, including herbal/nutraceuticals, anti-inflammatory/immune system therapies, device-based treatments, and other alternative approaches.

Bottom Line

Off-label medications that may offer benefit for patients with treatment-resistant depression (TRD) include pimavanserin, antihypertensive agents, ketamine, scopolamine, botulinum toxin, mifepristone, estrogens, buprenorphine, and N-acetylcysteine. Although some evidence supports use of these agents as adjuncts for TRD, an individualized risk/benefit analysis is required.

Related Resource

Drug Brand Names

Amlodipine • Katerzia, Norvasc
Atenolol • Tenormin
Bisoprolol • Zebeta
Buprenorphine • Sublocade, Subutex
Carvedilol • Coreg
Enalapril • Vasotec
Esketamine • Spravato
Estradiol transdermal • Estraderm
Ketamine • Ketalar
Mifepristone • Mifeprex
Pimavanserin • Nuplazid
Progesterone • Prometrium
Propranolol • Inderal
Ramipril • Altace
Verapamil • Calan, Verelan

[embed:render:related:node:227598]

References

1. Fava M, Dirks B, Freeman M, et al. A phase 2, randomized, double-blind, placebo-controlled study of adjunctive pimavanserin in patients with major depressive disorder and an inadequate response to therapy (CLARITY). J Clin Psychiatry. 2019;80(6):19m12928.

2. Jha MK, Fava M, Freeman MP, et al. Effect of adjunctive pimavanserin on sleep/wakefulness in patients with major depressive disorder: secondary analysis from CLARITY. J Clin Psychiatry. 2020;82(1):20m13425.

3. ACADIA Pharmaceuticals announces top-line results from the Phase 3 CLARITY study evaluating pimavanserin for the adjunctive treatment of major depressive disorder. News release. Acadia Pharmaceuticals Inc. Published July 20, 2020. https://ir.acadia-pharm.com/news-releases/news-release-details/acadia-pharmaceuticals-announces-top-line-results-phase-3-0

4. Kessing LV, Rytgaard HC, Ekstrom CT, et al. Antihypertensive drugs and risk of depression: a nationwide population-based study. Hypertension. 2020;76(4):1263-1279.

5. Williams NR, Heifets BD, Blasey C, et al. Attenuation of antidepressant effects of ketamine by opioid receptor antagonism. Am J Psychiatry. 2018;175(12):1205-1215.

6. Han Y, Chen J, Zou D, et al. Efficacy of ketamine in the rapid treatment of major depressive disorder: a meta-analysis of randomized, double-blind, placebo-controlled studies. Neuropsychiatr Dis Treat. 2016;12:2859-2867.

7. Wan LB, Levitch CF, Perez AM, et al. Ketamine safety and tolerability in clinical trials for treatment-resistant depression. J Clin Psychiatry. 2015;76(3):247-252.

8. Hasselmann, H. Scopolamine and depression: a role for muscarinic antagonism? CNS Neurol Disord Drug Targets. 2014;13(4):673-683.

9. Drevets WC, Zarate CA Jr, Furey ML. Antidepressant effects of the muscarinic cholinergic receptor antagonist scopolamine: a review. Biol Psychiatry. 2013;73(12):1156-1163.

10. Stearns TP, Shad MU, Guzman GC. Glabellar botulinum toxin injections in major depressive disorder: a critical review. Prim Care Companion CNS Disord. 2018;20(5): 18r02298.

11. Block TS, Kushner H, Kalin N, et al. Combined analysis of mifepristone for psychotic depression: plasma levels associated with clinical response. Biol Psychiatry. 2018;84(1):46-54.

12. Rubinow DR, Johnson SL, Schmidt PJ, et al. Efficacy of estradiol in perimenopausal depression: so much promise and so few answers. Depress Anxiety. 2015;32(8):539-549.

13. Schmidt PJ, Ben Dor R, Martinez PE, et al. Effects of estradiol withdrawal on mood in women with past perimenopausal depression: a randomized clinical trial. JAMA Psychiatry. 2015;72(7):714-726.

14. Gordon JL, Rubinow DR, Eisenlohr-Moul TA, et al. Efficacy of transdermal estradiol and micronized progesterone in the prevention of depressive symptoms in the menopause transition: a randomized clinical trial. JAMA Psychiatry. 2018;75(2):149-157.

15. Fava M, Thase ME, Trivedi MH, et al. Opioid system modulation with buprenorphine/samidorphan combination for major depressive disorder: two randomized controlled studies. Mol Psychiatry. 2020;25(7):1580-1591.

16. Fernandes BS, Dean OM, Dodd S, et al. N-Acetylcysteine in depressive symptoms and functionality: a systematic review and meta-analysis. J Clin Psychiatry. 2016;77(4):e457-466.

References

1. Fava M, Dirks B, Freeman M, et al. A phase 2, randomized, double-blind, placebo-controlled study of adjunctive pimavanserin in patients with major depressive disorder and an inadequate response to therapy (CLARITY). J Clin Psychiatry. 2019;80(6):19m12928.

2. Jha MK, Fava M, Freeman MP, et al. Effect of adjunctive pimavanserin on sleep/wakefulness in patients with major depressive disorder: secondary analysis from CLARITY. J Clin Psychiatry. 2020;82(1):20m13425.

3. ACADIA Pharmaceuticals announces top-line results from the Phase 3 CLARITY study evaluating pimavanserin for the adjunctive treatment of major depressive disorder. News release. Acadia Pharmaceuticals Inc. Published July 20, 2020. https://ir.acadia-pharm.com/news-releases/news-release-details/acadia-pharmaceuticals-announces-top-line-results-phase-3-0

4. Kessing LV, Rytgaard HC, Ekstrom CT, et al. Antihypertensive drugs and risk of depression: a nationwide population-based study. Hypertension. 2020;76(4):1263-1279.

5. Williams NR, Heifets BD, Blasey C, et al. Attenuation of antidepressant effects of ketamine by opioid receptor antagonism. Am J Psychiatry. 2018;175(12):1205-1215.

6. Han Y, Chen J, Zou D, et al. Efficacy of ketamine in the rapid treatment of major depressive disorder: a meta-analysis of randomized, double-blind, placebo-controlled studies. Neuropsychiatr Dis Treat. 2016;12:2859-2867.

7. Wan LB, Levitch CF, Perez AM, et al. Ketamine safety and tolerability in clinical trials for treatment-resistant depression. J Clin Psychiatry. 2015;76(3):247-252.

8. Hasselmann, H. Scopolamine and depression: a role for muscarinic antagonism? CNS Neurol Disord Drug Targets. 2014;13(4):673-683.

9. Drevets WC, Zarate CA Jr, Furey ML. Antidepressant effects of the muscarinic cholinergic receptor antagonist scopolamine: a review. Biol Psychiatry. 2013;73(12):1156-1163.

10. Stearns TP, Shad MU, Guzman GC. Glabellar botulinum toxin injections in major depressive disorder: a critical review. Prim Care Companion CNS Disord. 2018;20(5): 18r02298.

11. Block TS, Kushner H, Kalin N, et al. Combined analysis of mifepristone for psychotic depression: plasma levels associated with clinical response. Biol Psychiatry. 2018;84(1):46-54.

12. Rubinow DR, Johnson SL, Schmidt PJ, et al. Efficacy of estradiol in perimenopausal depression: so much promise and so few answers. Depress Anxiety. 2015;32(8):539-549.

13. Schmidt PJ, Ben Dor R, Martinez PE, et al. Effects of estradiol withdrawal on mood in women with past perimenopausal depression: a randomized clinical trial. JAMA Psychiatry. 2015;72(7):714-726.

14. Gordon JL, Rubinow DR, Eisenlohr-Moul TA, et al. Efficacy of transdermal estradiol and micronized progesterone in the prevention of depressive symptoms in the menopause transition: a randomized clinical trial. JAMA Psychiatry. 2018;75(2):149-157.

15. Fava M, Thase ME, Trivedi MH, et al. Opioid system modulation with buprenorphine/samidorphan combination for major depressive disorder: two randomized controlled studies. Mol Psychiatry. 2020;25(7):1580-1591.

16. Fernandes BS, Dean OM, Dodd S, et al. N-Acetylcysteine in depressive symptoms and functionality: a systematic review and meta-analysis. J Clin Psychiatry. 2016;77(4):e457-466.

Issue
Current Psychiatry - 20(9)
Issue
Current Psychiatry - 20(9)
Page Number
38-43,49
Page Number
38-43,49
Publications
Publications
Topics
Article Type
Display Headline
Nontraditional therapies for treatment-resistant depression
Display Headline
Nontraditional therapies for treatment-resistant depression
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

What’s new in transcranial magnetic stimulation

Article Type
Changed
Fri, 02/21/2020 - 14:17
Display Headline
What’s new in transcranial magnetic stimulation

Therapeutic neuromodulation takes advantage of the brain’s electrochemical makeup. This allows for treatment devices that modulate neurocircuits relevant to behaviors disrupted in disorders such as major depressive disorder (MDD) (eg, sleep quality, appetite, cognitive, and executive functions). The default mode network (comprised of structures such as the medial prefrontal cortex [MPFC], the posterior cingulate cortex, the hippocampus, and their functional connectivity) serves as a prime example of circuitry that can be targeted by this approach.1

For 80 years, electroconvulsive therapy (ECT) has been an important neuromodulation option for patients with more severe illness. Recently, additional neuromodulatory approaches have been FDA-cleared, including transcranial magnetic stimulation (TMS), vagus nerve stimulation (VNS), and deep brain stimulation (DBS). Another approach, transcranial direct current stimulation (tDCS), has been extensively studied for its potential clinical utility but is not FDA-cleared. The Table provides descriptions of these therapies.

CP01803010_t1.PNG


Since being cleared by the FDA in 2008, TMS has arguably made the greatest strides in providing an alternate neuromodulation treatment option for patients with MDD, with >1,000 centers nationally and 7 TMS devices FDA-cleared for treatment of depression. In this article, we review recent developments in TMS.

 

An evolving therapeutic option

While primarily studied as a monotherapy for MDD, in clinical practice TMS (Box) is typically used as an adjunct to medication and psychotherapy.2,3 In this context, it has demonstrated efficacy for more difficult-to-treat mood disorders with an excellent safety and tolerability profile whether used with or without medication.4-6

To further improve the efficiency and efficacy of TMS while maintaining its safety and tolerability, researchers and clinicians have been exploring a few initiatives.

Box

Key points: Transcranial magnetic stimulation
  • Transcranial magnetic stimulation (TMS) utilizes intense, localized magnetic fields to alter activity in neural circuits implicated in the pathophysiology of depression
  • Randomized, sham-controlled acute trials have demonstrated the efficacy of TMS for treatment-resistant depression
  • Clinical availability of TMS has grown steadily over the past 10 years as >1,000 centers have been opened and additional devices have been FDA-cleared
  • TMS has the potential to avoid safety and tolerability concerns associated with antidepressant pharmacotherapy (eg, weight gain, sexual dysfunction) and electroconvulsive therapy (eg, cognitive deficits)
  • Greater sophistication in the choice of stimulation parameters, as well as other ongoing efforts to optimize the benefits of TMS, are yielding better clinical outcomes

Altered treatment parameters

One initiative is assessing the feasibility of altering various treatment parameters, such as the total number of treatment sessions (30 to 60 sessions); the frequency of sessions (eg, more than once daily); the total number of magnetic pulses per session (eg, >3,000); the stimulation coil localization (eg, left vs right dorsal lateral prefrontal cortex [DLPFC]; MPFC; and various methods to determine optimal coil placement (eg, EEG F3 coordinate or MRI-guided neuro-navigational methods). Such refinements offer the potential for enhanced efficacy, shorter treatment sessions, and/or improved tolerability. For example, lower frequency right DLPFC stimulations (eg, 1 Hz) can decrease the risk of seizures and improve overall tolerability. While this has not been studied as extensively as higher frequency left DLPFC stimulations (eg, 5 to 20 Hz), existing evidence supports similar efficacy between these 2 approaches.7

Theta burst stimulation. Some TMS devices can be adapted to deliver theta burst stimulation (TBS). This produces trains of triple, 50 Hz, pulsed bursts (usually with 200 ms inter-burst intervals occurring at a rate of 5 Hz; at 80% MT) to model naturally occurring theta rhythms. These bursts can be administered in stimulation protocols using intermittent TBS (iTBS) (eg, 10 bursts of triplets over 2 seconds every 10 seconds; 30 pulses per burst; for approximately 3 minutes; totaling 600 pulses) or continuous TBS (cTBS) bursts given in an uninterrupted train (eg, 40 seconds, 600 pulses). Evidence indicates these protocols facilitate long-term potentiation (ie, iTBS) and long-term depression (ie, cTBS), which in turn can modulate synaptic plasticity.

Continue to: While some clinicians are using...

 

 

While some clinicians are using TBS off-label, a recent non-inferiority trial (N = 395) reported similar efficacy and safety comparing standard 10 Hz TMS to an iTBS protocol at 120% of resting motor threshold (both over the left DLPFC).8 This has led to FDA clearance of the TMS device adapted to provide iTBS in this trial.8

From a more practical perspective, TBS has the potential to reduce the number of pulses (eg, 600 vs 3,000) and the total number of sessions required, as well as the duration of treatment sessions (eg, 37.5 minutes to <5 minutes). This can accelerate the time to response and decrease patient and staff commitment, with resulting cost savings.9 Despite this recent progress, ongoing research still needs to clarify issues such as the risk/benefit profile, particularly in younger and older populations, as well as assessment of duration of initial benefit and appropriate maintenance strategies.

New devices

Another initiative is the development of alternative TMS equipment. For example, newer coil designs with enhanced cooling ability allow for a substantial decrease in the required inter-train interval duration between stimulation trains, thus shortening the total session duration by approximately 50% (eg, from 37.5 to 19 minutes). The use of different coil arrays (eg, the H-coil capable of deeper vs surface stimulation) may allow for more direct stimulation of relevant neurocircuitry (eg, cingulate cortex), possibly improving efficacy and shortening time to onset of benefit. However, in head-to-head comparisons with single-coil devices, enhanced efficacy for depression has not been clearly demonstrated. One caveat is that the increase in depth of magnetic field penetration results in a loss of focality, resulting in the stimulation of larger brain areas. This might increase the risk of adverse effects such as seizures.

 

Increasing durability of effect

Because high relapse and recurrence rates compromise the initial benefit of any antidepressant therapy, appropriate maintenance strategies are essential. Several studies have evaluated strategies to maintain the acute benefit of TMS for treatment-resistant depression.

One was a 6-month, open-label TMS durability of effect trial for acute responders (n = 99) in the pivotal registration study.5 During this study, all participants were given antidepressant medication monotherapy. In addition, with early indication of relapse, patients received a reintroduction of TMS sessions (32/99 patients; mean number of sessions = 14.3). With this protocol, approximately 84% re-achieved their response status. The overall relapse rate was approximately 13%.5

Continue to: In a 1-year naturalistic study...

 

 

In a 1-year naturalistic study, 63% of patients (75/120) who met response or remission criteria after an acute course of TMS still met response criteria after 12 months. These patients received clinician-determined maintenance treatment that included reintroduction of TMS when indicated.3

In a prospective, 12-month, multisite, randomized pilot study, 67 patients with treatment-resistant MDD underwent an antidepressant medication washout and then received 30 sessions of TMS monotherapy.10 Those who met criteria for improvement (n = 49) were then randomized to once-monthly TMS or observation only. All patients remained medication-free but could receive TMS re-introduction if they deteriorated. At the end of the study, both groups demonstrated comparable outcomes, with a trend to a longer time before relapse among participants who received once-monthly TMS. Although these results are preliminary, they suggest that some patients could be treated both acutely and then maintained with TMS alone.

Re-introducing TMS in patients who show early signs of relapse after having an initial response achieves rates of sustained improvement that compare favorably with those of other strategies used to manage patients with treatment-resistant depression.

 

TMS vs ECT

The question often arises as to whether TMS is a viable alternate treatment to ECT. I believe the answer is unequivocally yes and no. By this, I mean some patients who in the past only had ECT as their next option when medications and psychotherapy were insufficient may now consider TMS. In support, there is evidence of comparable efficacy between TMS and ECT in a subgroup of patients who were considered clinically appropriate for ECT.11-13

How to best identify this group remains unclear, but investigators are exploring predictive biomarkers. For example, a large study (N = 1,188), with functional magnetic resonance imaging (fMRI) reported that depressed patients could be divided into 4 neurophysiological “biotypes” based on different patterns of aberrant connectivity in limbic and fronto-striatal networks.14 The authors further noted that such distinctions were helpful in predicting response in a subgroup of patients (n = 154) who received TMS.

Continue to: For now...

 

 

For now, experience indicates certain clinical factors may provide some guidance. Patients are usually better served by ECT if they:

  • have depressive episodes of longer duration (eg, >3 years)
  • have a high risk of suicide
  • have psychotic or catatonic features associated with their depression
  • have difficulty maintaining their physical well-being
  • have bipolar depression.

Although existing evidence supports a possible benefit with TMS for bipolar depression (used in combination with a mood stabilizer), the lack of a definitive trial (precluding FDA clearance for this indication) and the lack of insurance coverage both limit the routine use of TMS for this indication.15

One potential advantage of TMS over ECT is a lower cost.13 Transcranial magnetic stimulation also may make it possible to achieve similar efficacy as ECT with fewer cognitive adverse effects when used in combination with ECT to reduce the number of acute ECT treatments required or as part of a maintenance strategy after a patient experiences an acute response to ECT.13

Magnetic seizure therapy (MST) vs ECT. An experimental treatment, MST uses a TMS device capable of producing more intense magnetic fields sufficient to induce a seizure.16 The advantage of MST over ECT-induced seizures is better control of intra-cerebral current path and density, thus avoiding deeper cortical areas associated with memory (eg, hippocampus) and minimizing cognitive adverse effects. As with ECT, however, anesthesia and muscle relaxation are required. Presently, MST remains investigational.

Other potential indications

In addition to MDD, TMS is also being studied as a potential treatment for other neuropsychiatric disorders.

Continue to: Obsessive-compulsive disorder

 

 

Obsessive-compulsive disorder (OCD). A recent double-blind study that evaluated a deep TMS (DTMS) device reported a significantly better outcome based on the Yale-Brown Obsessive-Compulsive Scale score with active high-frequency (20 Hz) DTMS (n = 18) vs a sham control (n = 15).17 The initial benefit persisted up to 1 month after the end of treatment. The authors speculated that this benefit may be due to direct modulation of the anterior cingulate cortex. These results led to the first FDA clearance of a deep TMS device for treating OCD.

Cognition. Because TMS does not require a seizure to produce its antidepressant effect and does not require anesthesia, the risk of neurocognitive disruption is low. In fact, evidence suggests TMS may have beneficial cognitive effects.18

In an effort to take advantage of this benefit, researchers have explored providing psychoeducation and psychotherapy sessions (eg, behavioral activation) during TMS treatments (“online”).19,20 The rationale is that neurocircuitry subserving various cognitive functions may be in a heightened state of receptivity during a TMS treatment, which would allow patients to assimilate and better utilize the therapeutic information provided.19,20

Researchers are also looking at the use of TMS to treat patients with mild cognitive impairment or early dementia. These patients often experience comorbid depression, and TMS could potentially improve memory via both its pro-cognitive and antidepressant effects.1 The lack of effective treatments for dementia supports pursuing TMS as a therapeutic option for these patients.

Other neuropsychiatric disorders. In addition to early-onset cognitive problems, other neurologic indications with promising data for TMS include chronic pain syndromes, Parkinson’s disease, tinnitus, and migraine headaches (a hand-held FDA-cleared device is now available for treating migraines). In addition to OCD and bipolar depression, other psychiatric indications with promising data include schizophrenia (eg, refractory auditory hallucinations, negative symptoms), posttraumatic stress disorder, and various addictive disorders.21 Because results have been mixed for most of these disorders, definitive trials are needed to clearly characterize the potential role of TMS.

Continue to: An ongoing evolution

 

 

An ongoing evolution

Neuromodulation is undergoing a renaissance spurred on by the need for more effective treatments to manage some of our most challenging illnesses. Transcranial magnetic stimulation and other forms of therapeutic neuromodulation are welcome additions for managing treatment-resistant depression, OCD, and possibly other disorders. But perhaps their greatest value is as a bellwether for what’s to come. In addition to the ongoing refinements to existing neuro­modulation devices, newer modulation approaches (eg, temporal interference stimulation) and the search for reliable biomarkers may dramatically expand and enhance our clinical options.14,22

Bottom Line

Transcranial magnetic stimulation (TMS) continues to evolve as a nonpharmacologic treatment for mood disorders, obsessive-compulsive disorder, and potentially for other indications. Recent developments, including altered treatment parameters, new devices, and strategies for increasing the durability of antidepressant effects, have enhanced the benefits of TMS.

Related Resources

  • Ziemann U. Thirty years of transcranial magnetic stimulation: where do we stand? Exp Brain Res. 2017;235(4):973-984.
  • Janicak PG, Sackett V, Kudrna K, et al. Transcranial magnetic stimulation for the treatment of major depression: an update on recent advances. Current Psychiatry. 2016:15(6):49-56.

[embed:render:related:node:206992]

References

1. Koch G, Bonnì S, Pellicciari MC, et al. Transcranial magnetic stimulation of the precuneus enhances memory and neural activity in prodromal Alzheimer’s disease. Neuroimage. 2018;169: 302-310.
2. O’Reardon JP, Solvason B, Janicak PG, et al. Efficacy and safety of repetitive transcranial magnetic stimulation (rTMS) in the acute treatment of major depression: results of a multicenter randomized controlled trial. Biol Psychiatry. 2007;62(11):1208-1216.
3. Dunner DL, Aaronson ST, Sackheim HA, et al. A multisite, observational study of transcranial magnetic stimulation for patients with pharmacoresistant major depressive disorder: durability of benefit over a one-year follow-up period. J Clin Psychiatry. 2014;75(12):1394-1401.
4. Janicak PG, O’Reardon JP, Sampson SM, et al. Transcranial magnetic stimulation in the treatment of major depressive disorder: a comprehensive summary of safety experience from acute exposure, extended exposure, and during reintroduction treatment. J Clin Psychiatry. 2008;69:222-232.
5. Janicak PG, Nahas Z, Lisanby SH, et al. Durability of clinical benefit with transcranial magnetic stimulation (TMS) in the treatment of pharmacoresistant major depression: assessment of relapse during a 6-month, multisite, open-label study. Brain Stimul. 2010;3(4):187-199.
6. Janicak PG. Risk management issues in transcranial magnetic stimulation for treatment of major depression. In: Bermudes R, Lanocha K, Janicak PG (eds). Transcranial magnetic stimulation: clinical applications for psychiatric practice. Washington, DC: American Psychiatric Association Publishing; 2018.
7. Chen J, Zhou C, Wu B, et al. Left versus right repetitive transcranial magnetic stimulation in treating major depression: a meta-analysis of randomised controlled trials. Psychiatry Res. 2013;210(3):1260-1264.
8. 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(10131):1683-1692.
9. Chung SW, Hoy KE, Fitzgerald PB. Theta-burst stimulation: a new form of TMS treatment for depression? Depress Anxiety. 2015;32(3):182-192.
10. Philip NS, Dunner DL, Dowd SM, et al. Can medication free, treatment-resistant, depressed patients who initially respond to TMS be maintained off medications? A prospective, 12-month multisite randomized pilot study. Brain Stimul. 2016;9(2):251-257.
11. Ren J, Li H, Palaniyappan L, et al. Repetitive transcranial magnetic stimulation versus electroconvulsive therapy for major depression: a systematic review and meta-analysis. Prop Neuropsychopharmacol Biol Psychiatry. 2014;51:181-189.
12. Janicak PG, Dowd SM, Martis B, et al. Repetitive transcranial magnetic stimulation versus electroconvulsive therapy for major depressive: preliminary results of a randomized trial. Biol Psychiatry. 2002;51(8):659-667.
13. Lanocha K, Janicak PG. TMS for depression: relationship to ECT and other therapeutic neuromodulation approaches. In: Bermudes RA, Lanocha KI, Janicak PG (eds). Transcranial magnetic stimulation: clinical applications for psychiatric practice. Washington, DC: American Psychiatric Association Publishing; 2018.
14. Drysdale AT, Grosenick L, Downar J, et al. Resting-state connectivity biomarkers define neurophysiological subtypes of depression. Nat Med. 2017;23(1):28-38.
15. Aaronson ST, Croarkin PE. Transcranial magnetic stimulation for the treatment of other mood disorders. In: Bermudes R, Lanocha K, Janicak PG (eds). Transcranial magnetic stimulation: clinical applications for psychiatric practice. Washington, DC: American Psychiatric Association Publishing; 2018.
16. Cretaz E, Brunoni AR, Lafer B. Magnetic seizure therapy for unipolar and bipolar depression: a systematic review. Neural Plast. 2015;2015:521398. doi: 10.1155/2015/521398.
17. Carmi L, Alyagon U, Barnea-Ygael N, et al. Clinical and electrophysiological outcomes of deep TMS over the medial prefrontal and anterior cingulate cortices in OCD patients. Brain Stimul. 2018;11(1):158-165.
18. Martis B, Alam D, Dowd SM, et al. Neurocognitive effects of repetitive transcranial magnetic stimulation in severe major depression. Clin Neurophysiol. 2003;114:1125-1132.
19. Donse L, Padberg F, Sack AT, et al. Simultaneous rTMS and psychotherapy in major depressive disorder: Clinical outcomes and predictors from a large naturalistic study. Brain Stimul. 2018;11(2):337-345.
20. Russo GB, Tirrell E, Busch A, et al. Behavioral activation therapy during transcranial magnetic stimulation for major depressive disorder. J Affect Disord. 2018;236:101-104.
21. Pannu J, DE Souza DD, Samara Z, et al. Transcranial magnetic stimulation for disorders other than depression. In: Bermudes RA, Lanocha KI, Janicak PG (eds). Transcranial magnetic stimulation: clinical applications for psychiatric practice. Washington, DC: American Psychiatric Association Publishing; 2018.
22. Grossman N. Modulation without surgical intervention. Science. 2018;361:461-462.

Article PDF
Author and Disclosure Information

Philip G. Janicak, MD
Adjunct Professor Department of Psychiatry and Behavioral Sciences
Northwestern University Feinberg School of Medicine
Chicago, Illinois
Member, Current Psychiatry Editorial Board

Disclosure
Dr. Janicak is an unpaid consultant to Neuronetics, Inc., and has a financial relationship with Otsuka Pharmaceuticals.

Issue
Current Psychiatry - 18(3)
Publications
Topics
Page Number
10-16
Sections
Author and Disclosure Information

Philip G. Janicak, MD
Adjunct Professor Department of Psychiatry and Behavioral Sciences
Northwestern University Feinberg School of Medicine
Chicago, Illinois
Member, Current Psychiatry Editorial Board

Disclosure
Dr. Janicak is an unpaid consultant to Neuronetics, Inc., and has a financial relationship with Otsuka Pharmaceuticals.

Author and Disclosure Information

Philip G. Janicak, MD
Adjunct Professor Department of Psychiatry and Behavioral Sciences
Northwestern University Feinberg School of Medicine
Chicago, Illinois
Member, Current Psychiatry Editorial Board

Disclosure
Dr. Janicak is an unpaid consultant to Neuronetics, Inc., and has a financial relationship with Otsuka Pharmaceuticals.

Article PDF
Article PDF

Therapeutic neuromodulation takes advantage of the brain’s electrochemical makeup. This allows for treatment devices that modulate neurocircuits relevant to behaviors disrupted in disorders such as major depressive disorder (MDD) (eg, sleep quality, appetite, cognitive, and executive functions). The default mode network (comprised of structures such as the medial prefrontal cortex [MPFC], the posterior cingulate cortex, the hippocampus, and their functional connectivity) serves as a prime example of circuitry that can be targeted by this approach.1

For 80 years, electroconvulsive therapy (ECT) has been an important neuromodulation option for patients with more severe illness. Recently, additional neuromodulatory approaches have been FDA-cleared, including transcranial magnetic stimulation (TMS), vagus nerve stimulation (VNS), and deep brain stimulation (DBS). Another approach, transcranial direct current stimulation (tDCS), has been extensively studied for its potential clinical utility but is not FDA-cleared. The Table provides descriptions of these therapies.

CP01803010_t1.PNG


Since being cleared by the FDA in 2008, TMS has arguably made the greatest strides in providing an alternate neuromodulation treatment option for patients with MDD, with >1,000 centers nationally and 7 TMS devices FDA-cleared for treatment of depression. In this article, we review recent developments in TMS.

 

An evolving therapeutic option

While primarily studied as a monotherapy for MDD, in clinical practice TMS (Box) is typically used as an adjunct to medication and psychotherapy.2,3 In this context, it has demonstrated efficacy for more difficult-to-treat mood disorders with an excellent safety and tolerability profile whether used with or without medication.4-6

To further improve the efficiency and efficacy of TMS while maintaining its safety and tolerability, researchers and clinicians have been exploring a few initiatives.

Box

Key points: Transcranial magnetic stimulation
  • Transcranial magnetic stimulation (TMS) utilizes intense, localized magnetic fields to alter activity in neural circuits implicated in the pathophysiology of depression
  • Randomized, sham-controlled acute trials have demonstrated the efficacy of TMS for treatment-resistant depression
  • Clinical availability of TMS has grown steadily over the past 10 years as >1,000 centers have been opened and additional devices have been FDA-cleared
  • TMS has the potential to avoid safety and tolerability concerns associated with antidepressant pharmacotherapy (eg, weight gain, sexual dysfunction) and electroconvulsive therapy (eg, cognitive deficits)
  • Greater sophistication in the choice of stimulation parameters, as well as other ongoing efforts to optimize the benefits of TMS, are yielding better clinical outcomes

Altered treatment parameters

One initiative is assessing the feasibility of altering various treatment parameters, such as the total number of treatment sessions (30 to 60 sessions); the frequency of sessions (eg, more than once daily); the total number of magnetic pulses per session (eg, >3,000); the stimulation coil localization (eg, left vs right dorsal lateral prefrontal cortex [DLPFC]; MPFC; and various methods to determine optimal coil placement (eg, EEG F3 coordinate or MRI-guided neuro-navigational methods). Such refinements offer the potential for enhanced efficacy, shorter treatment sessions, and/or improved tolerability. For example, lower frequency right DLPFC stimulations (eg, 1 Hz) can decrease the risk of seizures and improve overall tolerability. While this has not been studied as extensively as higher frequency left DLPFC stimulations (eg, 5 to 20 Hz), existing evidence supports similar efficacy between these 2 approaches.7

Theta burst stimulation. Some TMS devices can be adapted to deliver theta burst stimulation (TBS). This produces trains of triple, 50 Hz, pulsed bursts (usually with 200 ms inter-burst intervals occurring at a rate of 5 Hz; at 80% MT) to model naturally occurring theta rhythms. These bursts can be administered in stimulation protocols using intermittent TBS (iTBS) (eg, 10 bursts of triplets over 2 seconds every 10 seconds; 30 pulses per burst; for approximately 3 minutes; totaling 600 pulses) or continuous TBS (cTBS) bursts given in an uninterrupted train (eg, 40 seconds, 600 pulses). Evidence indicates these protocols facilitate long-term potentiation (ie, iTBS) and long-term depression (ie, cTBS), which in turn can modulate synaptic plasticity.

Continue to: While some clinicians are using...

 

 

While some clinicians are using TBS off-label, a recent non-inferiority trial (N = 395) reported similar efficacy and safety comparing standard 10 Hz TMS to an iTBS protocol at 120% of resting motor threshold (both over the left DLPFC).8 This has led to FDA clearance of the TMS device adapted to provide iTBS in this trial.8

From a more practical perspective, TBS has the potential to reduce the number of pulses (eg, 600 vs 3,000) and the total number of sessions required, as well as the duration of treatment sessions (eg, 37.5 minutes to <5 minutes). This can accelerate the time to response and decrease patient and staff commitment, with resulting cost savings.9 Despite this recent progress, ongoing research still needs to clarify issues such as the risk/benefit profile, particularly in younger and older populations, as well as assessment of duration of initial benefit and appropriate maintenance strategies.

New devices

Another initiative is the development of alternative TMS equipment. For example, newer coil designs with enhanced cooling ability allow for a substantial decrease in the required inter-train interval duration between stimulation trains, thus shortening the total session duration by approximately 50% (eg, from 37.5 to 19 minutes). The use of different coil arrays (eg, the H-coil capable of deeper vs surface stimulation) may allow for more direct stimulation of relevant neurocircuitry (eg, cingulate cortex), possibly improving efficacy and shortening time to onset of benefit. However, in head-to-head comparisons with single-coil devices, enhanced efficacy for depression has not been clearly demonstrated. One caveat is that the increase in depth of magnetic field penetration results in a loss of focality, resulting in the stimulation of larger brain areas. This might increase the risk of adverse effects such as seizures.

 

Increasing durability of effect

Because high relapse and recurrence rates compromise the initial benefit of any antidepressant therapy, appropriate maintenance strategies are essential. Several studies have evaluated strategies to maintain the acute benefit of TMS for treatment-resistant depression.

One was a 6-month, open-label TMS durability of effect trial for acute responders (n = 99) in the pivotal registration study.5 During this study, all participants were given antidepressant medication monotherapy. In addition, with early indication of relapse, patients received a reintroduction of TMS sessions (32/99 patients; mean number of sessions = 14.3). With this protocol, approximately 84% re-achieved their response status. The overall relapse rate was approximately 13%.5

Continue to: In a 1-year naturalistic study...

 

 

In a 1-year naturalistic study, 63% of patients (75/120) who met response or remission criteria after an acute course of TMS still met response criteria after 12 months. These patients received clinician-determined maintenance treatment that included reintroduction of TMS when indicated.3

In a prospective, 12-month, multisite, randomized pilot study, 67 patients with treatment-resistant MDD underwent an antidepressant medication washout and then received 30 sessions of TMS monotherapy.10 Those who met criteria for improvement (n = 49) were then randomized to once-monthly TMS or observation only. All patients remained medication-free but could receive TMS re-introduction if they deteriorated. At the end of the study, both groups demonstrated comparable outcomes, with a trend to a longer time before relapse among participants who received once-monthly TMS. Although these results are preliminary, they suggest that some patients could be treated both acutely and then maintained with TMS alone.

Re-introducing TMS in patients who show early signs of relapse after having an initial response achieves rates of sustained improvement that compare favorably with those of other strategies used to manage patients with treatment-resistant depression.

 

TMS vs ECT

The question often arises as to whether TMS is a viable alternate treatment to ECT. I believe the answer is unequivocally yes and no. By this, I mean some patients who in the past only had ECT as their next option when medications and psychotherapy were insufficient may now consider TMS. In support, there is evidence of comparable efficacy between TMS and ECT in a subgroup of patients who were considered clinically appropriate for ECT.11-13

How to best identify this group remains unclear, but investigators are exploring predictive biomarkers. For example, a large study (N = 1,188), with functional magnetic resonance imaging (fMRI) reported that depressed patients could be divided into 4 neurophysiological “biotypes” based on different patterns of aberrant connectivity in limbic and fronto-striatal networks.14 The authors further noted that such distinctions were helpful in predicting response in a subgroup of patients (n = 154) who received TMS.

Continue to: For now...

 

 

For now, experience indicates certain clinical factors may provide some guidance. Patients are usually better served by ECT if they:

  • have depressive episodes of longer duration (eg, >3 years)
  • have a high risk of suicide
  • have psychotic or catatonic features associated with their depression
  • have difficulty maintaining their physical well-being
  • have bipolar depression.

Although existing evidence supports a possible benefit with TMS for bipolar depression (used in combination with a mood stabilizer), the lack of a definitive trial (precluding FDA clearance for this indication) and the lack of insurance coverage both limit the routine use of TMS for this indication.15

One potential advantage of TMS over ECT is a lower cost.13 Transcranial magnetic stimulation also may make it possible to achieve similar efficacy as ECT with fewer cognitive adverse effects when used in combination with ECT to reduce the number of acute ECT treatments required or as part of a maintenance strategy after a patient experiences an acute response to ECT.13

Magnetic seizure therapy (MST) vs ECT. An experimental treatment, MST uses a TMS device capable of producing more intense magnetic fields sufficient to induce a seizure.16 The advantage of MST over ECT-induced seizures is better control of intra-cerebral current path and density, thus avoiding deeper cortical areas associated with memory (eg, hippocampus) and minimizing cognitive adverse effects. As with ECT, however, anesthesia and muscle relaxation are required. Presently, MST remains investigational.

Other potential indications

In addition to MDD, TMS is also being studied as a potential treatment for other neuropsychiatric disorders.

Continue to: Obsessive-compulsive disorder

 

 

Obsessive-compulsive disorder (OCD). A recent double-blind study that evaluated a deep TMS (DTMS) device reported a significantly better outcome based on the Yale-Brown Obsessive-Compulsive Scale score with active high-frequency (20 Hz) DTMS (n = 18) vs a sham control (n = 15).17 The initial benefit persisted up to 1 month after the end of treatment. The authors speculated that this benefit may be due to direct modulation of the anterior cingulate cortex. These results led to the first FDA clearance of a deep TMS device for treating OCD.

Cognition. Because TMS does not require a seizure to produce its antidepressant effect and does not require anesthesia, the risk of neurocognitive disruption is low. In fact, evidence suggests TMS may have beneficial cognitive effects.18

In an effort to take advantage of this benefit, researchers have explored providing psychoeducation and psychotherapy sessions (eg, behavioral activation) during TMS treatments (“online”).19,20 The rationale is that neurocircuitry subserving various cognitive functions may be in a heightened state of receptivity during a TMS treatment, which would allow patients to assimilate and better utilize the therapeutic information provided.19,20

Researchers are also looking at the use of TMS to treat patients with mild cognitive impairment or early dementia. These patients often experience comorbid depression, and TMS could potentially improve memory via both its pro-cognitive and antidepressant effects.1 The lack of effective treatments for dementia supports pursuing TMS as a therapeutic option for these patients.

Other neuropsychiatric disorders. In addition to early-onset cognitive problems, other neurologic indications with promising data for TMS include chronic pain syndromes, Parkinson’s disease, tinnitus, and migraine headaches (a hand-held FDA-cleared device is now available for treating migraines). In addition to OCD and bipolar depression, other psychiatric indications with promising data include schizophrenia (eg, refractory auditory hallucinations, negative symptoms), posttraumatic stress disorder, and various addictive disorders.21 Because results have been mixed for most of these disorders, definitive trials are needed to clearly characterize the potential role of TMS.

Continue to: An ongoing evolution

 

 

An ongoing evolution

Neuromodulation is undergoing a renaissance spurred on by the need for more effective treatments to manage some of our most challenging illnesses. Transcranial magnetic stimulation and other forms of therapeutic neuromodulation are welcome additions for managing treatment-resistant depression, OCD, and possibly other disorders. But perhaps their greatest value is as a bellwether for what’s to come. In addition to the ongoing refinements to existing neuro­modulation devices, newer modulation approaches (eg, temporal interference stimulation) and the search for reliable biomarkers may dramatically expand and enhance our clinical options.14,22

Bottom Line

Transcranial magnetic stimulation (TMS) continues to evolve as a nonpharmacologic treatment for mood disorders, obsessive-compulsive disorder, and potentially for other indications. Recent developments, including altered treatment parameters, new devices, and strategies for increasing the durability of antidepressant effects, have enhanced the benefits of TMS.

Related Resources

  • Ziemann U. Thirty years of transcranial magnetic stimulation: where do we stand? Exp Brain Res. 2017;235(4):973-984.
  • Janicak PG, Sackett V, Kudrna K, et al. Transcranial magnetic stimulation for the treatment of major depression: an update on recent advances. Current Psychiatry. 2016:15(6):49-56.

[embed:render:related:node:206992]

Therapeutic neuromodulation takes advantage of the brain’s electrochemical makeup. This allows for treatment devices that modulate neurocircuits relevant to behaviors disrupted in disorders such as major depressive disorder (MDD) (eg, sleep quality, appetite, cognitive, and executive functions). The default mode network (comprised of structures such as the medial prefrontal cortex [MPFC], the posterior cingulate cortex, the hippocampus, and their functional connectivity) serves as a prime example of circuitry that can be targeted by this approach.1

For 80 years, electroconvulsive therapy (ECT) has been an important neuromodulation option for patients with more severe illness. Recently, additional neuromodulatory approaches have been FDA-cleared, including transcranial magnetic stimulation (TMS), vagus nerve stimulation (VNS), and deep brain stimulation (DBS). Another approach, transcranial direct current stimulation (tDCS), has been extensively studied for its potential clinical utility but is not FDA-cleared. The Table provides descriptions of these therapies.

CP01803010_t1.PNG


Since being cleared by the FDA in 2008, TMS has arguably made the greatest strides in providing an alternate neuromodulation treatment option for patients with MDD, with >1,000 centers nationally and 7 TMS devices FDA-cleared for treatment of depression. In this article, we review recent developments in TMS.

 

An evolving therapeutic option

While primarily studied as a monotherapy for MDD, in clinical practice TMS (Box) is typically used as an adjunct to medication and psychotherapy.2,3 In this context, it has demonstrated efficacy for more difficult-to-treat mood disorders with an excellent safety and tolerability profile whether used with or without medication.4-6

To further improve the efficiency and efficacy of TMS while maintaining its safety and tolerability, researchers and clinicians have been exploring a few initiatives.

Box

Key points: Transcranial magnetic stimulation
  • Transcranial magnetic stimulation (TMS) utilizes intense, localized magnetic fields to alter activity in neural circuits implicated in the pathophysiology of depression
  • Randomized, sham-controlled acute trials have demonstrated the efficacy of TMS for treatment-resistant depression
  • Clinical availability of TMS has grown steadily over the past 10 years as >1,000 centers have been opened and additional devices have been FDA-cleared
  • TMS has the potential to avoid safety and tolerability concerns associated with antidepressant pharmacotherapy (eg, weight gain, sexual dysfunction) and electroconvulsive therapy (eg, cognitive deficits)
  • Greater sophistication in the choice of stimulation parameters, as well as other ongoing efforts to optimize the benefits of TMS, are yielding better clinical outcomes

Altered treatment parameters

One initiative is assessing the feasibility of altering various treatment parameters, such as the total number of treatment sessions (30 to 60 sessions); the frequency of sessions (eg, more than once daily); the total number of magnetic pulses per session (eg, >3,000); the stimulation coil localization (eg, left vs right dorsal lateral prefrontal cortex [DLPFC]; MPFC; and various methods to determine optimal coil placement (eg, EEG F3 coordinate or MRI-guided neuro-navigational methods). Such refinements offer the potential for enhanced efficacy, shorter treatment sessions, and/or improved tolerability. For example, lower frequency right DLPFC stimulations (eg, 1 Hz) can decrease the risk of seizures and improve overall tolerability. While this has not been studied as extensively as higher frequency left DLPFC stimulations (eg, 5 to 20 Hz), existing evidence supports similar efficacy between these 2 approaches.7

Theta burst stimulation. Some TMS devices can be adapted to deliver theta burst stimulation (TBS). This produces trains of triple, 50 Hz, pulsed bursts (usually with 200 ms inter-burst intervals occurring at a rate of 5 Hz; at 80% MT) to model naturally occurring theta rhythms. These bursts can be administered in stimulation protocols using intermittent TBS (iTBS) (eg, 10 bursts of triplets over 2 seconds every 10 seconds; 30 pulses per burst; for approximately 3 minutes; totaling 600 pulses) or continuous TBS (cTBS) bursts given in an uninterrupted train (eg, 40 seconds, 600 pulses). Evidence indicates these protocols facilitate long-term potentiation (ie, iTBS) and long-term depression (ie, cTBS), which in turn can modulate synaptic plasticity.

Continue to: While some clinicians are using...

 

 

While some clinicians are using TBS off-label, a recent non-inferiority trial (N = 395) reported similar efficacy and safety comparing standard 10 Hz TMS to an iTBS protocol at 120% of resting motor threshold (both over the left DLPFC).8 This has led to FDA clearance of the TMS device adapted to provide iTBS in this trial.8

From a more practical perspective, TBS has the potential to reduce the number of pulses (eg, 600 vs 3,000) and the total number of sessions required, as well as the duration of treatment sessions (eg, 37.5 minutes to <5 minutes). This can accelerate the time to response and decrease patient and staff commitment, with resulting cost savings.9 Despite this recent progress, ongoing research still needs to clarify issues such as the risk/benefit profile, particularly in younger and older populations, as well as assessment of duration of initial benefit and appropriate maintenance strategies.

New devices

Another initiative is the development of alternative TMS equipment. For example, newer coil designs with enhanced cooling ability allow for a substantial decrease in the required inter-train interval duration between stimulation trains, thus shortening the total session duration by approximately 50% (eg, from 37.5 to 19 minutes). The use of different coil arrays (eg, the H-coil capable of deeper vs surface stimulation) may allow for more direct stimulation of relevant neurocircuitry (eg, cingulate cortex), possibly improving efficacy and shortening time to onset of benefit. However, in head-to-head comparisons with single-coil devices, enhanced efficacy for depression has not been clearly demonstrated. One caveat is that the increase in depth of magnetic field penetration results in a loss of focality, resulting in the stimulation of larger brain areas. This might increase the risk of adverse effects such as seizures.

 

Increasing durability of effect

Because high relapse and recurrence rates compromise the initial benefit of any antidepressant therapy, appropriate maintenance strategies are essential. Several studies have evaluated strategies to maintain the acute benefit of TMS for treatment-resistant depression.

One was a 6-month, open-label TMS durability of effect trial for acute responders (n = 99) in the pivotal registration study.5 During this study, all participants were given antidepressant medication monotherapy. In addition, with early indication of relapse, patients received a reintroduction of TMS sessions (32/99 patients; mean number of sessions = 14.3). With this protocol, approximately 84% re-achieved their response status. The overall relapse rate was approximately 13%.5

Continue to: In a 1-year naturalistic study...

 

 

In a 1-year naturalistic study, 63% of patients (75/120) who met response or remission criteria after an acute course of TMS still met response criteria after 12 months. These patients received clinician-determined maintenance treatment that included reintroduction of TMS when indicated.3

In a prospective, 12-month, multisite, randomized pilot study, 67 patients with treatment-resistant MDD underwent an antidepressant medication washout and then received 30 sessions of TMS monotherapy.10 Those who met criteria for improvement (n = 49) were then randomized to once-monthly TMS or observation only. All patients remained medication-free but could receive TMS re-introduction if they deteriorated. At the end of the study, both groups demonstrated comparable outcomes, with a trend to a longer time before relapse among participants who received once-monthly TMS. Although these results are preliminary, they suggest that some patients could be treated both acutely and then maintained with TMS alone.

Re-introducing TMS in patients who show early signs of relapse after having an initial response achieves rates of sustained improvement that compare favorably with those of other strategies used to manage patients with treatment-resistant depression.

 

TMS vs ECT

The question often arises as to whether TMS is a viable alternate treatment to ECT. I believe the answer is unequivocally yes and no. By this, I mean some patients who in the past only had ECT as their next option when medications and psychotherapy were insufficient may now consider TMS. In support, there is evidence of comparable efficacy between TMS and ECT in a subgroup of patients who were considered clinically appropriate for ECT.11-13

How to best identify this group remains unclear, but investigators are exploring predictive biomarkers. For example, a large study (N = 1,188), with functional magnetic resonance imaging (fMRI) reported that depressed patients could be divided into 4 neurophysiological “biotypes” based on different patterns of aberrant connectivity in limbic and fronto-striatal networks.14 The authors further noted that such distinctions were helpful in predicting response in a subgroup of patients (n = 154) who received TMS.

Continue to: For now...

 

 

For now, experience indicates certain clinical factors may provide some guidance. Patients are usually better served by ECT if they:

  • have depressive episodes of longer duration (eg, >3 years)
  • have a high risk of suicide
  • have psychotic or catatonic features associated with their depression
  • have difficulty maintaining their physical well-being
  • have bipolar depression.

Although existing evidence supports a possible benefit with TMS for bipolar depression (used in combination with a mood stabilizer), the lack of a definitive trial (precluding FDA clearance for this indication) and the lack of insurance coverage both limit the routine use of TMS for this indication.15

One potential advantage of TMS over ECT is a lower cost.13 Transcranial magnetic stimulation also may make it possible to achieve similar efficacy as ECT with fewer cognitive adverse effects when used in combination with ECT to reduce the number of acute ECT treatments required or as part of a maintenance strategy after a patient experiences an acute response to ECT.13

Magnetic seizure therapy (MST) vs ECT. An experimental treatment, MST uses a TMS device capable of producing more intense magnetic fields sufficient to induce a seizure.16 The advantage of MST over ECT-induced seizures is better control of intra-cerebral current path and density, thus avoiding deeper cortical areas associated with memory (eg, hippocampus) and minimizing cognitive adverse effects. As with ECT, however, anesthesia and muscle relaxation are required. Presently, MST remains investigational.

Other potential indications

In addition to MDD, TMS is also being studied as a potential treatment for other neuropsychiatric disorders.

Continue to: Obsessive-compulsive disorder

 

 

Obsessive-compulsive disorder (OCD). A recent double-blind study that evaluated a deep TMS (DTMS) device reported a significantly better outcome based on the Yale-Brown Obsessive-Compulsive Scale score with active high-frequency (20 Hz) DTMS (n = 18) vs a sham control (n = 15).17 The initial benefit persisted up to 1 month after the end of treatment. The authors speculated that this benefit may be due to direct modulation of the anterior cingulate cortex. These results led to the first FDA clearance of a deep TMS device for treating OCD.

Cognition. Because TMS does not require a seizure to produce its antidepressant effect and does not require anesthesia, the risk of neurocognitive disruption is low. In fact, evidence suggests TMS may have beneficial cognitive effects.18

In an effort to take advantage of this benefit, researchers have explored providing psychoeducation and psychotherapy sessions (eg, behavioral activation) during TMS treatments (“online”).19,20 The rationale is that neurocircuitry subserving various cognitive functions may be in a heightened state of receptivity during a TMS treatment, which would allow patients to assimilate and better utilize the therapeutic information provided.19,20

Researchers are also looking at the use of TMS to treat patients with mild cognitive impairment or early dementia. These patients often experience comorbid depression, and TMS could potentially improve memory via both its pro-cognitive and antidepressant effects.1 The lack of effective treatments for dementia supports pursuing TMS as a therapeutic option for these patients.

Other neuropsychiatric disorders. In addition to early-onset cognitive problems, other neurologic indications with promising data for TMS include chronic pain syndromes, Parkinson’s disease, tinnitus, and migraine headaches (a hand-held FDA-cleared device is now available for treating migraines). In addition to OCD and bipolar depression, other psychiatric indications with promising data include schizophrenia (eg, refractory auditory hallucinations, negative symptoms), posttraumatic stress disorder, and various addictive disorders.21 Because results have been mixed for most of these disorders, definitive trials are needed to clearly characterize the potential role of TMS.

Continue to: An ongoing evolution

 

 

An ongoing evolution

Neuromodulation is undergoing a renaissance spurred on by the need for more effective treatments to manage some of our most challenging illnesses. Transcranial magnetic stimulation and other forms of therapeutic neuromodulation are welcome additions for managing treatment-resistant depression, OCD, and possibly other disorders. But perhaps their greatest value is as a bellwether for what’s to come. In addition to the ongoing refinements to existing neuro­modulation devices, newer modulation approaches (eg, temporal interference stimulation) and the search for reliable biomarkers may dramatically expand and enhance our clinical options.14,22

Bottom Line

Transcranial magnetic stimulation (TMS) continues to evolve as a nonpharmacologic treatment for mood disorders, obsessive-compulsive disorder, and potentially for other indications. Recent developments, including altered treatment parameters, new devices, and strategies for increasing the durability of antidepressant effects, have enhanced the benefits of TMS.

Related Resources

  • Ziemann U. Thirty years of transcranial magnetic stimulation: where do we stand? Exp Brain Res. 2017;235(4):973-984.
  • Janicak PG, Sackett V, Kudrna K, et al. Transcranial magnetic stimulation for the treatment of major depression: an update on recent advances. Current Psychiatry. 2016:15(6):49-56.

[embed:render:related:node:206992]

References

1. Koch G, Bonnì S, Pellicciari MC, et al. Transcranial magnetic stimulation of the precuneus enhances memory and neural activity in prodromal Alzheimer’s disease. Neuroimage. 2018;169: 302-310.
2. O’Reardon JP, Solvason B, Janicak PG, et al. Efficacy and safety of repetitive transcranial magnetic stimulation (rTMS) in the acute treatment of major depression: results of a multicenter randomized controlled trial. Biol Psychiatry. 2007;62(11):1208-1216.
3. Dunner DL, Aaronson ST, Sackheim HA, et al. A multisite, observational study of transcranial magnetic stimulation for patients with pharmacoresistant major depressive disorder: durability of benefit over a one-year follow-up period. J Clin Psychiatry. 2014;75(12):1394-1401.
4. Janicak PG, O’Reardon JP, Sampson SM, et al. Transcranial magnetic stimulation in the treatment of major depressive disorder: a comprehensive summary of safety experience from acute exposure, extended exposure, and during reintroduction treatment. J Clin Psychiatry. 2008;69:222-232.
5. Janicak PG, Nahas Z, Lisanby SH, et al. Durability of clinical benefit with transcranial magnetic stimulation (TMS) in the treatment of pharmacoresistant major depression: assessment of relapse during a 6-month, multisite, open-label study. Brain Stimul. 2010;3(4):187-199.
6. Janicak PG. Risk management issues in transcranial magnetic stimulation for treatment of major depression. In: Bermudes R, Lanocha K, Janicak PG (eds). Transcranial magnetic stimulation: clinical applications for psychiatric practice. Washington, DC: American Psychiatric Association Publishing; 2018.
7. Chen J, Zhou C, Wu B, et al. Left versus right repetitive transcranial magnetic stimulation in treating major depression: a meta-analysis of randomised controlled trials. Psychiatry Res. 2013;210(3):1260-1264.
8. 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(10131):1683-1692.
9. Chung SW, Hoy KE, Fitzgerald PB. Theta-burst stimulation: a new form of TMS treatment for depression? Depress Anxiety. 2015;32(3):182-192.
10. Philip NS, Dunner DL, Dowd SM, et al. Can medication free, treatment-resistant, depressed patients who initially respond to TMS be maintained off medications? A prospective, 12-month multisite randomized pilot study. Brain Stimul. 2016;9(2):251-257.
11. Ren J, Li H, Palaniyappan L, et al. Repetitive transcranial magnetic stimulation versus electroconvulsive therapy for major depression: a systematic review and meta-analysis. Prop Neuropsychopharmacol Biol Psychiatry. 2014;51:181-189.
12. Janicak PG, Dowd SM, Martis B, et al. Repetitive transcranial magnetic stimulation versus electroconvulsive therapy for major depressive: preliminary results of a randomized trial. Biol Psychiatry. 2002;51(8):659-667.
13. Lanocha K, Janicak PG. TMS for depression: relationship to ECT and other therapeutic neuromodulation approaches. In: Bermudes RA, Lanocha KI, Janicak PG (eds). Transcranial magnetic stimulation: clinical applications for psychiatric practice. Washington, DC: American Psychiatric Association Publishing; 2018.
14. Drysdale AT, Grosenick L, Downar J, et al. Resting-state connectivity biomarkers define neurophysiological subtypes of depression. Nat Med. 2017;23(1):28-38.
15. Aaronson ST, Croarkin PE. Transcranial magnetic stimulation for the treatment of other mood disorders. In: Bermudes R, Lanocha K, Janicak PG (eds). Transcranial magnetic stimulation: clinical applications for psychiatric practice. Washington, DC: American Psychiatric Association Publishing; 2018.
16. Cretaz E, Brunoni AR, Lafer B. Magnetic seizure therapy for unipolar and bipolar depression: a systematic review. Neural Plast. 2015;2015:521398. doi: 10.1155/2015/521398.
17. Carmi L, Alyagon U, Barnea-Ygael N, et al. Clinical and electrophysiological outcomes of deep TMS over the medial prefrontal and anterior cingulate cortices in OCD patients. Brain Stimul. 2018;11(1):158-165.
18. Martis B, Alam D, Dowd SM, et al. Neurocognitive effects of repetitive transcranial magnetic stimulation in severe major depression. Clin Neurophysiol. 2003;114:1125-1132.
19. Donse L, Padberg F, Sack AT, et al. Simultaneous rTMS and psychotherapy in major depressive disorder: Clinical outcomes and predictors from a large naturalistic study. Brain Stimul. 2018;11(2):337-345.
20. Russo GB, Tirrell E, Busch A, et al. Behavioral activation therapy during transcranial magnetic stimulation for major depressive disorder. J Affect Disord. 2018;236:101-104.
21. Pannu J, DE Souza DD, Samara Z, et al. Transcranial magnetic stimulation for disorders other than depression. In: Bermudes RA, Lanocha KI, Janicak PG (eds). Transcranial magnetic stimulation: clinical applications for psychiatric practice. Washington, DC: American Psychiatric Association Publishing; 2018.
22. Grossman N. Modulation without surgical intervention. Science. 2018;361:461-462.

References

1. Koch G, Bonnì S, Pellicciari MC, et al. Transcranial magnetic stimulation of the precuneus enhances memory and neural activity in prodromal Alzheimer’s disease. Neuroimage. 2018;169: 302-310.
2. O’Reardon JP, Solvason B, Janicak PG, et al. Efficacy and safety of repetitive transcranial magnetic stimulation (rTMS) in the acute treatment of major depression: results of a multicenter randomized controlled trial. Biol Psychiatry. 2007;62(11):1208-1216.
3. Dunner DL, Aaronson ST, Sackheim HA, et al. A multisite, observational study of transcranial magnetic stimulation for patients with pharmacoresistant major depressive disorder: durability of benefit over a one-year follow-up period. J Clin Psychiatry. 2014;75(12):1394-1401.
4. Janicak PG, O’Reardon JP, Sampson SM, et al. Transcranial magnetic stimulation in the treatment of major depressive disorder: a comprehensive summary of safety experience from acute exposure, extended exposure, and during reintroduction treatment. J Clin Psychiatry. 2008;69:222-232.
5. Janicak PG, Nahas Z, Lisanby SH, et al. Durability of clinical benefit with transcranial magnetic stimulation (TMS) in the treatment of pharmacoresistant major depression: assessment of relapse during a 6-month, multisite, open-label study. Brain Stimul. 2010;3(4):187-199.
6. Janicak PG. Risk management issues in transcranial magnetic stimulation for treatment of major depression. In: Bermudes R, Lanocha K, Janicak PG (eds). Transcranial magnetic stimulation: clinical applications for psychiatric practice. Washington, DC: American Psychiatric Association Publishing; 2018.
7. Chen J, Zhou C, Wu B, et al. Left versus right repetitive transcranial magnetic stimulation in treating major depression: a meta-analysis of randomised controlled trials. Psychiatry Res. 2013;210(3):1260-1264.
8. 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(10131):1683-1692.
9. Chung SW, Hoy KE, Fitzgerald PB. Theta-burst stimulation: a new form of TMS treatment for depression? Depress Anxiety. 2015;32(3):182-192.
10. Philip NS, Dunner DL, Dowd SM, et al. Can medication free, treatment-resistant, depressed patients who initially respond to TMS be maintained off medications? A prospective, 12-month multisite randomized pilot study. Brain Stimul. 2016;9(2):251-257.
11. Ren J, Li H, Palaniyappan L, et al. Repetitive transcranial magnetic stimulation versus electroconvulsive therapy for major depression: a systematic review and meta-analysis. Prop Neuropsychopharmacol Biol Psychiatry. 2014;51:181-189.
12. Janicak PG, Dowd SM, Martis B, et al. Repetitive transcranial magnetic stimulation versus electroconvulsive therapy for major depressive: preliminary results of a randomized trial. Biol Psychiatry. 2002;51(8):659-667.
13. Lanocha K, Janicak PG. TMS for depression: relationship to ECT and other therapeutic neuromodulation approaches. In: Bermudes RA, Lanocha KI, Janicak PG (eds). Transcranial magnetic stimulation: clinical applications for psychiatric practice. Washington, DC: American Psychiatric Association Publishing; 2018.
14. Drysdale AT, Grosenick L, Downar J, et al. Resting-state connectivity biomarkers define neurophysiological subtypes of depression. Nat Med. 2017;23(1):28-38.
15. Aaronson ST, Croarkin PE. Transcranial magnetic stimulation for the treatment of other mood disorders. In: Bermudes R, Lanocha K, Janicak PG (eds). Transcranial magnetic stimulation: clinical applications for psychiatric practice. Washington, DC: American Psychiatric Association Publishing; 2018.
16. Cretaz E, Brunoni AR, Lafer B. Magnetic seizure therapy for unipolar and bipolar depression: a systematic review. Neural Plast. 2015;2015:521398. doi: 10.1155/2015/521398.
17. Carmi L, Alyagon U, Barnea-Ygael N, et al. Clinical and electrophysiological outcomes of deep TMS over the medial prefrontal and anterior cingulate cortices in OCD patients. Brain Stimul. 2018;11(1):158-165.
18. Martis B, Alam D, Dowd SM, et al. Neurocognitive effects of repetitive transcranial magnetic stimulation in severe major depression. Clin Neurophysiol. 2003;114:1125-1132.
19. Donse L, Padberg F, Sack AT, et al. Simultaneous rTMS and psychotherapy in major depressive disorder: Clinical outcomes and predictors from a large naturalistic study. Brain Stimul. 2018;11(2):337-345.
20. Russo GB, Tirrell E, Busch A, et al. Behavioral activation therapy during transcranial magnetic stimulation for major depressive disorder. J Affect Disord. 2018;236:101-104.
21. Pannu J, DE Souza DD, Samara Z, et al. Transcranial magnetic stimulation for disorders other than depression. In: Bermudes RA, Lanocha KI, Janicak PG (eds). Transcranial magnetic stimulation: clinical applications for psychiatric practice. Washington, DC: American Psychiatric Association Publishing; 2018.
22. Grossman N. Modulation without surgical intervention. Science. 2018;361:461-462.

Issue
Current Psychiatry - 18(3)
Issue
Current Psychiatry - 18(3)
Page Number
10-16
Page Number
10-16
Publications
Publications
Topics
Article Type
Display Headline
What’s new in transcranial magnetic stimulation
Display Headline
What’s new in transcranial magnetic stimulation
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media
Image
Teambase ID
18001256.SIG
Disable zoom
Off

Advances in transcranial magnetic stimulation for managing major depressive disorders

Article Type
Changed
Thu, 03/28/2019 - 15:07
Display Headline
Advances in transcranial magnetic stimulation for managing major depressive disorders

Since 2008, the FDA has cleared 4 transcranial magnetic stimulation (TMS) devices for treating depression (Related Resources). In that time, the availability of TMS has steadily grown within and outside the United States.

Parallel with increasing clinical utilization of this technology, research continues into the benefit of TMS for treatment-resistant depression; such research includes additional, supportive, acute, sham-controlled trials; comparison trials with electroconvulsive therapy (ECT) for more severe episodes of depression; short- and long-term real-world outcome studies; exploration of alternative treatment parameters to further enhance its efficacy; and the development of other TMS approaches. In this article, we review recent developments in the application of TMS to treat major depressive disorder—in particular, treatment-resistant depression (Box).
 

RTEmagicC_CP015060049_b1.jpg.jpg


Therapeutic neuromodulation
The underlying premise of neuromodulation is that the brain is an electrochemical organ that can be modulated by pharmacotherapy or device-based approaches, or their combination.1 ECT is the prototypic device-based neuromodulation approach, and remains one of the most effective treatments for severe depression.

More recently, however, other methods have been, and continue to be, developed to treat patients who do not achieve adequate benefit from psychotherapy or medical therapy, or both, and who might not be an ideal candidate for ECT (Table,1). In addition to the potential therapeutic benefit of these alternative strategies, some could avoid safety and tolerability concerns associated with medication (weight gain, sexual dysfunction) and ECT (eg, cognitive deficits).

RTEmagicC_CP015060049_t1.jpg.jpg


TMS, which utilizes intense, localized magnetic fields to alter activity in neural circuits implicated in the pathophysiology of depression, represents an important example of this initiative.2


TMS has established efficacy for depression
Sham-controlled trials. Several randomized, sham-controlled acute trials have demonstrated the efficacy of TMS for treatment-resistant depression.

A recent meta-analysis considered 18 studies (N = 1,970) that met the authors’ criteria for inclusion.3 They found that TMS monotherapy was statistically and clinically more effective than a sham procedure based on:

 

  • improvement in depressive symptoms (mean decrease in baseline Hamilton Depression Rating Scale [HDRS] score, −4.53 [95% CI, −6.11 to −2.96])
  • response rate; response was 3 times more likely with TMS (relative risk 3.38 [95% CI, 2.24 to 5.10])
  • remission rate; remission was 5 times more likely with TMS (relative risk, 5.07 [95% CI, 2.50 to 10.30]).

Another meta-analysis (7 studies, N = 279) considered TMS as an augmentation strategy to standard medication for treatment-resistant depression.4 The authors reported that, based on change in HDRS scores, the pooled standardized mean difference between active and sham TMS augmentation was 0.86 (P < .00001). Furthermore, the pooled response rate with TMS augmentation was 46.6%, compared with 22.1% with the sham procedure (P < .0003).

Acute naturalistic TMS studies. The efficacy of TMS is supported by a large, naturalistic study of 307 patients with treatment-resistant depression who were assessed at baseline and during a standard course of TMS.5 Considering change score in the Clinician Global Impressions-Severity (CGI-S) scale, significant improvement was seen from baseline to end of treatment (−1.9 ± 1.4; P < .0001), with a clinician-assessed response rate of 58.0% and remission rate of 37.1%. Of note: Self-reported quality-of-life measures (on the Medical Outcomes Study 36-Item Short-Form Health Survey and EuroQol 5-Dimensions) also significantly improved during this relatively brief period.6

Maintenance strategies after acute TMS response. Most patients referred for TMS have a depressive illness characterized by a chronic, relapsing course and inadequate response to pharmacotherapy or psychotherapy, or their combination. An effective maintenance strategy after acute response to TMS is paramount. This includes:

 

  • prolonged tapering schedule after an acute TMS course is completed
  • maintenance medication or psychotherapy, or both
  • scheduled periodic maintenance TMS sessions (usually as an augmentation strategy)
  • reintroduction of TMS as needed with early signs of relapse. In this context, several trials have assessed the durability of acute TMS benefit.

A semi-controlled maintenance study followed 99 patients who had at least a 25% decrease in baseline HDRS score after acute TMS treatment.7 They were then tapered from their TMS sessions over 3 weeks while an antidepressant was titrated up. If, at any time during the subsequent 6 months, early signs of depression relapse were noted (ie, change of at least 1 point on the CGI-S for 2 consecutive weeks), TMS was reintroduced. At the end of the trial, 10 patients (13%) had relapsed and 38 (38%) had an exacerbation of symptoms sufficient to warrant reintroduction of TMS. Of those, 32 (84%) re-achieved mood stability.

In another study, 50 patients who had achieved remission during an acute course of TMS were followed for 3 months.8 After TMS taper and continued pharmacotherapy or naturalistic follow-up, 29 (58%) remained in remission; 2 (4%) maintained partial response; and 1 (2%) relapsed.

 

 

In a controlled, pilot, maintenance trial, 67 unmedicated patients with treatment-resistant depression received an acute course of TMS.9 Forty-seven of the responders were then randomized to a 1-year follow-up trial with or without a scheduled monthly TMS session. All patients could receive reintroduction TMS if they met criteria for symptom worsening.

Both groups had a similar outcome. The number of patients who did not require TMS reintroduction was 9 of 23 (39%) in the scheduled TMS group vs 9 of 26 (35%) in the no-scheduled TMS group (P < .1). Although no difference was noted between groups, the authors commented that these preliminary results will help inform larger, more definitive trials. They concluded that both acute and maintenance TMS monotherapy might be an option—for some patients.

A long-term, naturalistic outcomes study followed 257 treatment-resistant depressed patients for 1 year after they responded to an acute course of TMS.10 In addition to most patients receiving ongoing maintenance medication, they also could receive reintroduction of TMS if symptoms became worse.

Compared with pre-TMS baseline, there was a statistically significant reduction in the mean total score on the CGI-S scale (primary outcome, P < .0001) at the end of acute treatment that was sustained at follow-up. Ninety-six patients (36.2%) required reintroduction of TMS and 75 of 120 (62.5%) who initially met response or remission criteria after acute treatment continued to meet response criteria after 1 year. The authors concluded that TMS demonstrated both a statistically and clinically meaningful durability of acute benefit during this time frame.


TMS and electroconvulsive therapy
For more than 75 years, ECT has consistently proved to be an effective treatment for major depressive disorder. Although the use of ECT has fluctuated over this period, one practice survey estimated that 100,000 patients receive ECT annually.11

ECT has limitations, however, including cost, the need for general anesthesia, and cognitive deficits that range from short-term confusion to anterograde and retrograde amnesia, which can persist for weeks beyond active treatment.12 Despite increasing awareness of mental illness, stigma also remains a significant barrier to receiving ECT.

TMS vs ECT. Several trials have directly compared ECT and TMS:

 

  • A recent meta-analysis of 9 trials included 384 patients with depression who were considered clinically appropriate for ECT and were randomized to one or the other treatment.13 Both modalities produced a significant reduction in baseline HDRS score, but ECT (15.4 point reduction) was superior to TMS (9.3 point reduction) in the degree of improvement (P < .01).
  • Another meta-analysis of 9 trials (N = 425) found ECT superior to TMS in terms of response (P < .03) and remission (P < .006) rates, based on improvement in the HDRS score.14 When psychotic depressed patients were excluded, however, TMS produced effects equivalent to ECT.

In contrast to what was seen with ECT, cognitive testing of patients who received TMS revealed no deterioration in any domain. Furthermore, one of the comparison studies observed a modest, but statistically significant, improvement in patient’s working memory-executive function, objective memory, and fine-motor speed over the course of TMS treatment.15

TMS plus ECT. A 2-week, randomized, single-blind, controlled pilot study (N = 22) examined the combination of TMS and ECT as acute treatment of depression.16 Patients were assigned to receive either unilateral non-dominant (UND) ECT 3 days a week or a combination of 1 UND ECT treatment followed by 4 days of TMS. At the conclusion of treatment, UND ECT plus TMS group produced comparable efficacy and fewer adverse effects compared with the UND ECT-only group.

TMS maintenance after acute ECT response. Most patients who are referred for ECT have a depressive illness characterized by repeated episodes and incomplete response to pharmacotherapy or psychotherapy, or both. The need for an effective maintenance strategy after the acute response is therefore critical. Medication or ECT, or both, are commonly used to maintain acute benefit but, regrettably, a recent systematic review of the durability of benefit with such strategies found a substantial percentage (approximately 50%) of patients relapsed within the first year.17

 

  • In this context, a case series report found that 1 or 2 weekly, sequential, bilateral TMS treatments after a successful acute course of ECT maintained response in 5 of 6 patients over 6 to 12 months.18
  • Another case series (N = 6) transitioned stable patients from maintenance ECT to maintenance TMS, primarily because of adverse effects with ECT.19 With a mean frequency of 1 TMS treatment every 3.5 weeks, all 6 patients remained stable for as long as 6 months. Subsequently, 2 patients relapsed—1 at 8 months and 1 at 9 months.
 

 

Advantages of maintenance TMS over maintenance ECT include lower cost, fewer adverse effects (particularly cognitive deficits), and the ability to remain independent during the period of the treatment sessions.

TMS as an assessment tool for ECT response. TMS can be used to study excitability in cortical circuits. In a study, EEG potentials evoked by TMS before and after a course of ECT in 8 severely depressed patients revealed an increase in frontal cortical excitability, compared with baseline.20 Such findings support the ability of ECT to produce synaptic potentiation in humans. Furthermore, to the extent that depression presents with alterations in frontal cortical excitability, serial EEG-TMS measurements might be an effective tool to guide and monitor treatment progress with ECT, as well as other forms of therapeutic modulation.

Summing up: TMS and ECT. Although a definitive comparative study is needed, available evidence suggests that TMS might be an alternative treatment in a subgroup of patients who are referred for ECT. Factors that might warrant considering TMS over ECT include:

 

  • patient preference
  • fear of anesthesia
  • concern about cognitive deficits
  • stigma.

Although TMS might offer a workable alternative to ECT for acute and maintenance treatment of depression in selected patients, further refinement of the delivery of TMS is also needed to (1) enhance its efficacy and (2) identify clinical and biological markers to better define this select population.


Standard TMS treatment parameters
Superficial TMS. Superficial TMS for depression typically involves a single coil placed over the left dorsolateral prefrontal cortex. The standard, FDA-approved protocol includes stimulating at 110% of motor threshold with 75, 4-second trains at 10 Hz (ie, 40 stimulations) interspersed by 26-second intertrain intervals. Without interruption, a standard treatment session takes 37.5 minutes and delivers a total of 3,000 pulses. Most patients require 20 to 30 sessions, on a Monday-through-Friday schedule, to achieve optimal benefit.

This approach stimulates to a depth of approximately 2 or 3 cm. The coil usually is placed over the left dorsolateral prefrontal cortex because earlier studies indicated that decreased activity in this part of the brain correlates with symptoms of depression. When TMS is administered in a rapid repetitive fashion (at >1 Hz; typically, at 10 Hz), blood flow and metabolism in that area of the brain are increased. In addition, imaging studies indicate that trans-synaptic connections with deeper parts of the brain also allow modulation of other relevant neural circuits.

An alternate approach, less well-studied, involves low-frequency stimulation over the right dorsolateral prefrontal cortex. Parameters differ from what is used in left high-frequency dorsolateral prefrontal cortex TMS: frequency <1 Hz; train durations as long as 15 minutes; an intertrain interval of 25 to 180 seconds; 120 to 900 stimulations per train; and 2,400 to 18,000 total stimulations.

One hypothesis is that this low-frequency approach selectively stimulates inhibitory interneurons, decreases local neuronal activity and diminishes blood flow to deeper structures, such as the amygdala. Although right low-frequency TMS, compared with left high-frequency TMS, has potential advantages of better tolerability and decreased risk for seizures, its relative efficacy is unclear.

Deep TMS. Studies also are pursuing different coil configurations that allow for more direct stimulation of relevant structures (eg, prefrontal neuronal pathways associated with the reward system).

One of these coil designs (ie, the H-coil), coupled to a Magstim TMS stimulator, recently received FDA clearance for treatment-resistant depression. In the pivotal, sham-controlled study, patients received 20 treatment sessions over 4 weeks.21 The treatment protocol consisted of a helmet-like coil placed over the medial and lateral prefrontal cortex. Stimulation parameters included an 18-Hz frequency; stimulation intensity of 120% motor threshold; stimulation train duration of 2 seconds; and an intertrain interval of 20 seconds. The treatment sessions lasted 20.2 minutes and delivered a total of 1,980 stimulations.

Based on the 21-item HDRS, the active treatment coil group achieved a significantly greater decrease in baseline score (6.39 vs 3.28; P < .008); a greater response rate (37% vs 27.8%; P < .03); and a greater remission rate (30.4% vs 15.8%; P < .016) compared with the sham coil group.

Next, in what is the only randomized, controlled maintenance assessment to date, the same patients were followed for an additional 12 weeks, continuing blinded treatments twice weekly. At the end of the second phase, the active treatment group also demonstrated greater benefit than the sham group (P < .03). One seizure did occur, possibly related to excessive alcohol use; but this raises the question of whether treating at a higher frequency (18 Hz) with greater depth and less focality might increase the risk of seizure.

 

 

To assess the potential advantages, as well as the relative safety, of this approach over standard TMS delivery, an adequately designed and powered trial comparing the H-coil and a single-coil device is needed.


Alternate TMS approaches
Efforts to improve the clinical effectiveness of TMS for treating depression include several approaches.

Theta burst stimulation (TBS) is a patterned form of TMS pulse delivery that utilizes high and low frequencies in the same stimulus train (eg, three 50-Hz bursts delivered 5 times a second). Such a pulse sequence can modulate long-term depression and long-term potentiation mechanisms that induce plasticity in areas such as the hippocampus.22

Intermittent TBS (iTBS) administers stimulations over a relatively brief duration (eg, 2 seconds) or intermittently (eg, every 10 seconds) for a specific period (eg, 190 seconds [600 pulses in total]) over the left dorsolateral prefrontal cortex. This technique induces long-term potentiation and produces effects similar to those of high-frequency TMS.

In contrast, continuous TBS (cTBS) administers a continuous train (eg, 40 seconds [600 total pulses]) over the right dorsolateral prefrontal cortex. This induces long-term depression and produces effects similar to low-frequency TMS.

Recent studies using different delivery paradigms have generated mixed results:

Study 1: Fifty-six patients with depression received active treatment; 17 others, a sham procedure.23 This study used 3 different conditions:

 

  • a combination of low-frequency and high-frequency TMS stimulation, administered over the right and left dorsolateral prefrontal cortices, respectively
  • a combination of iTBS over the left dorsolateral prefrontal cortex and cTBS over the right dorsolateral prefrontal cortex
  • a sham procedure, in which no magnetic field was created.

Neither active treatment arm separated from the sham procedure based on change scores in the 21-item HDRS (P = not significant).

Study 2: Sixty treatment-resistant depression patients were assigned to cTBS, iTBS, a combination of the 2 procedures, or a sham procedure.24 After 2 weeks, the active treatment arms produced the greatest benefit, based on change in scores on the 17-item HDRS, which differed significantly among the 4 groups (F value = 6.166; P < .001); the iTBS and combination arms demonstrated the most robust effect.

There were also significantly more responders in the iTBS (40.0%) and combination groups (66.7%) than in the cTBS (25.0%) and sham groups (13.3%) (P < .010). A lower level of treatment refractoriness predicted a better outcome.

Study 3: Twenty-nine depressed patients were randomized to cTBS over the right dorsolateral prefrontal cortex or a sham procedure.25 Overall, there was no difference between groups; however, actively treated patients who were unmedicated (n = 3) or remained on a stable dosage of medication during treatment (n = 8) did experience a significantly greater reduction in the HDRS score.

Study 4: In a pilot trial, 32 depressed patients were randomized to 30 sessions of adjunctive combined iTBS plus cTBS or bilateral sham TBS.26 Based on reduction from the baseline Montgomery-Åsberg Depression Rating Scale score, 9 patients in the active treatment group and 4 in the sham group achieved response (odds ratio, 3.86; P < .048).

If at least comparable efficacy can be clearly demonstrated, advantages of TBS over standard TMS include a significantly reduced administration time, which might allow for more patients to be treated and reduce associated costs of treatment.27

Magnetic low-field synchronized stimulation is produced by rotating spherical rare-earth magnets that are synchronized to an individual’s alpha frequency. A recent 6-week, double-blind, sham-controlled trial (N = 202) reported that, in the intention-to-treat population, there was no difference in outcome between treatment arms. In patients who completed the study according to protocol (120 of 202), however, active treatment was significantly better in decreasing baseline HDRS score (P < .033).28

Magnetic seizure therapy (MST) is an experimental approach to treating patients with more severe depression that is resistant to medical therapy. The primary aim is to use TMS to induce a seizure, thus achieving the same efficacy as provided by ECT but without the adverse cognitive effects of ECT. With MST, the TMS device uses much higher stimulation settings to produce a seizure—the goal being to avoid direct electrical current to the brain’s memory centers.29

A pilot study considered the clinical and cognitive effects of MST in a group of 26 treatment-resistant depression patients (10 randomized; 16 open-label).30 Based on reduction in baseline HDRS scores at the end of the trial, 69% of patients achieved response and 46% met remission criteria; however, one-half of patients relapsed within 6 months.

Importantly, no cognitive adverse effects were observed. Furthermore, the antidepressant and anti-anxiety effects of MST were associated with localized metabolic changes in brain areas implicated in the pathophysiology of depression.

 

 

The investigators concluded that MST might constitute an effective, well-tolerated, and safe treatment for patients unable to benefit from available medical therapies for depression. In addition to confirmation of acute benefit in more definitive trials, the issue of durability of effect needs further clarification.


TMS is a key component of neuropsychiatric practice
It has been 3 decades since Barker et al31 developed the technology to deliver intense, localized magnetic pulses to specific areas of the nervous system. During this period, the role of TMS as a probe of the central and peripheral nervous systems has expanded to include various therapeutic applications, primarily focusing on treatment-resistant major depressive disorder.

Now, increasing sophistication in the choice of stimulation parameters and other ongoing efforts to optimize the benefits of TMS are yielding improved clinical outcomes. Research is still needed to better define the place of TMS in the management of subtypes of depression that are particularly difficult to treat and that do not benefit adequately from medications or psychotherapy or their combination.

Growing support from controlled trials, systematic reviews, meta-analyses, naturalistic outcome studies, and professional guidelines indicate that TMS has an increasingly important role in clinical practice.

 


Bottom Line
Clinical use of transcranial magnetic stimulation (TMS) continues to expand, particularly for treatment-resistant depression. At the same time, ongoing research seeks to refine the application of TMS to enhance and expand its benefits.

 


Related Resources

 

  • www.Brainsway.com
  • www.Magstim.com
  • www.MagVenture.com

 

     


    Disclosures
    Dr. Janicak receives research support from Neuronetics, Inc; Sunovion Pharmaceuticals, Inc; and Ortho-McNeil-Janssen. He has served as a consultant to Neuronetics, Inc. Dr. Sackett, Ms. Kudrna, and Dr. Cutler report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
    References


    1. Janicak PG, Dowd SM, Rado JT, et al. The re-emerging role of therapeutic neuromodulation. Current Psychiatry. 2010;9(11):66-70,72-74.
    2. Janicak PG, Dokucu ME. Transcranial magnetic stimulation for the treatment of major depression. Neuropsychiatr Dis Treat. 2015;11:1549-1560.
    3. Gaynes BN, Lloyd SW, Lux L, et al. Repetitive transcranial magnetic stimulation for treatment-resistant depression: a systematic review and meta-analysis. J Clin Psychiatry. 2014;75(5):477-489; quiz 489.
    4. Liu B, Zhang Y, Zhang L, et al. Repetitive transcranial magnetic stimulation as an augmentative strategy for treatment-resistant depression, a meta-analysis of randomized, double-blind and sham-controlled study. BMC Psychiatry. 2014;14:342.
    5. Carpenter LL, Janicak PG, Aaronson ST, et al. Transcranial magnetic stimulation (TMS) for major depression: a multisite, naturalistic, observational study of acute treatment outcomes in clinical practice. Depress Anxiety. 2012;29(7):587-596.
    6. Janicak PG, Dunner DL, Aaronson ST, et al. Transcranial magnetic stimulation (TMS) for major depression: a multisite, naturalistic, observational study of quality of life outcome measures in clinical practice. CNS Spectr. 2013;18(6):322-332.
    7. Janicak PG, Nahas Z, Lisanby SH, et al. Durability of clinical benefit with transcranial magnetic stimulation (TMS) in the treatment of pharmacoresistant major depression: assessment of relapse during a 6-month, multisite, open-label study. Brain Stimul. 2010;3(4):187-199.
    8. Mantovani A, Pavlicova M, Avery D, et al. Long-term efficacy of repeated daily prefrontal transcranial magnetic stimulation (TMS) in treatment-resistant depression. Depress Anxiety. 2012;29(10):883-890.
    9. Philip NS, Dunner DL, Dowd SM, et al. Can medication free, treatment-resistant, depressed patients who initially respond to TMS be maintained off medications? A prospective, 12-month multisite randomized pilot study. Brain Stimul. 2016;9(2):251-257.
    10. Dunner DL, Aaronson ST, Sackeim HA, et al. A multisite, naturalistic, observational study of transcranial magnetic stimulation for patients with pharmacoresistant major depressive disorder: durability of benefit over a 1-year follow-up period. J Clin Psychiatry. 2014;75(12):1394-1401.
    11. Hermann RC, Dorwart RA, Hoover CW. Variation in ECT use in the United States. Am J Psychiatry. 1995;152(6):869-875.
    12. Sackeim HA. Memory and ECT: from polarization to reconciliation. J ECT. 2000;16(2):87-96.
    13. Micallef-Trigona B. Comparing the effects of repetitive transcranial magnetic stimulation and electroconvulsive therapy in the treatment of depression: a systematic review and meta-analysis. Depress Res Treat. 2014;2014:135049. doi: 10.1155/2014/135049.
    14. Ren J, Li H, Palaniyappan L, et al. Repetitive transcranial magnetic stimulation versus electroconvulsive therapy for major depression: a systematic review and meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry. 2014;51:181-189.
    15. Martis B, Alam D, Dowd SM, et al. Neurocognitive effects of repetitive transcranial magnetic stimulation in severe major depression. Clin Neurophysiol. 2003;114(6):1125-1132.
    16. Pridmore S, Rybak M, Turnier-Shea Y, et al. Comparison of transcranial magnetic stimulation and electroconvulsive therapy in depression. In: Miyoshi K, Shapiro CM, Gaviria M, et al, eds. Contemporary neuropsychiatry. Tokyo, Japan: Springer; 2001:237-241.
    17. Jelovac A, Kolshus E, McLoughlin DM. Relapse following successful electroconvulsive therapy for major depression: a meta-analysis. Neuropsychopharmacology. 2013;38(12):2467-2474.
    18. Noda Y, Daskalakis Z, Ramos C, et al. Repetitive transcranial magnetic stimulation to maintain treatment response to electroconvulsive therapy in depression: a case series. Front Psychiatry. 2013;4:73.
    19. Cristancho MA, Helmer A, Connolly R, et al. Transcranial magnetic stimulation maintenance as a substitute for maintenance electroconvulsive therapy: a case series. J ECT. 2013;29(2):106-108.
    20. Casarotto S, Canali P, Rosanova M, et al. Assessing the effects of electroconvulsive therapy on cortical excitability by means of transcranial magnetic stimulation and electroencephalography. Brain Topogr. 2013;26(2):326-337.
    21. 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(1):64-73.
    22. Daskalakis ZJ. Theta-burst transcranial magnetic stimulation in depression: when less may be more. Brain. 2014;137(pt 7):1860-1862.
    23. Prasser J, Schecklmann M, Poeppl TB, et al. Bilateral prefrontal rTMS and theta burst TMS as an add-on treatment for depression: a randomized placebo controlled trial. World J Biol Psychiatry. 2015;16(1):57-65.
    24. Li CT, Chen MH, Juan CH, et al. Efficacy of prefrontal theta-burst stimulation in refractory depression: a randomized sham-controlled study. Brain. 2014;137(pt 7):2088-2098.
    25. Chistyakov A, Kreinin B, Marmor S, et al. Preliminary assessment of the therapeutic efficacy of continuous theta-burst magnetic stimulation (cTBS) in major depression: a double-blind sham-controlled study. J Affect Disord. 2015;170:225-229.
    26. Plewnia C, Pasqualetti P, Große S, et al. Treatment of major depression with bilateral theta burst stimulation: a randomized controlled pilot trial. J Affect Disord. 2014;156:219-223.
    27. Chung SW, Hoy KE, Fitzgerald PB. Theta-burst stimulation: a new form of TMS treatment for depression? Depress Anxiety. 2015;32(3):182-192.
    28. Leuchter AF, Cook IA, Feifel D, et al. Efficacy and safety of low-field synchronized transcranial magnetic stimulation (sTMS) for treatment of major depression. Brain Stimul. 2015;8(4):787-794.
    29. Cretaz E, Brunoni AR, Lafer B. Magnetic seizure therapy for unipolar and bipolar depression: a systematic review. Neural Plast. 2015;2015:521398. doi: 10.1155/2015/521398.
    30. Kayser S, Bewernick BH, Matusch A, et al. Magnetic seizure therapy in treatment-resistant depression: clinical, neuropsychological and metabolic effects. Psychol Med. 2015;45(5):1073-1092.
    31. Barker AT, Jalinous R, Freeston IL. Non-invasive magnetic stimulation of human motor cortex. Lancet. 1985;1(8437):1106-1107.

    Article PDF
    Author and Disclosure Information

     

    Philip G. Janicak, MD
    Director

    Vincent Sackett, MD
    Associate Director

    Karyn Kudrna, BSN, RN
    Coordinator

    Bradley Cutler, MD
    Associate Director

    Transcranial Magnetic Stimulation Center
    Linden Oaks Medical Group
    Naperville, Illinois


    Dr. Janicak is also Adjunct Professor, Northwestern University Department of Psychiatry and Behavioral Sciences, Chicago, Illinois; and Member, Current Psychiatry Editorial Board.

    Dr. Cutler is also Assistant Professor, University of Illinois College of Medicine, Chicago, Illinois.

    Issue
    Current Psychiatry - 15(6)
    Publications
    Topics
    Page Number
    49-56
    Legacy Keywords
    TMS, transcranial magnetic stimulation, major depressive disorders, MDD, depressive disorder, depressive disorder, depression, depressed, suicide, ECT, electroconvulsive therapy, theta burst stimulation, TBS, magnetic seizure therapy, MST
    Sections
    Author and Disclosure Information

     

    Philip G. Janicak, MD
    Director

    Vincent Sackett, MD
    Associate Director

    Karyn Kudrna, BSN, RN
    Coordinator

    Bradley Cutler, MD
    Associate Director

    Transcranial Magnetic Stimulation Center
    Linden Oaks Medical Group
    Naperville, Illinois


    Dr. Janicak is also Adjunct Professor, Northwestern University Department of Psychiatry and Behavioral Sciences, Chicago, Illinois; and Member, Current Psychiatry Editorial Board.

    Dr. Cutler is also Assistant Professor, University of Illinois College of Medicine, Chicago, Illinois.

    Author and Disclosure Information

     

    Philip G. Janicak, MD
    Director

    Vincent Sackett, MD
    Associate Director

    Karyn Kudrna, BSN, RN
    Coordinator

    Bradley Cutler, MD
    Associate Director

    Transcranial Magnetic Stimulation Center
    Linden Oaks Medical Group
    Naperville, Illinois


    Dr. Janicak is also Adjunct Professor, Northwestern University Department of Psychiatry and Behavioral Sciences, Chicago, Illinois; and Member, Current Psychiatry Editorial Board.

    Dr. Cutler is also Assistant Professor, University of Illinois College of Medicine, Chicago, Illinois.

    Article PDF
    Article PDF

    Since 2008, the FDA has cleared 4 transcranial magnetic stimulation (TMS) devices for treating depression (Related Resources). In that time, the availability of TMS has steadily grown within and outside the United States.

    Parallel with increasing clinical utilization of this technology, research continues into the benefit of TMS for treatment-resistant depression; such research includes additional, supportive, acute, sham-controlled trials; comparison trials with electroconvulsive therapy (ECT) for more severe episodes of depression; short- and long-term real-world outcome studies; exploration of alternative treatment parameters to further enhance its efficacy; and the development of other TMS approaches. In this article, we review recent developments in the application of TMS to treat major depressive disorder—in particular, treatment-resistant depression (Box).
     

    RTEmagicC_CP015060049_b1.jpg.jpg


    Therapeutic neuromodulation
    The underlying premise of neuromodulation is that the brain is an electrochemical organ that can be modulated by pharmacotherapy or device-based approaches, or their combination.1 ECT is the prototypic device-based neuromodulation approach, and remains one of the most effective treatments for severe depression.

    More recently, however, other methods have been, and continue to be, developed to treat patients who do not achieve adequate benefit from psychotherapy or medical therapy, or both, and who might not be an ideal candidate for ECT (Table,1). In addition to the potential therapeutic benefit of these alternative strategies, some could avoid safety and tolerability concerns associated with medication (weight gain, sexual dysfunction) and ECT (eg, cognitive deficits).

    RTEmagicC_CP015060049_t1.jpg.jpg


    TMS, which utilizes intense, localized magnetic fields to alter activity in neural circuits implicated in the pathophysiology of depression, represents an important example of this initiative.2


    TMS has established efficacy for depression
    Sham-controlled trials. Several randomized, sham-controlled acute trials have demonstrated the efficacy of TMS for treatment-resistant depression.

    A recent meta-analysis considered 18 studies (N = 1,970) that met the authors’ criteria for inclusion.3 They found that TMS monotherapy was statistically and clinically more effective than a sham procedure based on:

     

    • improvement in depressive symptoms (mean decrease in baseline Hamilton Depression Rating Scale [HDRS] score, −4.53 [95% CI, −6.11 to −2.96])
    • response rate; response was 3 times more likely with TMS (relative risk 3.38 [95% CI, 2.24 to 5.10])
    • remission rate; remission was 5 times more likely with TMS (relative risk, 5.07 [95% CI, 2.50 to 10.30]).

    Another meta-analysis (7 studies, N = 279) considered TMS as an augmentation strategy to standard medication for treatment-resistant depression.4 The authors reported that, based on change in HDRS scores, the pooled standardized mean difference between active and sham TMS augmentation was 0.86 (P < .00001). Furthermore, the pooled response rate with TMS augmentation was 46.6%, compared with 22.1% with the sham procedure (P < .0003).

    Acute naturalistic TMS studies. The efficacy of TMS is supported by a large, naturalistic study of 307 patients with treatment-resistant depression who were assessed at baseline and during a standard course of TMS.5 Considering change score in the Clinician Global Impressions-Severity (CGI-S) scale, significant improvement was seen from baseline to end of treatment (−1.9 ± 1.4; P < .0001), with a clinician-assessed response rate of 58.0% and remission rate of 37.1%. Of note: Self-reported quality-of-life measures (on the Medical Outcomes Study 36-Item Short-Form Health Survey and EuroQol 5-Dimensions) also significantly improved during this relatively brief period.6

    Maintenance strategies after acute TMS response. Most patients referred for TMS have a depressive illness characterized by a chronic, relapsing course and inadequate response to pharmacotherapy or psychotherapy, or their combination. An effective maintenance strategy after acute response to TMS is paramount. This includes:

     

    • prolonged tapering schedule after an acute TMS course is completed
    • maintenance medication or psychotherapy, or both
    • scheduled periodic maintenance TMS sessions (usually as an augmentation strategy)
    • reintroduction of TMS as needed with early signs of relapse. In this context, several trials have assessed the durability of acute TMS benefit.

    A semi-controlled maintenance study followed 99 patients who had at least a 25% decrease in baseline HDRS score after acute TMS treatment.7 They were then tapered from their TMS sessions over 3 weeks while an antidepressant was titrated up. If, at any time during the subsequent 6 months, early signs of depression relapse were noted (ie, change of at least 1 point on the CGI-S for 2 consecutive weeks), TMS was reintroduced. At the end of the trial, 10 patients (13%) had relapsed and 38 (38%) had an exacerbation of symptoms sufficient to warrant reintroduction of TMS. Of those, 32 (84%) re-achieved mood stability.

    In another study, 50 patients who had achieved remission during an acute course of TMS were followed for 3 months.8 After TMS taper and continued pharmacotherapy or naturalistic follow-up, 29 (58%) remained in remission; 2 (4%) maintained partial response; and 1 (2%) relapsed.

     

     

    In a controlled, pilot, maintenance trial, 67 unmedicated patients with treatment-resistant depression received an acute course of TMS.9 Forty-seven of the responders were then randomized to a 1-year follow-up trial with or without a scheduled monthly TMS session. All patients could receive reintroduction TMS if they met criteria for symptom worsening.

    Both groups had a similar outcome. The number of patients who did not require TMS reintroduction was 9 of 23 (39%) in the scheduled TMS group vs 9 of 26 (35%) in the no-scheduled TMS group (P < .1). Although no difference was noted between groups, the authors commented that these preliminary results will help inform larger, more definitive trials. They concluded that both acute and maintenance TMS monotherapy might be an option—for some patients.

    A long-term, naturalistic outcomes study followed 257 treatment-resistant depressed patients for 1 year after they responded to an acute course of TMS.10 In addition to most patients receiving ongoing maintenance medication, they also could receive reintroduction of TMS if symptoms became worse.

    Compared with pre-TMS baseline, there was a statistically significant reduction in the mean total score on the CGI-S scale (primary outcome, P < .0001) at the end of acute treatment that was sustained at follow-up. Ninety-six patients (36.2%) required reintroduction of TMS and 75 of 120 (62.5%) who initially met response or remission criteria after acute treatment continued to meet response criteria after 1 year. The authors concluded that TMS demonstrated both a statistically and clinically meaningful durability of acute benefit during this time frame.


    TMS and electroconvulsive therapy
    For more than 75 years, ECT has consistently proved to be an effective treatment for major depressive disorder. Although the use of ECT has fluctuated over this period, one practice survey estimated that 100,000 patients receive ECT annually.11

    ECT has limitations, however, including cost, the need for general anesthesia, and cognitive deficits that range from short-term confusion to anterograde and retrograde amnesia, which can persist for weeks beyond active treatment.12 Despite increasing awareness of mental illness, stigma also remains a significant barrier to receiving ECT.

    TMS vs ECT. Several trials have directly compared ECT and TMS:

     

    • A recent meta-analysis of 9 trials included 384 patients with depression who were considered clinically appropriate for ECT and were randomized to one or the other treatment.13 Both modalities produced a significant reduction in baseline HDRS score, but ECT (15.4 point reduction) was superior to TMS (9.3 point reduction) in the degree of improvement (P < .01).
    • Another meta-analysis of 9 trials (N = 425) found ECT superior to TMS in terms of response (P < .03) and remission (P < .006) rates, based on improvement in the HDRS score.14 When psychotic depressed patients were excluded, however, TMS produced effects equivalent to ECT.

    In contrast to what was seen with ECT, cognitive testing of patients who received TMS revealed no deterioration in any domain. Furthermore, one of the comparison studies observed a modest, but statistically significant, improvement in patient’s working memory-executive function, objective memory, and fine-motor speed over the course of TMS treatment.15

    TMS plus ECT. A 2-week, randomized, single-blind, controlled pilot study (N = 22) examined the combination of TMS and ECT as acute treatment of depression.16 Patients were assigned to receive either unilateral non-dominant (UND) ECT 3 days a week or a combination of 1 UND ECT treatment followed by 4 days of TMS. At the conclusion of treatment, UND ECT plus TMS group produced comparable efficacy and fewer adverse effects compared with the UND ECT-only group.

    TMS maintenance after acute ECT response. Most patients who are referred for ECT have a depressive illness characterized by repeated episodes and incomplete response to pharmacotherapy or psychotherapy, or both. The need for an effective maintenance strategy after the acute response is therefore critical. Medication or ECT, or both, are commonly used to maintain acute benefit but, regrettably, a recent systematic review of the durability of benefit with such strategies found a substantial percentage (approximately 50%) of patients relapsed within the first year.17

     

    • In this context, a case series report found that 1 or 2 weekly, sequential, bilateral TMS treatments after a successful acute course of ECT maintained response in 5 of 6 patients over 6 to 12 months.18
    • Another case series (N = 6) transitioned stable patients from maintenance ECT to maintenance TMS, primarily because of adverse effects with ECT.19 With a mean frequency of 1 TMS treatment every 3.5 weeks, all 6 patients remained stable for as long as 6 months. Subsequently, 2 patients relapsed—1 at 8 months and 1 at 9 months.
     

     

    Advantages of maintenance TMS over maintenance ECT include lower cost, fewer adverse effects (particularly cognitive deficits), and the ability to remain independent during the period of the treatment sessions.

    TMS as an assessment tool for ECT response. TMS can be used to study excitability in cortical circuits. In a study, EEG potentials evoked by TMS before and after a course of ECT in 8 severely depressed patients revealed an increase in frontal cortical excitability, compared with baseline.20 Such findings support the ability of ECT to produce synaptic potentiation in humans. Furthermore, to the extent that depression presents with alterations in frontal cortical excitability, serial EEG-TMS measurements might be an effective tool to guide and monitor treatment progress with ECT, as well as other forms of therapeutic modulation.

    Summing up: TMS and ECT. Although a definitive comparative study is needed, available evidence suggests that TMS might be an alternative treatment in a subgroup of patients who are referred for ECT. Factors that might warrant considering TMS over ECT include:

     

    • patient preference
    • fear of anesthesia
    • concern about cognitive deficits
    • stigma.

    Although TMS might offer a workable alternative to ECT for acute and maintenance treatment of depression in selected patients, further refinement of the delivery of TMS is also needed to (1) enhance its efficacy and (2) identify clinical and biological markers to better define this select population.


    Standard TMS treatment parameters
    Superficial TMS. Superficial TMS for depression typically involves a single coil placed over the left dorsolateral prefrontal cortex. The standard, FDA-approved protocol includes stimulating at 110% of motor threshold with 75, 4-second trains at 10 Hz (ie, 40 stimulations) interspersed by 26-second intertrain intervals. Without interruption, a standard treatment session takes 37.5 minutes and delivers a total of 3,000 pulses. Most patients require 20 to 30 sessions, on a Monday-through-Friday schedule, to achieve optimal benefit.

    This approach stimulates to a depth of approximately 2 or 3 cm. The coil usually is placed over the left dorsolateral prefrontal cortex because earlier studies indicated that decreased activity in this part of the brain correlates with symptoms of depression. When TMS is administered in a rapid repetitive fashion (at >1 Hz; typically, at 10 Hz), blood flow and metabolism in that area of the brain are increased. In addition, imaging studies indicate that trans-synaptic connections with deeper parts of the brain also allow modulation of other relevant neural circuits.

    An alternate approach, less well-studied, involves low-frequency stimulation over the right dorsolateral prefrontal cortex. Parameters differ from what is used in left high-frequency dorsolateral prefrontal cortex TMS: frequency <1 Hz; train durations as long as 15 minutes; an intertrain interval of 25 to 180 seconds; 120 to 900 stimulations per train; and 2,400 to 18,000 total stimulations.

    One hypothesis is that this low-frequency approach selectively stimulates inhibitory interneurons, decreases local neuronal activity and diminishes blood flow to deeper structures, such as the amygdala. Although right low-frequency TMS, compared with left high-frequency TMS, has potential advantages of better tolerability and decreased risk for seizures, its relative efficacy is unclear.

    Deep TMS. Studies also are pursuing different coil configurations that allow for more direct stimulation of relevant structures (eg, prefrontal neuronal pathways associated with the reward system).

    One of these coil designs (ie, the H-coil), coupled to a Magstim TMS stimulator, recently received FDA clearance for treatment-resistant depression. In the pivotal, sham-controlled study, patients received 20 treatment sessions over 4 weeks.21 The treatment protocol consisted of a helmet-like coil placed over the medial and lateral prefrontal cortex. Stimulation parameters included an 18-Hz frequency; stimulation intensity of 120% motor threshold; stimulation train duration of 2 seconds; and an intertrain interval of 20 seconds. The treatment sessions lasted 20.2 minutes and delivered a total of 1,980 stimulations.

    Based on the 21-item HDRS, the active treatment coil group achieved a significantly greater decrease in baseline score (6.39 vs 3.28; P < .008); a greater response rate (37% vs 27.8%; P < .03); and a greater remission rate (30.4% vs 15.8%; P < .016) compared with the sham coil group.

    Next, in what is the only randomized, controlled maintenance assessment to date, the same patients were followed for an additional 12 weeks, continuing blinded treatments twice weekly. At the end of the second phase, the active treatment group also demonstrated greater benefit than the sham group (P < .03). One seizure did occur, possibly related to excessive alcohol use; but this raises the question of whether treating at a higher frequency (18 Hz) with greater depth and less focality might increase the risk of seizure.

     

     

    To assess the potential advantages, as well as the relative safety, of this approach over standard TMS delivery, an adequately designed and powered trial comparing the H-coil and a single-coil device is needed.


    Alternate TMS approaches
    Efforts to improve the clinical effectiveness of TMS for treating depression include several approaches.

    Theta burst stimulation (TBS) is a patterned form of TMS pulse delivery that utilizes high and low frequencies in the same stimulus train (eg, three 50-Hz bursts delivered 5 times a second). Such a pulse sequence can modulate long-term depression and long-term potentiation mechanisms that induce plasticity in areas such as the hippocampus.22

    Intermittent TBS (iTBS) administers stimulations over a relatively brief duration (eg, 2 seconds) or intermittently (eg, every 10 seconds) for a specific period (eg, 190 seconds [600 pulses in total]) over the left dorsolateral prefrontal cortex. This technique induces long-term potentiation and produces effects similar to those of high-frequency TMS.

    In contrast, continuous TBS (cTBS) administers a continuous train (eg, 40 seconds [600 total pulses]) over the right dorsolateral prefrontal cortex. This induces long-term depression and produces effects similar to low-frequency TMS.

    Recent studies using different delivery paradigms have generated mixed results:

    Study 1: Fifty-six patients with depression received active treatment; 17 others, a sham procedure.23 This study used 3 different conditions:

     

    • a combination of low-frequency and high-frequency TMS stimulation, administered over the right and left dorsolateral prefrontal cortices, respectively
    • a combination of iTBS over the left dorsolateral prefrontal cortex and cTBS over the right dorsolateral prefrontal cortex
    • a sham procedure, in which no magnetic field was created.

    Neither active treatment arm separated from the sham procedure based on change scores in the 21-item HDRS (P = not significant).

    Study 2: Sixty treatment-resistant depression patients were assigned to cTBS, iTBS, a combination of the 2 procedures, or a sham procedure.24 After 2 weeks, the active treatment arms produced the greatest benefit, based on change in scores on the 17-item HDRS, which differed significantly among the 4 groups (F value = 6.166; P < .001); the iTBS and combination arms demonstrated the most robust effect.

    There were also significantly more responders in the iTBS (40.0%) and combination groups (66.7%) than in the cTBS (25.0%) and sham groups (13.3%) (P < .010). A lower level of treatment refractoriness predicted a better outcome.

    Study 3: Twenty-nine depressed patients were randomized to cTBS over the right dorsolateral prefrontal cortex or a sham procedure.25 Overall, there was no difference between groups; however, actively treated patients who were unmedicated (n = 3) or remained on a stable dosage of medication during treatment (n = 8) did experience a significantly greater reduction in the HDRS score.

    Study 4: In a pilot trial, 32 depressed patients were randomized to 30 sessions of adjunctive combined iTBS plus cTBS or bilateral sham TBS.26 Based on reduction from the baseline Montgomery-Åsberg Depression Rating Scale score, 9 patients in the active treatment group and 4 in the sham group achieved response (odds ratio, 3.86; P < .048).

    If at least comparable efficacy can be clearly demonstrated, advantages of TBS over standard TMS include a significantly reduced administration time, which might allow for more patients to be treated and reduce associated costs of treatment.27

    Magnetic low-field synchronized stimulation is produced by rotating spherical rare-earth magnets that are synchronized to an individual’s alpha frequency. A recent 6-week, double-blind, sham-controlled trial (N = 202) reported that, in the intention-to-treat population, there was no difference in outcome between treatment arms. In patients who completed the study according to protocol (120 of 202), however, active treatment was significantly better in decreasing baseline HDRS score (P < .033).28

    Magnetic seizure therapy (MST) is an experimental approach to treating patients with more severe depression that is resistant to medical therapy. The primary aim is to use TMS to induce a seizure, thus achieving the same efficacy as provided by ECT but without the adverse cognitive effects of ECT. With MST, the TMS device uses much higher stimulation settings to produce a seizure—the goal being to avoid direct electrical current to the brain’s memory centers.29

    A pilot study considered the clinical and cognitive effects of MST in a group of 26 treatment-resistant depression patients (10 randomized; 16 open-label).30 Based on reduction in baseline HDRS scores at the end of the trial, 69% of patients achieved response and 46% met remission criteria; however, one-half of patients relapsed within 6 months.

    Importantly, no cognitive adverse effects were observed. Furthermore, the antidepressant and anti-anxiety effects of MST were associated with localized metabolic changes in brain areas implicated in the pathophysiology of depression.

     

     

    The investigators concluded that MST might constitute an effective, well-tolerated, and safe treatment for patients unable to benefit from available medical therapies for depression. In addition to confirmation of acute benefit in more definitive trials, the issue of durability of effect needs further clarification.


    TMS is a key component of neuropsychiatric practice
    It has been 3 decades since Barker et al31 developed the technology to deliver intense, localized magnetic pulses to specific areas of the nervous system. During this period, the role of TMS as a probe of the central and peripheral nervous systems has expanded to include various therapeutic applications, primarily focusing on treatment-resistant major depressive disorder.

    Now, increasing sophistication in the choice of stimulation parameters and other ongoing efforts to optimize the benefits of TMS are yielding improved clinical outcomes. Research is still needed to better define the place of TMS in the management of subtypes of depression that are particularly difficult to treat and that do not benefit adequately from medications or psychotherapy or their combination.

    Growing support from controlled trials, systematic reviews, meta-analyses, naturalistic outcome studies, and professional guidelines indicate that TMS has an increasingly important role in clinical practice.

     


    Bottom Line
    Clinical use of transcranial magnetic stimulation (TMS) continues to expand, particularly for treatment-resistant depression. At the same time, ongoing research seeks to refine the application of TMS to enhance and expand its benefits.

     


    Related Resources

     

    • www.Brainsway.com
    • www.Magstim.com
    • www.MagVenture.com

     

       


      Disclosures
      Dr. Janicak receives research support from Neuronetics, Inc; Sunovion Pharmaceuticals, Inc; and Ortho-McNeil-Janssen. He has served as a consultant to Neuronetics, Inc. Dr. Sackett, Ms. Kudrna, and Dr. Cutler report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

      Since 2008, the FDA has cleared 4 transcranial magnetic stimulation (TMS) devices for treating depression (Related Resources). In that time, the availability of TMS has steadily grown within and outside the United States.

      Parallel with increasing clinical utilization of this technology, research continues into the benefit of TMS for treatment-resistant depression; such research includes additional, supportive, acute, sham-controlled trials; comparison trials with electroconvulsive therapy (ECT) for more severe episodes of depression; short- and long-term real-world outcome studies; exploration of alternative treatment parameters to further enhance its efficacy; and the development of other TMS approaches. In this article, we review recent developments in the application of TMS to treat major depressive disorder—in particular, treatment-resistant depression (Box).
       

      RTEmagicC_CP015060049_b1.jpg.jpg


      Therapeutic neuromodulation
      The underlying premise of neuromodulation is that the brain is an electrochemical organ that can be modulated by pharmacotherapy or device-based approaches, or their combination.1 ECT is the prototypic device-based neuromodulation approach, and remains one of the most effective treatments for severe depression.

      More recently, however, other methods have been, and continue to be, developed to treat patients who do not achieve adequate benefit from psychotherapy or medical therapy, or both, and who might not be an ideal candidate for ECT (Table,1). In addition to the potential therapeutic benefit of these alternative strategies, some could avoid safety and tolerability concerns associated with medication (weight gain, sexual dysfunction) and ECT (eg, cognitive deficits).

      RTEmagicC_CP015060049_t1.jpg.jpg


      TMS, which utilizes intense, localized magnetic fields to alter activity in neural circuits implicated in the pathophysiology of depression, represents an important example of this initiative.2


      TMS has established efficacy for depression
      Sham-controlled trials. Several randomized, sham-controlled acute trials have demonstrated the efficacy of TMS for treatment-resistant depression.

      A recent meta-analysis considered 18 studies (N = 1,970) that met the authors’ criteria for inclusion.3 They found that TMS monotherapy was statistically and clinically more effective than a sham procedure based on:

       

      • improvement in depressive symptoms (mean decrease in baseline Hamilton Depression Rating Scale [HDRS] score, −4.53 [95% CI, −6.11 to −2.96])
      • response rate; response was 3 times more likely with TMS (relative risk 3.38 [95% CI, 2.24 to 5.10])
      • remission rate; remission was 5 times more likely with TMS (relative risk, 5.07 [95% CI, 2.50 to 10.30]).

      Another meta-analysis (7 studies, N = 279) considered TMS as an augmentation strategy to standard medication for treatment-resistant depression.4 The authors reported that, based on change in HDRS scores, the pooled standardized mean difference between active and sham TMS augmentation was 0.86 (P < .00001). Furthermore, the pooled response rate with TMS augmentation was 46.6%, compared with 22.1% with the sham procedure (P < .0003).

      Acute naturalistic TMS studies. The efficacy of TMS is supported by a large, naturalistic study of 307 patients with treatment-resistant depression who were assessed at baseline and during a standard course of TMS.5 Considering change score in the Clinician Global Impressions-Severity (CGI-S) scale, significant improvement was seen from baseline to end of treatment (−1.9 ± 1.4; P < .0001), with a clinician-assessed response rate of 58.0% and remission rate of 37.1%. Of note: Self-reported quality-of-life measures (on the Medical Outcomes Study 36-Item Short-Form Health Survey and EuroQol 5-Dimensions) also significantly improved during this relatively brief period.6

      Maintenance strategies after acute TMS response. Most patients referred for TMS have a depressive illness characterized by a chronic, relapsing course and inadequate response to pharmacotherapy or psychotherapy, or their combination. An effective maintenance strategy after acute response to TMS is paramount. This includes:

       

      • prolonged tapering schedule after an acute TMS course is completed
      • maintenance medication or psychotherapy, or both
      • scheduled periodic maintenance TMS sessions (usually as an augmentation strategy)
      • reintroduction of TMS as needed with early signs of relapse. In this context, several trials have assessed the durability of acute TMS benefit.

      A semi-controlled maintenance study followed 99 patients who had at least a 25% decrease in baseline HDRS score after acute TMS treatment.7 They were then tapered from their TMS sessions over 3 weeks while an antidepressant was titrated up. If, at any time during the subsequent 6 months, early signs of depression relapse were noted (ie, change of at least 1 point on the CGI-S for 2 consecutive weeks), TMS was reintroduced. At the end of the trial, 10 patients (13%) had relapsed and 38 (38%) had an exacerbation of symptoms sufficient to warrant reintroduction of TMS. Of those, 32 (84%) re-achieved mood stability.

      In another study, 50 patients who had achieved remission during an acute course of TMS were followed for 3 months.8 After TMS taper and continued pharmacotherapy or naturalistic follow-up, 29 (58%) remained in remission; 2 (4%) maintained partial response; and 1 (2%) relapsed.

       

       

      In a controlled, pilot, maintenance trial, 67 unmedicated patients with treatment-resistant depression received an acute course of TMS.9 Forty-seven of the responders were then randomized to a 1-year follow-up trial with or without a scheduled monthly TMS session. All patients could receive reintroduction TMS if they met criteria for symptom worsening.

      Both groups had a similar outcome. The number of patients who did not require TMS reintroduction was 9 of 23 (39%) in the scheduled TMS group vs 9 of 26 (35%) in the no-scheduled TMS group (P < .1). Although no difference was noted between groups, the authors commented that these preliminary results will help inform larger, more definitive trials. They concluded that both acute and maintenance TMS monotherapy might be an option—for some patients.

      A long-term, naturalistic outcomes study followed 257 treatment-resistant depressed patients for 1 year after they responded to an acute course of TMS.10 In addition to most patients receiving ongoing maintenance medication, they also could receive reintroduction of TMS if symptoms became worse.

      Compared with pre-TMS baseline, there was a statistically significant reduction in the mean total score on the CGI-S scale (primary outcome, P < .0001) at the end of acute treatment that was sustained at follow-up. Ninety-six patients (36.2%) required reintroduction of TMS and 75 of 120 (62.5%) who initially met response or remission criteria after acute treatment continued to meet response criteria after 1 year. The authors concluded that TMS demonstrated both a statistically and clinically meaningful durability of acute benefit during this time frame.


      TMS and electroconvulsive therapy
      For more than 75 years, ECT has consistently proved to be an effective treatment for major depressive disorder. Although the use of ECT has fluctuated over this period, one practice survey estimated that 100,000 patients receive ECT annually.11

      ECT has limitations, however, including cost, the need for general anesthesia, and cognitive deficits that range from short-term confusion to anterograde and retrograde amnesia, which can persist for weeks beyond active treatment.12 Despite increasing awareness of mental illness, stigma also remains a significant barrier to receiving ECT.

      TMS vs ECT. Several trials have directly compared ECT and TMS:

       

      • A recent meta-analysis of 9 trials included 384 patients with depression who were considered clinically appropriate for ECT and were randomized to one or the other treatment.13 Both modalities produced a significant reduction in baseline HDRS score, but ECT (15.4 point reduction) was superior to TMS (9.3 point reduction) in the degree of improvement (P < .01).
      • Another meta-analysis of 9 trials (N = 425) found ECT superior to TMS in terms of response (P < .03) and remission (P < .006) rates, based on improvement in the HDRS score.14 When psychotic depressed patients were excluded, however, TMS produced effects equivalent to ECT.

      In contrast to what was seen with ECT, cognitive testing of patients who received TMS revealed no deterioration in any domain. Furthermore, one of the comparison studies observed a modest, but statistically significant, improvement in patient’s working memory-executive function, objective memory, and fine-motor speed over the course of TMS treatment.15

      TMS plus ECT. A 2-week, randomized, single-blind, controlled pilot study (N = 22) examined the combination of TMS and ECT as acute treatment of depression.16 Patients were assigned to receive either unilateral non-dominant (UND) ECT 3 days a week or a combination of 1 UND ECT treatment followed by 4 days of TMS. At the conclusion of treatment, UND ECT plus TMS group produced comparable efficacy and fewer adverse effects compared with the UND ECT-only group.

      TMS maintenance after acute ECT response. Most patients who are referred for ECT have a depressive illness characterized by repeated episodes and incomplete response to pharmacotherapy or psychotherapy, or both. The need for an effective maintenance strategy after the acute response is therefore critical. Medication or ECT, or both, are commonly used to maintain acute benefit but, regrettably, a recent systematic review of the durability of benefit with such strategies found a substantial percentage (approximately 50%) of patients relapsed within the first year.17

       

      • In this context, a case series report found that 1 or 2 weekly, sequential, bilateral TMS treatments after a successful acute course of ECT maintained response in 5 of 6 patients over 6 to 12 months.18
      • Another case series (N = 6) transitioned stable patients from maintenance ECT to maintenance TMS, primarily because of adverse effects with ECT.19 With a mean frequency of 1 TMS treatment every 3.5 weeks, all 6 patients remained stable for as long as 6 months. Subsequently, 2 patients relapsed—1 at 8 months and 1 at 9 months.
       

       

      Advantages of maintenance TMS over maintenance ECT include lower cost, fewer adverse effects (particularly cognitive deficits), and the ability to remain independent during the period of the treatment sessions.

      TMS as an assessment tool for ECT response. TMS can be used to study excitability in cortical circuits. In a study, EEG potentials evoked by TMS before and after a course of ECT in 8 severely depressed patients revealed an increase in frontal cortical excitability, compared with baseline.20 Such findings support the ability of ECT to produce synaptic potentiation in humans. Furthermore, to the extent that depression presents with alterations in frontal cortical excitability, serial EEG-TMS measurements might be an effective tool to guide and monitor treatment progress with ECT, as well as other forms of therapeutic modulation.

      Summing up: TMS and ECT. Although a definitive comparative study is needed, available evidence suggests that TMS might be an alternative treatment in a subgroup of patients who are referred for ECT. Factors that might warrant considering TMS over ECT include:

       

      • patient preference
      • fear of anesthesia
      • concern about cognitive deficits
      • stigma.

      Although TMS might offer a workable alternative to ECT for acute and maintenance treatment of depression in selected patients, further refinement of the delivery of TMS is also needed to (1) enhance its efficacy and (2) identify clinical and biological markers to better define this select population.


      Standard TMS treatment parameters
      Superficial TMS. Superficial TMS for depression typically involves a single coil placed over the left dorsolateral prefrontal cortex. The standard, FDA-approved protocol includes stimulating at 110% of motor threshold with 75, 4-second trains at 10 Hz (ie, 40 stimulations) interspersed by 26-second intertrain intervals. Without interruption, a standard treatment session takes 37.5 minutes and delivers a total of 3,000 pulses. Most patients require 20 to 30 sessions, on a Monday-through-Friday schedule, to achieve optimal benefit.

      This approach stimulates to a depth of approximately 2 or 3 cm. The coil usually is placed over the left dorsolateral prefrontal cortex because earlier studies indicated that decreased activity in this part of the brain correlates with symptoms of depression. When TMS is administered in a rapid repetitive fashion (at >1 Hz; typically, at 10 Hz), blood flow and metabolism in that area of the brain are increased. In addition, imaging studies indicate that trans-synaptic connections with deeper parts of the brain also allow modulation of other relevant neural circuits.

      An alternate approach, less well-studied, involves low-frequency stimulation over the right dorsolateral prefrontal cortex. Parameters differ from what is used in left high-frequency dorsolateral prefrontal cortex TMS: frequency <1 Hz; train durations as long as 15 minutes; an intertrain interval of 25 to 180 seconds; 120 to 900 stimulations per train; and 2,400 to 18,000 total stimulations.

      One hypothesis is that this low-frequency approach selectively stimulates inhibitory interneurons, decreases local neuronal activity and diminishes blood flow to deeper structures, such as the amygdala. Although right low-frequency TMS, compared with left high-frequency TMS, has potential advantages of better tolerability and decreased risk for seizures, its relative efficacy is unclear.

      Deep TMS. Studies also are pursuing different coil configurations that allow for more direct stimulation of relevant structures (eg, prefrontal neuronal pathways associated with the reward system).

      One of these coil designs (ie, the H-coil), coupled to a Magstim TMS stimulator, recently received FDA clearance for treatment-resistant depression. In the pivotal, sham-controlled study, patients received 20 treatment sessions over 4 weeks.21 The treatment protocol consisted of a helmet-like coil placed over the medial and lateral prefrontal cortex. Stimulation parameters included an 18-Hz frequency; stimulation intensity of 120% motor threshold; stimulation train duration of 2 seconds; and an intertrain interval of 20 seconds. The treatment sessions lasted 20.2 minutes and delivered a total of 1,980 stimulations.

      Based on the 21-item HDRS, the active treatment coil group achieved a significantly greater decrease in baseline score (6.39 vs 3.28; P < .008); a greater response rate (37% vs 27.8%; P < .03); and a greater remission rate (30.4% vs 15.8%; P < .016) compared with the sham coil group.

      Next, in what is the only randomized, controlled maintenance assessment to date, the same patients were followed for an additional 12 weeks, continuing blinded treatments twice weekly. At the end of the second phase, the active treatment group also demonstrated greater benefit than the sham group (P < .03). One seizure did occur, possibly related to excessive alcohol use; but this raises the question of whether treating at a higher frequency (18 Hz) with greater depth and less focality might increase the risk of seizure.

       

       

      To assess the potential advantages, as well as the relative safety, of this approach over standard TMS delivery, an adequately designed and powered trial comparing the H-coil and a single-coil device is needed.


      Alternate TMS approaches
      Efforts to improve the clinical effectiveness of TMS for treating depression include several approaches.

      Theta burst stimulation (TBS) is a patterned form of TMS pulse delivery that utilizes high and low frequencies in the same stimulus train (eg, three 50-Hz bursts delivered 5 times a second). Such a pulse sequence can modulate long-term depression and long-term potentiation mechanisms that induce plasticity in areas such as the hippocampus.22

      Intermittent TBS (iTBS) administers stimulations over a relatively brief duration (eg, 2 seconds) or intermittently (eg, every 10 seconds) for a specific period (eg, 190 seconds [600 pulses in total]) over the left dorsolateral prefrontal cortex. This technique induces long-term potentiation and produces effects similar to those of high-frequency TMS.

      In contrast, continuous TBS (cTBS) administers a continuous train (eg, 40 seconds [600 total pulses]) over the right dorsolateral prefrontal cortex. This induces long-term depression and produces effects similar to low-frequency TMS.

      Recent studies using different delivery paradigms have generated mixed results:

      Study 1: Fifty-six patients with depression received active treatment; 17 others, a sham procedure.23 This study used 3 different conditions:

       

      • a combination of low-frequency and high-frequency TMS stimulation, administered over the right and left dorsolateral prefrontal cortices, respectively
      • a combination of iTBS over the left dorsolateral prefrontal cortex and cTBS over the right dorsolateral prefrontal cortex
      • a sham procedure, in which no magnetic field was created.

      Neither active treatment arm separated from the sham procedure based on change scores in the 21-item HDRS (P = not significant).

      Study 2: Sixty treatment-resistant depression patients were assigned to cTBS, iTBS, a combination of the 2 procedures, or a sham procedure.24 After 2 weeks, the active treatment arms produced the greatest benefit, based on change in scores on the 17-item HDRS, which differed significantly among the 4 groups (F value = 6.166; P < .001); the iTBS and combination arms demonstrated the most robust effect.

      There were also significantly more responders in the iTBS (40.0%) and combination groups (66.7%) than in the cTBS (25.0%) and sham groups (13.3%) (P < .010). A lower level of treatment refractoriness predicted a better outcome.

      Study 3: Twenty-nine depressed patients were randomized to cTBS over the right dorsolateral prefrontal cortex or a sham procedure.25 Overall, there was no difference between groups; however, actively treated patients who were unmedicated (n = 3) or remained on a stable dosage of medication during treatment (n = 8) did experience a significantly greater reduction in the HDRS score.

      Study 4: In a pilot trial, 32 depressed patients were randomized to 30 sessions of adjunctive combined iTBS plus cTBS or bilateral sham TBS.26 Based on reduction from the baseline Montgomery-Åsberg Depression Rating Scale score, 9 patients in the active treatment group and 4 in the sham group achieved response (odds ratio, 3.86; P < .048).

      If at least comparable efficacy can be clearly demonstrated, advantages of TBS over standard TMS include a significantly reduced administration time, which might allow for more patients to be treated and reduce associated costs of treatment.27

      Magnetic low-field synchronized stimulation is produced by rotating spherical rare-earth magnets that are synchronized to an individual’s alpha frequency. A recent 6-week, double-blind, sham-controlled trial (N = 202) reported that, in the intention-to-treat population, there was no difference in outcome between treatment arms. In patients who completed the study according to protocol (120 of 202), however, active treatment was significantly better in decreasing baseline HDRS score (P < .033).28

      Magnetic seizure therapy (MST) is an experimental approach to treating patients with more severe depression that is resistant to medical therapy. The primary aim is to use TMS to induce a seizure, thus achieving the same efficacy as provided by ECT but without the adverse cognitive effects of ECT. With MST, the TMS device uses much higher stimulation settings to produce a seizure—the goal being to avoid direct electrical current to the brain’s memory centers.29

      A pilot study considered the clinical and cognitive effects of MST in a group of 26 treatment-resistant depression patients (10 randomized; 16 open-label).30 Based on reduction in baseline HDRS scores at the end of the trial, 69% of patients achieved response and 46% met remission criteria; however, one-half of patients relapsed within 6 months.

      Importantly, no cognitive adverse effects were observed. Furthermore, the antidepressant and anti-anxiety effects of MST were associated with localized metabolic changes in brain areas implicated in the pathophysiology of depression.

       

       

      The investigators concluded that MST might constitute an effective, well-tolerated, and safe treatment for patients unable to benefit from available medical therapies for depression. In addition to confirmation of acute benefit in more definitive trials, the issue of durability of effect needs further clarification.


      TMS is a key component of neuropsychiatric practice
      It has been 3 decades since Barker et al31 developed the technology to deliver intense, localized magnetic pulses to specific areas of the nervous system. During this period, the role of TMS as a probe of the central and peripheral nervous systems has expanded to include various therapeutic applications, primarily focusing on treatment-resistant major depressive disorder.

      Now, increasing sophistication in the choice of stimulation parameters and other ongoing efforts to optimize the benefits of TMS are yielding improved clinical outcomes. Research is still needed to better define the place of TMS in the management of subtypes of depression that are particularly difficult to treat and that do not benefit adequately from medications or psychotherapy or their combination.

      Growing support from controlled trials, systematic reviews, meta-analyses, naturalistic outcome studies, and professional guidelines indicate that TMS has an increasingly important role in clinical practice.

       


      Bottom Line
      Clinical use of transcranial magnetic stimulation (TMS) continues to expand, particularly for treatment-resistant depression. At the same time, ongoing research seeks to refine the application of TMS to enhance and expand its benefits.

       


      Related Resources

       

      • www.Brainsway.com
      • www.Magstim.com
      • www.MagVenture.com

       

         


        Disclosures
        Dr. Janicak receives research support from Neuronetics, Inc; Sunovion Pharmaceuticals, Inc; and Ortho-McNeil-Janssen. He has served as a consultant to Neuronetics, Inc. Dr. Sackett, Ms. Kudrna, and Dr. Cutler report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
        References


        1. Janicak PG, Dowd SM, Rado JT, et al. The re-emerging role of therapeutic neuromodulation. Current Psychiatry. 2010;9(11):66-70,72-74.
        2. Janicak PG, Dokucu ME. Transcranial magnetic stimulation for the treatment of major depression. Neuropsychiatr Dis Treat. 2015;11:1549-1560.
        3. Gaynes BN, Lloyd SW, Lux L, et al. Repetitive transcranial magnetic stimulation for treatment-resistant depression: a systematic review and meta-analysis. J Clin Psychiatry. 2014;75(5):477-489; quiz 489.
        4. Liu B, Zhang Y, Zhang L, et al. Repetitive transcranial magnetic stimulation as an augmentative strategy for treatment-resistant depression, a meta-analysis of randomized, double-blind and sham-controlled study. BMC Psychiatry. 2014;14:342.
        5. Carpenter LL, Janicak PG, Aaronson ST, et al. Transcranial magnetic stimulation (TMS) for major depression: a multisite, naturalistic, observational study of acute treatment outcomes in clinical practice. Depress Anxiety. 2012;29(7):587-596.
        6. Janicak PG, Dunner DL, Aaronson ST, et al. Transcranial magnetic stimulation (TMS) for major depression: a multisite, naturalistic, observational study of quality of life outcome measures in clinical practice. CNS Spectr. 2013;18(6):322-332.
        7. Janicak PG, Nahas Z, Lisanby SH, et al. Durability of clinical benefit with transcranial magnetic stimulation (TMS) in the treatment of pharmacoresistant major depression: assessment of relapse during a 6-month, multisite, open-label study. Brain Stimul. 2010;3(4):187-199.
        8. Mantovani A, Pavlicova M, Avery D, et al. Long-term efficacy of repeated daily prefrontal transcranial magnetic stimulation (TMS) in treatment-resistant depression. Depress Anxiety. 2012;29(10):883-890.
        9. Philip NS, Dunner DL, Dowd SM, et al. Can medication free, treatment-resistant, depressed patients who initially respond to TMS be maintained off medications? A prospective, 12-month multisite randomized pilot study. Brain Stimul. 2016;9(2):251-257.
        10. Dunner DL, Aaronson ST, Sackeim HA, et al. A multisite, naturalistic, observational study of transcranial magnetic stimulation for patients with pharmacoresistant major depressive disorder: durability of benefit over a 1-year follow-up period. J Clin Psychiatry. 2014;75(12):1394-1401.
        11. Hermann RC, Dorwart RA, Hoover CW. Variation in ECT use in the United States. Am J Psychiatry. 1995;152(6):869-875.
        12. Sackeim HA. Memory and ECT: from polarization to reconciliation. J ECT. 2000;16(2):87-96.
        13. Micallef-Trigona B. Comparing the effects of repetitive transcranial magnetic stimulation and electroconvulsive therapy in the treatment of depression: a systematic review and meta-analysis. Depress Res Treat. 2014;2014:135049. doi: 10.1155/2014/135049.
        14. Ren J, Li H, Palaniyappan L, et al. Repetitive transcranial magnetic stimulation versus electroconvulsive therapy for major depression: a systematic review and meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry. 2014;51:181-189.
        15. Martis B, Alam D, Dowd SM, et al. Neurocognitive effects of repetitive transcranial magnetic stimulation in severe major depression. Clin Neurophysiol. 2003;114(6):1125-1132.
        16. Pridmore S, Rybak M, Turnier-Shea Y, et al. Comparison of transcranial magnetic stimulation and electroconvulsive therapy in depression. In: Miyoshi K, Shapiro CM, Gaviria M, et al, eds. Contemporary neuropsychiatry. Tokyo, Japan: Springer; 2001:237-241.
        17. Jelovac A, Kolshus E, McLoughlin DM. Relapse following successful electroconvulsive therapy for major depression: a meta-analysis. Neuropsychopharmacology. 2013;38(12):2467-2474.
        18. Noda Y, Daskalakis Z, Ramos C, et al. Repetitive transcranial magnetic stimulation to maintain treatment response to electroconvulsive therapy in depression: a case series. Front Psychiatry. 2013;4:73.
        19. Cristancho MA, Helmer A, Connolly R, et al. Transcranial magnetic stimulation maintenance as a substitute for maintenance electroconvulsive therapy: a case series. J ECT. 2013;29(2):106-108.
        20. Casarotto S, Canali P, Rosanova M, et al. Assessing the effects of electroconvulsive therapy on cortical excitability by means of transcranial magnetic stimulation and electroencephalography. Brain Topogr. 2013;26(2):326-337.
        21. 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(1):64-73.
        22. Daskalakis ZJ. Theta-burst transcranial magnetic stimulation in depression: when less may be more. Brain. 2014;137(pt 7):1860-1862.
        23. Prasser J, Schecklmann M, Poeppl TB, et al. Bilateral prefrontal rTMS and theta burst TMS as an add-on treatment for depression: a randomized placebo controlled trial. World J Biol Psychiatry. 2015;16(1):57-65.
        24. Li CT, Chen MH, Juan CH, et al. Efficacy of prefrontal theta-burst stimulation in refractory depression: a randomized sham-controlled study. Brain. 2014;137(pt 7):2088-2098.
        25. Chistyakov A, Kreinin B, Marmor S, et al. Preliminary assessment of the therapeutic efficacy of continuous theta-burst magnetic stimulation (cTBS) in major depression: a double-blind sham-controlled study. J Affect Disord. 2015;170:225-229.
        26. Plewnia C, Pasqualetti P, Große S, et al. Treatment of major depression with bilateral theta burst stimulation: a randomized controlled pilot trial. J Affect Disord. 2014;156:219-223.
        27. Chung SW, Hoy KE, Fitzgerald PB. Theta-burst stimulation: a new form of TMS treatment for depression? Depress Anxiety. 2015;32(3):182-192.
        28. Leuchter AF, Cook IA, Feifel D, et al. Efficacy and safety of low-field synchronized transcranial magnetic stimulation (sTMS) for treatment of major depression. Brain Stimul. 2015;8(4):787-794.
        29. Cretaz E, Brunoni AR, Lafer B. Magnetic seizure therapy for unipolar and bipolar depression: a systematic review. Neural Plast. 2015;2015:521398. doi: 10.1155/2015/521398.
        30. Kayser S, Bewernick BH, Matusch A, et al. Magnetic seizure therapy in treatment-resistant depression: clinical, neuropsychological and metabolic effects. Psychol Med. 2015;45(5):1073-1092.
        31. Barker AT, Jalinous R, Freeston IL. Non-invasive magnetic stimulation of human motor cortex. Lancet. 1985;1(8437):1106-1107.

        References


        1. Janicak PG, Dowd SM, Rado JT, et al. The re-emerging role of therapeutic neuromodulation. Current Psychiatry. 2010;9(11):66-70,72-74.
        2. Janicak PG, Dokucu ME. Transcranial magnetic stimulation for the treatment of major depression. Neuropsychiatr Dis Treat. 2015;11:1549-1560.
        3. Gaynes BN, Lloyd SW, Lux L, et al. Repetitive transcranial magnetic stimulation for treatment-resistant depression: a systematic review and meta-analysis. J Clin Psychiatry. 2014;75(5):477-489; quiz 489.
        4. Liu B, Zhang Y, Zhang L, et al. Repetitive transcranial magnetic stimulation as an augmentative strategy for treatment-resistant depression, a meta-analysis of randomized, double-blind and sham-controlled study. BMC Psychiatry. 2014;14:342.
        5. Carpenter LL, Janicak PG, Aaronson ST, et al. Transcranial magnetic stimulation (TMS) for major depression: a multisite, naturalistic, observational study of acute treatment outcomes in clinical practice. Depress Anxiety. 2012;29(7):587-596.
        6. Janicak PG, Dunner DL, Aaronson ST, et al. Transcranial magnetic stimulation (TMS) for major depression: a multisite, naturalistic, observational study of quality of life outcome measures in clinical practice. CNS Spectr. 2013;18(6):322-332.
        7. Janicak PG, Nahas Z, Lisanby SH, et al. Durability of clinical benefit with transcranial magnetic stimulation (TMS) in the treatment of pharmacoresistant major depression: assessment of relapse during a 6-month, multisite, open-label study. Brain Stimul. 2010;3(4):187-199.
        8. Mantovani A, Pavlicova M, Avery D, et al. Long-term efficacy of repeated daily prefrontal transcranial magnetic stimulation (TMS) in treatment-resistant depression. Depress Anxiety. 2012;29(10):883-890.
        9. Philip NS, Dunner DL, Dowd SM, et al. Can medication free, treatment-resistant, depressed patients who initially respond to TMS be maintained off medications? A prospective, 12-month multisite randomized pilot study. Brain Stimul. 2016;9(2):251-257.
        10. Dunner DL, Aaronson ST, Sackeim HA, et al. A multisite, naturalistic, observational study of transcranial magnetic stimulation for patients with pharmacoresistant major depressive disorder: durability of benefit over a 1-year follow-up period. J Clin Psychiatry. 2014;75(12):1394-1401.
        11. Hermann RC, Dorwart RA, Hoover CW. Variation in ECT use in the United States. Am J Psychiatry. 1995;152(6):869-875.
        12. Sackeim HA. Memory and ECT: from polarization to reconciliation. J ECT. 2000;16(2):87-96.
        13. Micallef-Trigona B. Comparing the effects of repetitive transcranial magnetic stimulation and electroconvulsive therapy in the treatment of depression: a systematic review and meta-analysis. Depress Res Treat. 2014;2014:135049. doi: 10.1155/2014/135049.
        14. Ren J, Li H, Palaniyappan L, et al. Repetitive transcranial magnetic stimulation versus electroconvulsive therapy for major depression: a systematic review and meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry. 2014;51:181-189.
        15. Martis B, Alam D, Dowd SM, et al. Neurocognitive effects of repetitive transcranial magnetic stimulation in severe major depression. Clin Neurophysiol. 2003;114(6):1125-1132.
        16. Pridmore S, Rybak M, Turnier-Shea Y, et al. Comparison of transcranial magnetic stimulation and electroconvulsive therapy in depression. In: Miyoshi K, Shapiro CM, Gaviria M, et al, eds. Contemporary neuropsychiatry. Tokyo, Japan: Springer; 2001:237-241.
        17. Jelovac A, Kolshus E, McLoughlin DM. Relapse following successful electroconvulsive therapy for major depression: a meta-analysis. Neuropsychopharmacology. 2013;38(12):2467-2474.
        18. Noda Y, Daskalakis Z, Ramos C, et al. Repetitive transcranial magnetic stimulation to maintain treatment response to electroconvulsive therapy in depression: a case series. Front Psychiatry. 2013;4:73.
        19. Cristancho MA, Helmer A, Connolly R, et al. Transcranial magnetic stimulation maintenance as a substitute for maintenance electroconvulsive therapy: a case series. J ECT. 2013;29(2):106-108.
        20. Casarotto S, Canali P, Rosanova M, et al. Assessing the effects of electroconvulsive therapy on cortical excitability by means of transcranial magnetic stimulation and electroencephalography. Brain Topogr. 2013;26(2):326-337.
        21. 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(1):64-73.
        22. Daskalakis ZJ. Theta-burst transcranial magnetic stimulation in depression: when less may be more. Brain. 2014;137(pt 7):1860-1862.
        23. Prasser J, Schecklmann M, Poeppl TB, et al. Bilateral prefrontal rTMS and theta burst TMS as an add-on treatment for depression: a randomized placebo controlled trial. World J Biol Psychiatry. 2015;16(1):57-65.
        24. Li CT, Chen MH, Juan CH, et al. Efficacy of prefrontal theta-burst stimulation in refractory depression: a randomized sham-controlled study. Brain. 2014;137(pt 7):2088-2098.
        25. Chistyakov A, Kreinin B, Marmor S, et al. Preliminary assessment of the therapeutic efficacy of continuous theta-burst magnetic stimulation (cTBS) in major depression: a double-blind sham-controlled study. J Affect Disord. 2015;170:225-229.
        26. Plewnia C, Pasqualetti P, Große S, et al. Treatment of major depression with bilateral theta burst stimulation: a randomized controlled pilot trial. J Affect Disord. 2014;156:219-223.
        27. Chung SW, Hoy KE, Fitzgerald PB. Theta-burst stimulation: a new form of TMS treatment for depression? Depress Anxiety. 2015;32(3):182-192.
        28. Leuchter AF, Cook IA, Feifel D, et al. Efficacy and safety of low-field synchronized transcranial magnetic stimulation (sTMS) for treatment of major depression. Brain Stimul. 2015;8(4):787-794.
        29. Cretaz E, Brunoni AR, Lafer B. Magnetic seizure therapy for unipolar and bipolar depression: a systematic review. Neural Plast. 2015;2015:521398. doi: 10.1155/2015/521398.
        30. Kayser S, Bewernick BH, Matusch A, et al. Magnetic seizure therapy in treatment-resistant depression: clinical, neuropsychological and metabolic effects. Psychol Med. 2015;45(5):1073-1092.
        31. Barker AT, Jalinous R, Freeston IL. Non-invasive magnetic stimulation of human motor cortex. Lancet. 1985;1(8437):1106-1107.

        Issue
        Current Psychiatry - 15(6)
        Issue
        Current Psychiatry - 15(6)
        Page Number
        49-56
        Page Number
        49-56
        Publications
        Publications
        Topics
        Article Type
        Display Headline
        Advances in transcranial magnetic stimulation for managing major depressive disorders
        Display Headline
        Advances in transcranial magnetic stimulation for managing major depressive disorders
        Legacy Keywords
        TMS, transcranial magnetic stimulation, major depressive disorders, MDD, depressive disorder, depressive disorder, depression, depressed, suicide, ECT, electroconvulsive therapy, theta burst stimulation, TBS, magnetic seizure therapy, MST
        Legacy Keywords
        TMS, transcranial magnetic stimulation, major depressive disorders, MDD, depressive disorder, depressive disorder, depression, depressed, suicide, ECT, electroconvulsive therapy, theta burst stimulation, TBS, magnetic seizure therapy, MST
        Sections
        Disallow All Ads
        Alternative CME
        Use ProPublica
        Article PDF Media
        Image
        Disable zoom
        Off

        The re-emerging role of therapeutic neuromodulation

        Article Type
        Changed
        Thu, 02/07/2019 - 12:28
        Display Headline
        The re-emerging role of therapeutic neuromodulation

        Discuss this article at http://currentpsychiatry.blogspot.com/2010/11/therapeutic-neuromodulation.html#comments

        The brain is an electrochemical organ, and its activity can be modulated for therapeutic purposes by electrical, pharmacologic, or combined approaches. In general, neuromodulation induces electrical current in peripheral or central nervous tissue, which is accomplished by various techniques, including:

        • electroconvulsive therapy (ECT)
        • vagus nerve stimulation (VNS)
        • transcranial magnetic stimulation (TMS)
        • deep brain stimulation (DBS).

        It is thought that therapeutic benefit occurs by regulating functional disturbances in relevant distributed neural circuits.1 Depending on the stimulation method, the frequencies chosen may excite or inhibit different or the same areas of the brain in varying patterns. Unlike medication, neuromodulation impacts the brain episodically, which may mitigate adaptation to the therapy’s beneficial effects and avoid systemic adverse effects.

        Neuromodulation techniques are categorized based on their risk level as invasive or noninvasive and seizurogenic or nonseizurogenic (Table 1). Although these and other approaches are being considered for various neuropsychiatric disorders (Table 2), the most common application is for severe, treatment-resistant depression. Therefore, this article focuses on FDA-approved neuromodulation treatments for depression, with limited discussion of other indications.

        Table 1

        Therapeutic neuromodulation: Categorization based on risk

         

        Noninvasive, nonseizurogenic
          TMS, tDCS, CES
        Noninvasive, seizurogenic
          ECT, MST, FEAST
        Invasive, nonseizurogenic
          VNS, DBS, EpCS
        CES: cranial electrotherapy stimulation; DBS: deep brain stimulation; ECT: electroconvulsive therapy; EpCS: epidural prefrontal cortical stimulation; FEAST: focal electrically administered seizure therapy; MST: magnetic seizure therapy; tDCS: transcranial direct current stimulation; TMS: transcranial magnetic stimulation; VNS: vagus nerve stimulation

        Table 2

        Approved and investigational indications of neuromodulation

         

        ApproachDescriptionClinical application
        CESUses small pulses of electrical current delivered across the head focused on the hypothalamic region with electrodes usually placed on the ear at the mastoid near the faceDepression
        Anxiety
        Sleep disorders
        DBS‘Functional neurosurgical’ procedure that uses electrical current to directly modulate specific areas of the CNSDepression
        OCD*
        Parkinson’s disease* Dystonia*
        ECTShort-term electrical stimulation sufficient to induce a seizureDepression*
        Schizophrenia
        Mania
        EpCSUses implantable stimulating paddles that do not come in contact with the brain and target the anterior frontal poles and the lateral prefrontal cortexDepression Pain
        FEASTAn alternate form of ECT that involves passage of electrical current unidirectionally from a small anode to a larger cathode electrodeDepression
        MSTIntense, high-frequency magnetic pulses sufficient to induce a seizureDepression
        tDCSSustained, low-intensity constant current flow usually passing from anode to cathode electrodes placed on the scalpDepression
        TMSUse of intense high- or low-frequency magnetic pulses to produce neuronal excitation or inhibitionDepression*
        PTSD
        OCD
        Schizophrenia Substance use disorders Tinnitus
        VNSUse of intermittent mild electrical pulses to the left vagus nerve, whose afferent fibers impact structures such as the locus ceruleus and the raphe nucleusEpilepsy*
        Depression*
        *FDA-approved indications
        CES: cranial electrotherapy stimulation; DBS: deep brain stimulation; ECT: electroconvulsive therapy; EpCS: epidural prefrontal cortical stimulation; FEAST: focal electrically administered seizure therapy; MST: magnetic seizure therapy; OCD: obsessive-compulsive disorder; PTSD: posttraumatic stress disorder; tDCS: transcranial direct current stimulation; TMS: transcranial magnetic stimulation; VNS: vagus nerve stimulation

        ECT: Oldest and most effective

        ECT has remained the most effective therapeutic neuromodulation technique for more than 7 decades. It is indicated primarily for severe depressive episodes (eg, psychotic, melancholic), particularly in older patients.

         

        ECT delivers electrical current to the CNS that is sufficient to produce a seizure. Under modified conditions, a typical course of 6 to 12 sessions can resolve severe depressive episodes and may also benefit other disorders, such as bipolar mania and acute psychosis. Although ECT is potentially life-saving, its use was markedly curtailed with the advent of effective antidepressants in the 1950s. Multiple factors impede its use, including:

         

        • access and expertise are limited in many areas
        • cognition is at least temporarily adversely affected
        • relapse rates after acute benefit are high
        • cost
        • public perception often is negative.

        Studies are addressing several of these concerns. For example, the National Institute of Mental Health-sponsored Consortium on Research with ECT (CORE) group is considering how to more effectively maintain acute benefits of ECT. They compared the potential merits of maintenance ECT with maintenance pharmacotherapy (nortriptyline plus lithium) over 6 months. Although the 2 strategies had comparable results, retention rates were <50% and about one-third relapsed in both groups.2,3 Potential alternative strategies include a more frequent ECT maintenance schedule and/or combining maintenance ECT with medication(s).

         

        Magnetic seizure therapy (MST) and focal electrically administered seizure therapy (FEAST) are attempts to produce similar efficacy and less cognitive disruption compared with ECT.4,5 Work also continues on electrode placement (eg, bifrontal) and alteration of waveform characteristics (eg, ultra-brief) to maintain or enhance efficacy while minimizing adverse effects.6,7

         

         

        Stimulating the vagus nerve

        VNS was introduced for treating refractory epilepsy in 1997. In 2005, it became the first FDA-approved implantable device for managing chronic or recurrent treatment-resistant depression.

        The vagus nerve is the principal parasympathetic, efferent tract regulating heart rate, intestinal motility, and gastric acid secretion. Information about pain, hunger, and satiety is conveyed by these fibers to the median raphe nucleus and locus coeruleus, brain regions with significant serotonergic and noradrenergic innervation. These neurotransmitters also are believed to play a pivotal role in major depression.

        With VNS, a pacemaker-like pulse generator is surgically implanted subcutaneously in the patient’s upper left chest. Wires extend from this device to the left vagus nerve (80% of whose fibers are afferent) located in the neck, to which the pulse generator sends electrical signals every few seconds (Table 3). The right vagus nerve is not used because it provides parasympathetic innervation to the heart. A clinician adjusts stimulation parameters using a computer and a noninvasive handheld device. Common adverse effects include voice alteration or hoarseness, cough, and shortness of breath, which occur during active stimulation because of the proximity of the electrodes to the laryngeal and pharyngeal branches of the vagus nerve. These effects may improve by adjusting stimulation intensity. The device permits a wide range of duty cycles, but preclinical animal studies indicate that >50% activation periods may damage the vagus nerve. If patients become too uncomfortable, they may deactivate the device with a magnet held over the implantation area.

        Two open-label studies evaluated VNS to treat major depression. The first involved 10 weeks of stimulation in 59 subjects with chronic or recurrent, nonpsychotic, unipolar or bipolar depression who failed at least 2 adequate antidepressant trials in the current episode.8 Stable doses of concomitant antidepressants or mood stabilizers were allowed. After 3 months, 18 (31%) patients responded within an average of 45.5 days, and nearly 15% achieved remission. Response was defined as 50% reduction in baseline Hamilton Depression Rating Scale-28 (HDRS-28) score; remission was defined as HDRS-28 score ≤10. Further, clinical response did not differ between unipolar and bipolar depression patients.

        In the second trial, 74 patients with treatment-resistant depression received fixed dose antidepressants and VNS for 3 months, followed by 9 months of flexibly dosed VNS and antidepressants.9 At 3 months, response (≥50% reduction in HDRS-28 score) and remission (HDRS-28 score <10) rates were 37% and 17%, respectively, and increased to 53% and 33% at 1 year.

        A sham-controlled trial of VNS in 235 depressed patients used similar inclusion and exclusion criteria as in the open-label study by Sackeim et al.8,10 Two weeks after device implantation, patients were randomized to active treatment (stimulator turned on) or sham control (stimulator left off). At 3 months, the primary outcome measure—response rate based on HDRS-24 score—did not differ significantly between the active and control groups (15% vs 10%, respectively). There was, however, a significantly greater improvement in Inventory of Depressive Symptomatology-Self Report Scale scores with active VNS vs sham VNS.

         

        Patients on sham treatment then were switched to active treatment and both groups were followed for 12 additional months, at which time response and remission rates nearly doubled for both groups.11 In a post-hoc analysis, the same investigators found significant improvement with VNS compared with a naturalistic, matched control group with similar treatment-resistant depression.12 The FDA considered this adequate to support efficacy and approved the device for chronic or recurrent treatment-resistant depression in an episode not responsive to at least 4 adequate treatment trials with pharmacotherapy or ECT. Perhaps because post-hoc analyses typically are not sufficient to gain FDA approval, most insurance companies do not reimburse for VNS treatment of depression, and VNS is not frequently used for refractory depression.

        Table 3

        Vagus nerve stimulation treatment parameters

         

        ParameterUnitsRangeMedian value at 12 months in pivotal study
        Output currentMilliamps (mA)0 to 3.51
        Signal frequencyHertz (Hz)1.3020
        Pulse widthMicroseconds (µsec)130 to 1,000500
        Duty cycle: ON time*Seconds7 to 6030
        Duty cycle: OFF time*Minutes0.2 to 1805
        *Stimulation cycle is 24 hours per day
        Source: Epilepsy patient’s manual for vagus nerve stimulation with the VNS Therapy™ system. Houston, TX: Cyberonics, Inc.; 2002, 2004. Depression physician’s manual. Houston, TX: Cyberonics, Inc.; 2005

        A newer option: TMS

        TMS is the most recently FDA-approved therapeutic neuromodulation technique for treating depression. In October 2008, a TMS device became available for patients failing to respond to 1 adequate antidepressant trial during the current episode.

        TMS delivers intense, intermittent magnetic pulses produced by an electrical charge into a ferromagnetic coil. The pulse intensity is similar to that produced by MRI. The coil usually is placed on the scalp over the left dorsolateral prefrontal cortex (DLPFC) and pulses are delivered in a rapid, repetitive train, causing neuronal depolarization in a small area of the adjacent cerebral cortex, as well as distal effects in other relevant neural circuits (Table 4). TMS typically is administered on an outpatient basis. A standard treatment course for depression consists of 5 treatment sessions per week for 4 to 8 weeks, depending on symptom severity and how quickly patients respond.

         

         

        TMS initially was examined in several small, open-label studies that looked at various treatment parameters and stimulation sites. Several sham-controlled studies generally found TMS efficacious and further refined treatment administration. Its role in treating depression—and possibly other psychiatric disorders—has been supported by 2 recent meta-analyses.13,14

        O’Reardon et al15 conducted the largest double-blind trial of active vs sham TMS (N=301) for moderately treatment-resistant major depression. This study began with a 4- to 6-week, blinded, randomized phase followed by 6 weeks of open-label TMS for initial nonresponders. The third phase reintroduced TMS over 6 months as needed to augment maintenance antidepressants. This trial utilized the most aggressive treatment parameters to date (ie, 10 Hz; 75 4-second trains; 26-second inter-train interval; 120% motor threshold) delivering 3,000 pulses per treatment over an average of 24 sessions. Compared with the sham procedure, patients who received active TMS showed significantly higher response rates on the Montgomery-Åsberg Depression Rating Scale (MADRS) at weeks 4 and 6. Similar results were found for the 17- and 24-item HDRS. At 6 weeks, remission rate—defined as a MADRS score <10—was significantly higher in the active treatment group (14%) compared with the sham procedure (6%). A post-hoc analysis found that the most robust benefit occurred in patients with only 1 failed adequate antidepressant trial (effect size=0.83).16 This administration protocol was well tolerated, with no deaths or seizures and a low rate of discontinuation because of adverse events (5%).17 The most common adverse effects were application site pain or discomfort and headaches.

         

        Recently, the second largest (N=190) sham-controlled trial of TMS for treatment-resistant major depression was published.18 This National Institute of Mental Health-sponsored, multiphase study included an initial 2-week, treatment-free period; 3 weeks of daily treatments over the left DLPFC using the same device and parameters as in the O’Reardon study; and an additional 3 weeks of treatment in patients who were improving. Those not responding to initial treatment were crossed over to open-label active TMS. This study advanced TMS development by:

        • using a novel somatosensory system that produced similar sensations with sham and active TMS
        • assessing the success of maintaining the blind
        • establishing a rigorous clinical rating system
        • utilizing MRI-guided adjustment of coil placement in a subset of patients.

        The authors concluded that active TMS was significantly better than sham treatment in achieving remission (14% vs 5%). In addition, the raters, treaters, and patients were effectively blinded to the treatment condition. MRI-assisted coil placement found that in 33% of the sample, site placement determined by standardized assessment was over the premotor cortex rather than the prefrontal cortex, so the coil was moved 1 additional cm anteriorly in these patients. Similar to those observed by O’Reardon et al, adverse effects of active TMS were generally mild to moderate, did not differ by treatment condition, and led to a low discontinuation rate (5.5%).

        Table 4

        Treatment parameters of transcranial magnetic stimulation

         

        ParameterComment
        Motor thresholdLowest intensity over primary motor cortex to produce contraction of the first dorsal interosseous or abductor pollicis brevis muscle; visual or electromyographically monitored
        Stimulus coil locationMost common: Left dorsolateral prefrontal cortex (DLPFC) Less common: Right DLPFC, vertex
        Stimulus pulse(s) or train 
          Intensity80% to 120% of MT
          Frequency≤1 to 20 Hz
          Duration≤1 millisecond
          Interpulse interval50 to 100 milliseconds
        Stimulus train duration3 to 6 seconds
        Inter-train interval20 to 60 seconds
        Source: Janicak PG, Krasuski J, Beedle D, et al. Transcranial magnetic stimulation for neuropsychiatric disorders. Psychiatr Times. 1999;16:56-63

        Deep brain stimulation

        DBS is a “functional neurosurgical” procedure that delivers electrical current directly to specific areas within the brain.19 Its mechanism of action remains uncertain; depolarization blockade, synaptic inhibition, and “neural jamming” are leading hypotheses. In contrast to conventional ablative surgeries, DBS is reversible and adjustable. Implantation involves positioning pacemaker-like battery devices subcutaneously in the left and right upper chest. Electrodes attached to wires are run subcutaneously behind the ears and, with stereotactic guidance, placed through burr holes in the skull into specific CNS areas implicated in the pathophysiology of conditions such as Parkinson’s disease, refractory depression, and severe obsessive-compulsive disorder (OCD).

         

        Antidepressant effects. The FDA recently approved DBS under its humanitarian device exemption program for intractable, severe, disabling OCD based on promising results from open and blind trials that stimulated areas such as the internal capsule and adjacent ventral striatum.20-22 These studies reported that DBS of the caudate nucleus for OCD and subthalamic nucleus for Parkinson’s disease also produced antidepressant effects. Subsequently, trials targeting the subgenual region (Brodmann’s area 25), the ventral capsule/ventral striatum, and nucleus accumbens demonstrated antidepressant effects.23-27 Pending the results of ongoing pilot trials, large, multi-center studies using different devices and target areas are being planned to clarify the role of DBS for patients with severe, disabling, refractory depression.

         

         

        Adverse effects of DBS can be:

        • surgical-related (eg, seizure, bleeding, infection)
        • device-related (eg, lead breakage, malfunction)
        • stimulation-related (eg, paresthesia, dysarthria, memory disruption, cognitive changes, psychiatric symptoms).

        The most serious risk is intracranial bleeding, which occurs in 2% to 3% of patients. Clearly, the risk-benefit ratio must be carefully considered.

        Cost and reimbursement

        Cost of treatment and potential for third-party reimbursement are important considerations for any risk-benefit analysis. Many patients who seek neuromodulation treatments will not have insurance or other coverage entitlements.28-30 Further, newer treatments are not routinely covered by insurance; however, individual case coverage may be allowed and some device manufacturers have programs to assist providers and patients obtain coverage.28-30 Even ECT, which has long been a covered treatment for major depression, is still considered investigational for other disorders. Thus, it is important to pre-certify with the patient’s health insurance provider before initiating treatment.

         

        Coverage, however, is not the only consideration when weighing cost effectiveness. Economic studies can assist with clinical and ethical decisions relating to treatment choice.31 These studies, however, need to be critically evaluated (eg, what costs were included in the analysis). Although direct costs are easier to evaluate, indirect costs—such as the patient’s ability to continue to work while receiving the treatment, caretaker availability during treatment, and whether treatment is an inpatient or outpatient procedure—are more difficult to evaluate and should be discussed with the patient. Because these specialized options have the potential to further benefit patients with depression and other neuropsychiatric disorders, it is essential to balance the pressures of cost containment with the need for more effective and better tolerated treatments.32-34

        Related Resource

        • Brunoni AR, Teng CT, Correa C, et al. Neuromodulation approaches for the treatment of major depression: challenges and recommendations from a working group meeting. Arq Neuropsiquiatr. 2010;68(3):433-451.

        Drug Brand Names

        • Lithium • Eskalith, Lithobid
        • Nortriptyline • Aventyl, Pamelor
        References

         

        1. Janicak PG, Pavuluri M, Marder S. Principles and practice of psychopharmacotherapy. 5th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 323-359. In press.

        2. Kellner CH, Knapp RG, Petrides G, et al. Continuation electroconvulsive therapy vs pharmacotherapy for relapse prevention in major depression. A multisite study from the Consortium for Research in Electroconvulsive Therapy (CORE). Arch Gen Psychiatry. 2006;63:1337-1344.

        3. Rasmussen KG, Mueller M, Rummans TA, et al. Is 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 Psychiatry. 2009;70(2):232-237.

        4. Spellman T, McClintock SM, Terrace H, et al. Differential effects of high-dose magnetic seizure therapy and electroconvulsive shock on cognitive function. Biol Psychiatry. 2008;63:1163-1170.

        5. Spellman T, Peterchev AV, Lisanby SH. Focal electrically administered seizure therapy: a novel form of ECT illustrates the roles of current directionality, polarity, and electrode configuration in seizure induction. Neuropsychopharmacology. 2009;34(8):2002-2010.

        6. Kellner CH, Knapp R, Husain MM, et al. Bifrontal, bitemporal and right unilateral electrode placement in ECT: randomised trial. Br J Psychiatry. 2010;196:226-234.

        7. 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 Stimulat. 2008;1:71-83.

        8. Sackeim HA, Rush JA, George MS, et al. Vagus nerve stimulation (VNSTM) for treatment-resistant depression: efficacy, side effects, and predictors of outcome. Neuropsychopharmacology. 2001;25(5):713-728.

        9. Schlaepfer TE, Frick C, Zobel A, et al. Vagus nerve stimulation for depression: efficacy and safety in a European study. Psychol Med. 2008;38(5):651-661.

        10. Rush AJ, Marangell LB, Sackeim HA, et al. Vagus nerve stimulation for treatment-resistant depression: a randomized controlled acute phase trial. Biol Psychiatry. 2005;58:347-354.

        11. Rush AJ, Sackeim HA, Marangell LB, et al. Effects of 12 months of vagus nerve stimulation in treatment resistant depression: a naturalistic study. Biol Psychiatry. 2005;58(5):355-363.

        12. George MS, Rush AJ, Marangell LB, et al. A one-year comparison of vagus nerve stimulation with treatment as usual for treatment-resistant depression. Biol Psychiatry. 2005;58:364-373.

        13. Schutter DJ. Antidepressant efficacy of high-frequency transcranial magnetic stimulation over the left dordolateral prefrontal cortex in double-blind sham-controlled designs: a meta-analysis. Psychol Med. 2009;39:65-75.

        14. Slotema CW, Blom JD, Hoek HW, et al. Should we expand the toolbox of psychiatric treatment methods to include repetitive transcranial magnetic stimulation (rTMS)? A meta-analysis of the efficacy of rTMS in psychiatric disorders. J Clin Psychiatry. 2010;71(7):873-884.

        15. O’Reardon JP, Solvason HB, Janicak PG, et al. Efficacy and safety of transcranial magnetic stimulation in the acute treatment of major depression: a multisite randomized controlled trial. Biol Psychiatry. 2007;62:1208-1216.

        16. 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(2):522-534.

        17. Janicak PG, O’Reardon JP, Sampson SM, et al. Transcranial magnetic stimulation in the treatment of major depressive disorder: a comprehensive summary of safety experience from acute exposure, extended exposure, and during reintroduction treatment. J Clin Psychiatry. 2008;69:222-232.

        18. George MS, Lisanby SH, Avery D, et al. Daily left prefrontal transcranial magnetic stimulation therapy for major depressive disorder: a sham controlled randomized trial. Arch Gen Psychiatry. 2010;67(5):507-516.

        19. Pilitsis JG, Bakay RAE. Deep brain stimulation for psychiatric disorders. Psychopharm Rev. 2007;42(9):67-74.

        20. Greenberg BD, Gabriels LA, Malone DA, et al. Deep brain stimulation of the ventral internal capsule/ventral striatum for obsessive-compulsive disorder: worldwide experience. Mol Psychiatry. 2010;15(1):64-79.

        21. Mallet L, Plolsan M, Jaafari N, et al. Subthalamic nucleus stimulation in severe obsessive-compulsive disorder. N Engl J Med. 2008;359:2121-2134.

        22. Goodman WK, Foote KD, Greenberg BD, et al. Deep brain stimulation for intractable obsessive compulsive disorder: pilot study using a blinded, staggered-onset design. Biol Psychiatry. 2010;67:535-542.

        23. Mayberg HS, Lozano AM, McNeely HE, et al. Deep brain stimulation for treatment-resistant depression. Neuron. 2005;45(5):651-660.

        24. Lozano AM, Mayberg HS, Giacobbe P, et al. Subcallosal cingulated gyrus deep brain stimulation for treatment-resistant depression. Biol Psychiatry. 2008;64:461-467.

        25. McNeely HE, Mayberg HS, Lozano AM, et al. Neuropsychological impact of Cg25 deep brain stimulation for treatment-resistant depression: preliminary results over 12 months. J Nerv Ment Dis. 2008;196(5):405-410.

        26. Malone DA, Dougherty DD, Rezai AR, et al. Deep brain stimulation of the ventral capsule/ventral striatum for treatment-resistant depression. Biol Psychiatry. 2009;65(4):267-275.

        27. Schlaepfer TE, Cohen MX, Frick C, et al. Deep brain stimulation to reward circuitry alleviates anhedonia in refractory major depression. Neuropsychopharmacology. 2008;33(2):368-377.

        28. Health insurance coverage NeuroStar TMS Therapy® Web site. Available at: http://www.neurostartms.com/TMSHealthInsurance/Health-Insurance-Coverage.aspx. Accessed June 2, 2010.

        29. VNS insurance information Vagus nerve stimulation therapy for treatment-resistant depression Web site. Available at: http://www.vnstherapy.com/depression/insuranceinformation/coverage.asp. Accessed June 2, 2010.

        30. Insurance coverage—DBS therapy for OCD Available at: http://www.medtronic.com/your-health/obsessive-compulsive-disorder-ocd/getting-therapy/insurance-coverage/index.htm. Accessed June 2, 2010.

        31. Simpson KN, Welch MJ, Kozel FA, et al. Cost-effectiveness of transcranial magnetic stimulation in the treatment of major depression: a health economics analysis. Adv Ther. 2009;26(3):346-368.

        32. Rado J, Dowd SM, Janicak PG. The emerging role of transcranial magnetic stimulation (TMS) for treatment of psychiatric disorders. Dir Psychiatry. 2008;28(25):215-331.

        33. Dougherty DD, Rauch SL. Somatic therapies for treatment-resistant depression: new neurotherapeutic interventions. Psychiatr Clin N Am. 2007;30:31-37.

        34. Olfson M, Marcus S, Sackeim HA, et al. Use of ECT for the inpatient treatment of recurrent major depression. Am J Psychiatry. 1998;155:22-29.

        Article PDF
        Author and Disclosure Information

        Philip G. Janicak, MD
        Professor, Department of Psychiatry, Rush University Medical Center, Chicago, IL
         

        Sheila M. Dowd, PhD
        Assistant Professor, Department of Psychiatry, Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL
         

        Jeffrey T. Rado, MD
        Assistant Professor, Department of Psychiatry and Medicine, Rush University Medical Center, Chicago, IL
         

        Mary Jane Welch, DNP, APRN, BC, CIP
        Assistant Professor, College of Nursing, Director, Human Subjects Protection, Rush University Medical Center, Chicago, IL

         

        DisclosuresDr. Janicak receives research/grant support from and is a consultant to and speaker for Bristol-Myers Squibb/Otsuka and Neuronetics, Inc. Dr. Dowd receives research/grant support from Neuronetics, Inc. and Otsuka and is a consultant to Neuronetics, Inc. Dr. Rado receives research/grant support from Eli Lilly and Company, Neuronetics, Inc., and Otsuka, is a consultant to Neuronetics, Inc., and is a speaker for Eli Lilly and Company. Dr. Welch reports no financial relationship with any company whose products are mentioned in this article, or with manufacturers of competing products.

        Issue
        Current Psychiatry - 09(11)
        Publications
        Topics
        Page Number
        66-70,72-74
        Sections
        Author and Disclosure Information

        Philip G. Janicak, MD
        Professor, Department of Psychiatry, Rush University Medical Center, Chicago, IL
         

        Sheila M. Dowd, PhD
        Assistant Professor, Department of Psychiatry, Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL
         

        Jeffrey T. Rado, MD
        Assistant Professor, Department of Psychiatry and Medicine, Rush University Medical Center, Chicago, IL
         

        Mary Jane Welch, DNP, APRN, BC, CIP
        Assistant Professor, College of Nursing, Director, Human Subjects Protection, Rush University Medical Center, Chicago, IL

         

        DisclosuresDr. Janicak receives research/grant support from and is a consultant to and speaker for Bristol-Myers Squibb/Otsuka and Neuronetics, Inc. Dr. Dowd receives research/grant support from Neuronetics, Inc. and Otsuka and is a consultant to Neuronetics, Inc. Dr. Rado receives research/grant support from Eli Lilly and Company, Neuronetics, Inc., and Otsuka, is a consultant to Neuronetics, Inc., and is a speaker for Eli Lilly and Company. Dr. Welch reports no financial relationship with any company whose products are mentioned in this article, or with manufacturers of competing products.

        Author and Disclosure Information

        Philip G. Janicak, MD
        Professor, Department of Psychiatry, Rush University Medical Center, Chicago, IL
         

        Sheila M. Dowd, PhD
        Assistant Professor, Department of Psychiatry, Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL
         

        Jeffrey T. Rado, MD
        Assistant Professor, Department of Psychiatry and Medicine, Rush University Medical Center, Chicago, IL
         

        Mary Jane Welch, DNP, APRN, BC, CIP
        Assistant Professor, College of Nursing, Director, Human Subjects Protection, Rush University Medical Center, Chicago, IL

         

        DisclosuresDr. Janicak receives research/grant support from and is a consultant to and speaker for Bristol-Myers Squibb/Otsuka and Neuronetics, Inc. Dr. Dowd receives research/grant support from Neuronetics, Inc. and Otsuka and is a consultant to Neuronetics, Inc. Dr. Rado receives research/grant support from Eli Lilly and Company, Neuronetics, Inc., and Otsuka, is a consultant to Neuronetics, Inc., and is a speaker for Eli Lilly and Company. Dr. Welch reports no financial relationship with any company whose products are mentioned in this article, or with manufacturers of competing products.

        Article PDF
        Article PDF

        Discuss this article at http://currentpsychiatry.blogspot.com/2010/11/therapeutic-neuromodulation.html#comments

        The brain is an electrochemical organ, and its activity can be modulated for therapeutic purposes by electrical, pharmacologic, or combined approaches. In general, neuromodulation induces electrical current in peripheral or central nervous tissue, which is accomplished by various techniques, including:

        • electroconvulsive therapy (ECT)
        • vagus nerve stimulation (VNS)
        • transcranial magnetic stimulation (TMS)
        • deep brain stimulation (DBS).

        It is thought that therapeutic benefit occurs by regulating functional disturbances in relevant distributed neural circuits.1 Depending on the stimulation method, the frequencies chosen may excite or inhibit different or the same areas of the brain in varying patterns. Unlike medication, neuromodulation impacts the brain episodically, which may mitigate adaptation to the therapy’s beneficial effects and avoid systemic adverse effects.

        Neuromodulation techniques are categorized based on their risk level as invasive or noninvasive and seizurogenic or nonseizurogenic (Table 1). Although these and other approaches are being considered for various neuropsychiatric disorders (Table 2), the most common application is for severe, treatment-resistant depression. Therefore, this article focuses on FDA-approved neuromodulation treatments for depression, with limited discussion of other indications.

        Table 1

        Therapeutic neuromodulation: Categorization based on risk

         

        Noninvasive, nonseizurogenic
          TMS, tDCS, CES
        Noninvasive, seizurogenic
          ECT, MST, FEAST
        Invasive, nonseizurogenic
          VNS, DBS, EpCS
        CES: cranial electrotherapy stimulation; DBS: deep brain stimulation; ECT: electroconvulsive therapy; EpCS: epidural prefrontal cortical stimulation; FEAST: focal electrically administered seizure therapy; MST: magnetic seizure therapy; tDCS: transcranial direct current stimulation; TMS: transcranial magnetic stimulation; VNS: vagus nerve stimulation

        Table 2

        Approved and investigational indications of neuromodulation

         

        ApproachDescriptionClinical application
        CESUses small pulses of electrical current delivered across the head focused on the hypothalamic region with electrodes usually placed on the ear at the mastoid near the faceDepression
        Anxiety
        Sleep disorders
        DBS‘Functional neurosurgical’ procedure that uses electrical current to directly modulate specific areas of the CNSDepression
        OCD*
        Parkinson’s disease* Dystonia*
        ECTShort-term electrical stimulation sufficient to induce a seizureDepression*
        Schizophrenia
        Mania
        EpCSUses implantable stimulating paddles that do not come in contact with the brain and target the anterior frontal poles and the lateral prefrontal cortexDepression Pain
        FEASTAn alternate form of ECT that involves passage of electrical current unidirectionally from a small anode to a larger cathode electrodeDepression
        MSTIntense, high-frequency magnetic pulses sufficient to induce a seizureDepression
        tDCSSustained, low-intensity constant current flow usually passing from anode to cathode electrodes placed on the scalpDepression
        TMSUse of intense high- or low-frequency magnetic pulses to produce neuronal excitation or inhibitionDepression*
        PTSD
        OCD
        Schizophrenia Substance use disorders Tinnitus
        VNSUse of intermittent mild electrical pulses to the left vagus nerve, whose afferent fibers impact structures such as the locus ceruleus and the raphe nucleusEpilepsy*
        Depression*
        *FDA-approved indications
        CES: cranial electrotherapy stimulation; DBS: deep brain stimulation; ECT: electroconvulsive therapy; EpCS: epidural prefrontal cortical stimulation; FEAST: focal electrically administered seizure therapy; MST: magnetic seizure therapy; OCD: obsessive-compulsive disorder; PTSD: posttraumatic stress disorder; tDCS: transcranial direct current stimulation; TMS: transcranial magnetic stimulation; VNS: vagus nerve stimulation

        ECT: Oldest and most effective

        ECT has remained the most effective therapeutic neuromodulation technique for more than 7 decades. It is indicated primarily for severe depressive episodes (eg, psychotic, melancholic), particularly in older patients.

         

        ECT delivers electrical current to the CNS that is sufficient to produce a seizure. Under modified conditions, a typical course of 6 to 12 sessions can resolve severe depressive episodes and may also benefit other disorders, such as bipolar mania and acute psychosis. Although ECT is potentially life-saving, its use was markedly curtailed with the advent of effective antidepressants in the 1950s. Multiple factors impede its use, including:

         

        • access and expertise are limited in many areas
        • cognition is at least temporarily adversely affected
        • relapse rates after acute benefit are high
        • cost
        • public perception often is negative.

        Studies are addressing several of these concerns. For example, the National Institute of Mental Health-sponsored Consortium on Research with ECT (CORE) group is considering how to more effectively maintain acute benefits of ECT. They compared the potential merits of maintenance ECT with maintenance pharmacotherapy (nortriptyline plus lithium) over 6 months. Although the 2 strategies had comparable results, retention rates were <50% and about one-third relapsed in both groups.2,3 Potential alternative strategies include a more frequent ECT maintenance schedule and/or combining maintenance ECT with medication(s).

         

        Magnetic seizure therapy (MST) and focal electrically administered seizure therapy (FEAST) are attempts to produce similar efficacy and less cognitive disruption compared with ECT.4,5 Work also continues on electrode placement (eg, bifrontal) and alteration of waveform characteristics (eg, ultra-brief) to maintain or enhance efficacy while minimizing adverse effects.6,7

         

         

        Stimulating the vagus nerve

        VNS was introduced for treating refractory epilepsy in 1997. In 2005, it became the first FDA-approved implantable device for managing chronic or recurrent treatment-resistant depression.

        The vagus nerve is the principal parasympathetic, efferent tract regulating heart rate, intestinal motility, and gastric acid secretion. Information about pain, hunger, and satiety is conveyed by these fibers to the median raphe nucleus and locus coeruleus, brain regions with significant serotonergic and noradrenergic innervation. These neurotransmitters also are believed to play a pivotal role in major depression.

        With VNS, a pacemaker-like pulse generator is surgically implanted subcutaneously in the patient’s upper left chest. Wires extend from this device to the left vagus nerve (80% of whose fibers are afferent) located in the neck, to which the pulse generator sends electrical signals every few seconds (Table 3). The right vagus nerve is not used because it provides parasympathetic innervation to the heart. A clinician adjusts stimulation parameters using a computer and a noninvasive handheld device. Common adverse effects include voice alteration or hoarseness, cough, and shortness of breath, which occur during active stimulation because of the proximity of the electrodes to the laryngeal and pharyngeal branches of the vagus nerve. These effects may improve by adjusting stimulation intensity. The device permits a wide range of duty cycles, but preclinical animal studies indicate that >50% activation periods may damage the vagus nerve. If patients become too uncomfortable, they may deactivate the device with a magnet held over the implantation area.

        Two open-label studies evaluated VNS to treat major depression. The first involved 10 weeks of stimulation in 59 subjects with chronic or recurrent, nonpsychotic, unipolar or bipolar depression who failed at least 2 adequate antidepressant trials in the current episode.8 Stable doses of concomitant antidepressants or mood stabilizers were allowed. After 3 months, 18 (31%) patients responded within an average of 45.5 days, and nearly 15% achieved remission. Response was defined as 50% reduction in baseline Hamilton Depression Rating Scale-28 (HDRS-28) score; remission was defined as HDRS-28 score ≤10. Further, clinical response did not differ between unipolar and bipolar depression patients.

        In the second trial, 74 patients with treatment-resistant depression received fixed dose antidepressants and VNS for 3 months, followed by 9 months of flexibly dosed VNS and antidepressants.9 At 3 months, response (≥50% reduction in HDRS-28 score) and remission (HDRS-28 score <10) rates were 37% and 17%, respectively, and increased to 53% and 33% at 1 year.

        A sham-controlled trial of VNS in 235 depressed patients used similar inclusion and exclusion criteria as in the open-label study by Sackeim et al.8,10 Two weeks after device implantation, patients were randomized to active treatment (stimulator turned on) or sham control (stimulator left off). At 3 months, the primary outcome measure—response rate based on HDRS-24 score—did not differ significantly between the active and control groups (15% vs 10%, respectively). There was, however, a significantly greater improvement in Inventory of Depressive Symptomatology-Self Report Scale scores with active VNS vs sham VNS.

         

        Patients on sham treatment then were switched to active treatment and both groups were followed for 12 additional months, at which time response and remission rates nearly doubled for both groups.11 In a post-hoc analysis, the same investigators found significant improvement with VNS compared with a naturalistic, matched control group with similar treatment-resistant depression.12 The FDA considered this adequate to support efficacy and approved the device for chronic or recurrent treatment-resistant depression in an episode not responsive to at least 4 adequate treatment trials with pharmacotherapy or ECT. Perhaps because post-hoc analyses typically are not sufficient to gain FDA approval, most insurance companies do not reimburse for VNS treatment of depression, and VNS is not frequently used for refractory depression.

        Table 3

        Vagus nerve stimulation treatment parameters

         

        ParameterUnitsRangeMedian value at 12 months in pivotal study
        Output currentMilliamps (mA)0 to 3.51
        Signal frequencyHertz (Hz)1.3020
        Pulse widthMicroseconds (µsec)130 to 1,000500
        Duty cycle: ON time*Seconds7 to 6030
        Duty cycle: OFF time*Minutes0.2 to 1805
        *Stimulation cycle is 24 hours per day
        Source: Epilepsy patient’s manual for vagus nerve stimulation with the VNS Therapy™ system. Houston, TX: Cyberonics, Inc.; 2002, 2004. Depression physician’s manual. Houston, TX: Cyberonics, Inc.; 2005

        A newer option: TMS

        TMS is the most recently FDA-approved therapeutic neuromodulation technique for treating depression. In October 2008, a TMS device became available for patients failing to respond to 1 adequate antidepressant trial during the current episode.

        TMS delivers intense, intermittent magnetic pulses produced by an electrical charge into a ferromagnetic coil. The pulse intensity is similar to that produced by MRI. The coil usually is placed on the scalp over the left dorsolateral prefrontal cortex (DLPFC) and pulses are delivered in a rapid, repetitive train, causing neuronal depolarization in a small area of the adjacent cerebral cortex, as well as distal effects in other relevant neural circuits (Table 4). TMS typically is administered on an outpatient basis. A standard treatment course for depression consists of 5 treatment sessions per week for 4 to 8 weeks, depending on symptom severity and how quickly patients respond.

         

         

        TMS initially was examined in several small, open-label studies that looked at various treatment parameters and stimulation sites. Several sham-controlled studies generally found TMS efficacious and further refined treatment administration. Its role in treating depression—and possibly other psychiatric disorders—has been supported by 2 recent meta-analyses.13,14

        O’Reardon et al15 conducted the largest double-blind trial of active vs sham TMS (N=301) for moderately treatment-resistant major depression. This study began with a 4- to 6-week, blinded, randomized phase followed by 6 weeks of open-label TMS for initial nonresponders. The third phase reintroduced TMS over 6 months as needed to augment maintenance antidepressants. This trial utilized the most aggressive treatment parameters to date (ie, 10 Hz; 75 4-second trains; 26-second inter-train interval; 120% motor threshold) delivering 3,000 pulses per treatment over an average of 24 sessions. Compared with the sham procedure, patients who received active TMS showed significantly higher response rates on the Montgomery-Åsberg Depression Rating Scale (MADRS) at weeks 4 and 6. Similar results were found for the 17- and 24-item HDRS. At 6 weeks, remission rate—defined as a MADRS score <10—was significantly higher in the active treatment group (14%) compared with the sham procedure (6%). A post-hoc analysis found that the most robust benefit occurred in patients with only 1 failed adequate antidepressant trial (effect size=0.83).16 This administration protocol was well tolerated, with no deaths or seizures and a low rate of discontinuation because of adverse events (5%).17 The most common adverse effects were application site pain or discomfort and headaches.

         

        Recently, the second largest (N=190) sham-controlled trial of TMS for treatment-resistant major depression was published.18 This National Institute of Mental Health-sponsored, multiphase study included an initial 2-week, treatment-free period; 3 weeks of daily treatments over the left DLPFC using the same device and parameters as in the O’Reardon study; and an additional 3 weeks of treatment in patients who were improving. Those not responding to initial treatment were crossed over to open-label active TMS. This study advanced TMS development by:

        • using a novel somatosensory system that produced similar sensations with sham and active TMS
        • assessing the success of maintaining the blind
        • establishing a rigorous clinical rating system
        • utilizing MRI-guided adjustment of coil placement in a subset of patients.

        The authors concluded that active TMS was significantly better than sham treatment in achieving remission (14% vs 5%). In addition, the raters, treaters, and patients were effectively blinded to the treatment condition. MRI-assisted coil placement found that in 33% of the sample, site placement determined by standardized assessment was over the premotor cortex rather than the prefrontal cortex, so the coil was moved 1 additional cm anteriorly in these patients. Similar to those observed by O’Reardon et al, adverse effects of active TMS were generally mild to moderate, did not differ by treatment condition, and led to a low discontinuation rate (5.5%).

        Table 4

        Treatment parameters of transcranial magnetic stimulation

         

        ParameterComment
        Motor thresholdLowest intensity over primary motor cortex to produce contraction of the first dorsal interosseous or abductor pollicis brevis muscle; visual or electromyographically monitored
        Stimulus coil locationMost common: Left dorsolateral prefrontal cortex (DLPFC) Less common: Right DLPFC, vertex
        Stimulus pulse(s) or train 
          Intensity80% to 120% of MT
          Frequency≤1 to 20 Hz
          Duration≤1 millisecond
          Interpulse interval50 to 100 milliseconds
        Stimulus train duration3 to 6 seconds
        Inter-train interval20 to 60 seconds
        Source: Janicak PG, Krasuski J, Beedle D, et al. Transcranial magnetic stimulation for neuropsychiatric disorders. Psychiatr Times. 1999;16:56-63

        Deep brain stimulation

        DBS is a “functional neurosurgical” procedure that delivers electrical current directly to specific areas within the brain.19 Its mechanism of action remains uncertain; depolarization blockade, synaptic inhibition, and “neural jamming” are leading hypotheses. In contrast to conventional ablative surgeries, DBS is reversible and adjustable. Implantation involves positioning pacemaker-like battery devices subcutaneously in the left and right upper chest. Electrodes attached to wires are run subcutaneously behind the ears and, with stereotactic guidance, placed through burr holes in the skull into specific CNS areas implicated in the pathophysiology of conditions such as Parkinson’s disease, refractory depression, and severe obsessive-compulsive disorder (OCD).

         

        Antidepressant effects. The FDA recently approved DBS under its humanitarian device exemption program for intractable, severe, disabling OCD based on promising results from open and blind trials that stimulated areas such as the internal capsule and adjacent ventral striatum.20-22 These studies reported that DBS of the caudate nucleus for OCD and subthalamic nucleus for Parkinson’s disease also produced antidepressant effects. Subsequently, trials targeting the subgenual region (Brodmann’s area 25), the ventral capsule/ventral striatum, and nucleus accumbens demonstrated antidepressant effects.23-27 Pending the results of ongoing pilot trials, large, multi-center studies using different devices and target areas are being planned to clarify the role of DBS for patients with severe, disabling, refractory depression.

         

         

        Adverse effects of DBS can be:

        • surgical-related (eg, seizure, bleeding, infection)
        • device-related (eg, lead breakage, malfunction)
        • stimulation-related (eg, paresthesia, dysarthria, memory disruption, cognitive changes, psychiatric symptoms).

        The most serious risk is intracranial bleeding, which occurs in 2% to 3% of patients. Clearly, the risk-benefit ratio must be carefully considered.

        Cost and reimbursement

        Cost of treatment and potential for third-party reimbursement are important considerations for any risk-benefit analysis. Many patients who seek neuromodulation treatments will not have insurance or other coverage entitlements.28-30 Further, newer treatments are not routinely covered by insurance; however, individual case coverage may be allowed and some device manufacturers have programs to assist providers and patients obtain coverage.28-30 Even ECT, which has long been a covered treatment for major depression, is still considered investigational for other disorders. Thus, it is important to pre-certify with the patient’s health insurance provider before initiating treatment.

         

        Coverage, however, is not the only consideration when weighing cost effectiveness. Economic studies can assist with clinical and ethical decisions relating to treatment choice.31 These studies, however, need to be critically evaluated (eg, what costs were included in the analysis). Although direct costs are easier to evaluate, indirect costs—such as the patient’s ability to continue to work while receiving the treatment, caretaker availability during treatment, and whether treatment is an inpatient or outpatient procedure—are more difficult to evaluate and should be discussed with the patient. Because these specialized options have the potential to further benefit patients with depression and other neuropsychiatric disorders, it is essential to balance the pressures of cost containment with the need for more effective and better tolerated treatments.32-34

        Related Resource

        • Brunoni AR, Teng CT, Correa C, et al. Neuromodulation approaches for the treatment of major depression: challenges and recommendations from a working group meeting. Arq Neuropsiquiatr. 2010;68(3):433-451.

        Drug Brand Names

        • Lithium • Eskalith, Lithobid
        • Nortriptyline • Aventyl, Pamelor

        Discuss this article at http://currentpsychiatry.blogspot.com/2010/11/therapeutic-neuromodulation.html#comments

        The brain is an electrochemical organ, and its activity can be modulated for therapeutic purposes by electrical, pharmacologic, or combined approaches. In general, neuromodulation induces electrical current in peripheral or central nervous tissue, which is accomplished by various techniques, including:

        • electroconvulsive therapy (ECT)
        • vagus nerve stimulation (VNS)
        • transcranial magnetic stimulation (TMS)
        • deep brain stimulation (DBS).

        It is thought that therapeutic benefit occurs by regulating functional disturbances in relevant distributed neural circuits.1 Depending on the stimulation method, the frequencies chosen may excite or inhibit different or the same areas of the brain in varying patterns. Unlike medication, neuromodulation impacts the brain episodically, which may mitigate adaptation to the therapy’s beneficial effects and avoid systemic adverse effects.

        Neuromodulation techniques are categorized based on their risk level as invasive or noninvasive and seizurogenic or nonseizurogenic (Table 1). Although these and other approaches are being considered for various neuropsychiatric disorders (Table 2), the most common application is for severe, treatment-resistant depression. Therefore, this article focuses on FDA-approved neuromodulation treatments for depression, with limited discussion of other indications.

        Table 1

        Therapeutic neuromodulation: Categorization based on risk

         

        Noninvasive, nonseizurogenic
          TMS, tDCS, CES
        Noninvasive, seizurogenic
          ECT, MST, FEAST
        Invasive, nonseizurogenic
          VNS, DBS, EpCS
        CES: cranial electrotherapy stimulation; DBS: deep brain stimulation; ECT: electroconvulsive therapy; EpCS: epidural prefrontal cortical stimulation; FEAST: focal electrically administered seizure therapy; MST: magnetic seizure therapy; tDCS: transcranial direct current stimulation; TMS: transcranial magnetic stimulation; VNS: vagus nerve stimulation

        Table 2

        Approved and investigational indications of neuromodulation

         

        ApproachDescriptionClinical application
        CESUses small pulses of electrical current delivered across the head focused on the hypothalamic region with electrodes usually placed on the ear at the mastoid near the faceDepression
        Anxiety
        Sleep disorders
        DBS‘Functional neurosurgical’ procedure that uses electrical current to directly modulate specific areas of the CNSDepression
        OCD*
        Parkinson’s disease* Dystonia*
        ECTShort-term electrical stimulation sufficient to induce a seizureDepression*
        Schizophrenia
        Mania
        EpCSUses implantable stimulating paddles that do not come in contact with the brain and target the anterior frontal poles and the lateral prefrontal cortexDepression Pain
        FEASTAn alternate form of ECT that involves passage of electrical current unidirectionally from a small anode to a larger cathode electrodeDepression
        MSTIntense, high-frequency magnetic pulses sufficient to induce a seizureDepression
        tDCSSustained, low-intensity constant current flow usually passing from anode to cathode electrodes placed on the scalpDepression
        TMSUse of intense high- or low-frequency magnetic pulses to produce neuronal excitation or inhibitionDepression*
        PTSD
        OCD
        Schizophrenia Substance use disorders Tinnitus
        VNSUse of intermittent mild electrical pulses to the left vagus nerve, whose afferent fibers impact structures such as the locus ceruleus and the raphe nucleusEpilepsy*
        Depression*
        *FDA-approved indications
        CES: cranial electrotherapy stimulation; DBS: deep brain stimulation; ECT: electroconvulsive therapy; EpCS: epidural prefrontal cortical stimulation; FEAST: focal electrically administered seizure therapy; MST: magnetic seizure therapy; OCD: obsessive-compulsive disorder; PTSD: posttraumatic stress disorder; tDCS: transcranial direct current stimulation; TMS: transcranial magnetic stimulation; VNS: vagus nerve stimulation

        ECT: Oldest and most effective

        ECT has remained the most effective therapeutic neuromodulation technique for more than 7 decades. It is indicated primarily for severe depressive episodes (eg, psychotic, melancholic), particularly in older patients.

         

        ECT delivers electrical current to the CNS that is sufficient to produce a seizure. Under modified conditions, a typical course of 6 to 12 sessions can resolve severe depressive episodes and may also benefit other disorders, such as bipolar mania and acute psychosis. Although ECT is potentially life-saving, its use was markedly curtailed with the advent of effective antidepressants in the 1950s. Multiple factors impede its use, including:

         

        • access and expertise are limited in many areas
        • cognition is at least temporarily adversely affected
        • relapse rates after acute benefit are high
        • cost
        • public perception often is negative.

        Studies are addressing several of these concerns. For example, the National Institute of Mental Health-sponsored Consortium on Research with ECT (CORE) group is considering how to more effectively maintain acute benefits of ECT. They compared the potential merits of maintenance ECT with maintenance pharmacotherapy (nortriptyline plus lithium) over 6 months. Although the 2 strategies had comparable results, retention rates were <50% and about one-third relapsed in both groups.2,3 Potential alternative strategies include a more frequent ECT maintenance schedule and/or combining maintenance ECT with medication(s).

         

        Magnetic seizure therapy (MST) and focal electrically administered seizure therapy (FEAST) are attempts to produce similar efficacy and less cognitive disruption compared with ECT.4,5 Work also continues on electrode placement (eg, bifrontal) and alteration of waveform characteristics (eg, ultra-brief) to maintain or enhance efficacy while minimizing adverse effects.6,7

         

         

        Stimulating the vagus nerve

        VNS was introduced for treating refractory epilepsy in 1997. In 2005, it became the first FDA-approved implantable device for managing chronic or recurrent treatment-resistant depression.

        The vagus nerve is the principal parasympathetic, efferent tract regulating heart rate, intestinal motility, and gastric acid secretion. Information about pain, hunger, and satiety is conveyed by these fibers to the median raphe nucleus and locus coeruleus, brain regions with significant serotonergic and noradrenergic innervation. These neurotransmitters also are believed to play a pivotal role in major depression.

        With VNS, a pacemaker-like pulse generator is surgically implanted subcutaneously in the patient’s upper left chest. Wires extend from this device to the left vagus nerve (80% of whose fibers are afferent) located in the neck, to which the pulse generator sends electrical signals every few seconds (Table 3). The right vagus nerve is not used because it provides parasympathetic innervation to the heart. A clinician adjusts stimulation parameters using a computer and a noninvasive handheld device. Common adverse effects include voice alteration or hoarseness, cough, and shortness of breath, which occur during active stimulation because of the proximity of the electrodes to the laryngeal and pharyngeal branches of the vagus nerve. These effects may improve by adjusting stimulation intensity. The device permits a wide range of duty cycles, but preclinical animal studies indicate that >50% activation periods may damage the vagus nerve. If patients become too uncomfortable, they may deactivate the device with a magnet held over the implantation area.

        Two open-label studies evaluated VNS to treat major depression. The first involved 10 weeks of stimulation in 59 subjects with chronic or recurrent, nonpsychotic, unipolar or bipolar depression who failed at least 2 adequate antidepressant trials in the current episode.8 Stable doses of concomitant antidepressants or mood stabilizers were allowed. After 3 months, 18 (31%) patients responded within an average of 45.5 days, and nearly 15% achieved remission. Response was defined as 50% reduction in baseline Hamilton Depression Rating Scale-28 (HDRS-28) score; remission was defined as HDRS-28 score ≤10. Further, clinical response did not differ between unipolar and bipolar depression patients.

        In the second trial, 74 patients with treatment-resistant depression received fixed dose antidepressants and VNS for 3 months, followed by 9 months of flexibly dosed VNS and antidepressants.9 At 3 months, response (≥50% reduction in HDRS-28 score) and remission (HDRS-28 score <10) rates were 37% and 17%, respectively, and increased to 53% and 33% at 1 year.

        A sham-controlled trial of VNS in 235 depressed patients used similar inclusion and exclusion criteria as in the open-label study by Sackeim et al.8,10 Two weeks after device implantation, patients were randomized to active treatment (stimulator turned on) or sham control (stimulator left off). At 3 months, the primary outcome measure—response rate based on HDRS-24 score—did not differ significantly between the active and control groups (15% vs 10%, respectively). There was, however, a significantly greater improvement in Inventory of Depressive Symptomatology-Self Report Scale scores with active VNS vs sham VNS.

         

        Patients on sham treatment then were switched to active treatment and both groups were followed for 12 additional months, at which time response and remission rates nearly doubled for both groups.11 In a post-hoc analysis, the same investigators found significant improvement with VNS compared with a naturalistic, matched control group with similar treatment-resistant depression.12 The FDA considered this adequate to support efficacy and approved the device for chronic or recurrent treatment-resistant depression in an episode not responsive to at least 4 adequate treatment trials with pharmacotherapy or ECT. Perhaps because post-hoc analyses typically are not sufficient to gain FDA approval, most insurance companies do not reimburse for VNS treatment of depression, and VNS is not frequently used for refractory depression.

        Table 3

        Vagus nerve stimulation treatment parameters

         

        ParameterUnitsRangeMedian value at 12 months in pivotal study
        Output currentMilliamps (mA)0 to 3.51
        Signal frequencyHertz (Hz)1.3020
        Pulse widthMicroseconds (µsec)130 to 1,000500
        Duty cycle: ON time*Seconds7 to 6030
        Duty cycle: OFF time*Minutes0.2 to 1805
        *Stimulation cycle is 24 hours per day
        Source: Epilepsy patient’s manual for vagus nerve stimulation with the VNS Therapy™ system. Houston, TX: Cyberonics, Inc.; 2002, 2004. Depression physician’s manual. Houston, TX: Cyberonics, Inc.; 2005

        A newer option: TMS

        TMS is the most recently FDA-approved therapeutic neuromodulation technique for treating depression. In October 2008, a TMS device became available for patients failing to respond to 1 adequate antidepressant trial during the current episode.

        TMS delivers intense, intermittent magnetic pulses produced by an electrical charge into a ferromagnetic coil. The pulse intensity is similar to that produced by MRI. The coil usually is placed on the scalp over the left dorsolateral prefrontal cortex (DLPFC) and pulses are delivered in a rapid, repetitive train, causing neuronal depolarization in a small area of the adjacent cerebral cortex, as well as distal effects in other relevant neural circuits (Table 4). TMS typically is administered on an outpatient basis. A standard treatment course for depression consists of 5 treatment sessions per week for 4 to 8 weeks, depending on symptom severity and how quickly patients respond.

         

         

        TMS initially was examined in several small, open-label studies that looked at various treatment parameters and stimulation sites. Several sham-controlled studies generally found TMS efficacious and further refined treatment administration. Its role in treating depression—and possibly other psychiatric disorders—has been supported by 2 recent meta-analyses.13,14

        O’Reardon et al15 conducted the largest double-blind trial of active vs sham TMS (N=301) for moderately treatment-resistant major depression. This study began with a 4- to 6-week, blinded, randomized phase followed by 6 weeks of open-label TMS for initial nonresponders. The third phase reintroduced TMS over 6 months as needed to augment maintenance antidepressants. This trial utilized the most aggressive treatment parameters to date (ie, 10 Hz; 75 4-second trains; 26-second inter-train interval; 120% motor threshold) delivering 3,000 pulses per treatment over an average of 24 sessions. Compared with the sham procedure, patients who received active TMS showed significantly higher response rates on the Montgomery-Åsberg Depression Rating Scale (MADRS) at weeks 4 and 6. Similar results were found for the 17- and 24-item HDRS. At 6 weeks, remission rate—defined as a MADRS score <10—was significantly higher in the active treatment group (14%) compared with the sham procedure (6%). A post-hoc analysis found that the most robust benefit occurred in patients with only 1 failed adequate antidepressant trial (effect size=0.83).16 This administration protocol was well tolerated, with no deaths or seizures and a low rate of discontinuation because of adverse events (5%).17 The most common adverse effects were application site pain or discomfort and headaches.

         

        Recently, the second largest (N=190) sham-controlled trial of TMS for treatment-resistant major depression was published.18 This National Institute of Mental Health-sponsored, multiphase study included an initial 2-week, treatment-free period; 3 weeks of daily treatments over the left DLPFC using the same device and parameters as in the O’Reardon study; and an additional 3 weeks of treatment in patients who were improving. Those not responding to initial treatment were crossed over to open-label active TMS. This study advanced TMS development by:

        • using a novel somatosensory system that produced similar sensations with sham and active TMS
        • assessing the success of maintaining the blind
        • establishing a rigorous clinical rating system
        • utilizing MRI-guided adjustment of coil placement in a subset of patients.

        The authors concluded that active TMS was significantly better than sham treatment in achieving remission (14% vs 5%). In addition, the raters, treaters, and patients were effectively blinded to the treatment condition. MRI-assisted coil placement found that in 33% of the sample, site placement determined by standardized assessment was over the premotor cortex rather than the prefrontal cortex, so the coil was moved 1 additional cm anteriorly in these patients. Similar to those observed by O’Reardon et al, adverse effects of active TMS were generally mild to moderate, did not differ by treatment condition, and led to a low discontinuation rate (5.5%).

        Table 4

        Treatment parameters of transcranial magnetic stimulation

         

        ParameterComment
        Motor thresholdLowest intensity over primary motor cortex to produce contraction of the first dorsal interosseous or abductor pollicis brevis muscle; visual or electromyographically monitored
        Stimulus coil locationMost common: Left dorsolateral prefrontal cortex (DLPFC) Less common: Right DLPFC, vertex
        Stimulus pulse(s) or train 
          Intensity80% to 120% of MT
          Frequency≤1 to 20 Hz
          Duration≤1 millisecond
          Interpulse interval50 to 100 milliseconds
        Stimulus train duration3 to 6 seconds
        Inter-train interval20 to 60 seconds
        Source: Janicak PG, Krasuski J, Beedle D, et al. Transcranial magnetic stimulation for neuropsychiatric disorders. Psychiatr Times. 1999;16:56-63

        Deep brain stimulation

        DBS is a “functional neurosurgical” procedure that delivers electrical current directly to specific areas within the brain.19 Its mechanism of action remains uncertain; depolarization blockade, synaptic inhibition, and “neural jamming” are leading hypotheses. In contrast to conventional ablative surgeries, DBS is reversible and adjustable. Implantation involves positioning pacemaker-like battery devices subcutaneously in the left and right upper chest. Electrodes attached to wires are run subcutaneously behind the ears and, with stereotactic guidance, placed through burr holes in the skull into specific CNS areas implicated in the pathophysiology of conditions such as Parkinson’s disease, refractory depression, and severe obsessive-compulsive disorder (OCD).

         

        Antidepressant effects. The FDA recently approved DBS under its humanitarian device exemption program for intractable, severe, disabling OCD based on promising results from open and blind trials that stimulated areas such as the internal capsule and adjacent ventral striatum.20-22 These studies reported that DBS of the caudate nucleus for OCD and subthalamic nucleus for Parkinson’s disease also produced antidepressant effects. Subsequently, trials targeting the subgenual region (Brodmann’s area 25), the ventral capsule/ventral striatum, and nucleus accumbens demonstrated antidepressant effects.23-27 Pending the results of ongoing pilot trials, large, multi-center studies using different devices and target areas are being planned to clarify the role of DBS for patients with severe, disabling, refractory depression.

         

         

        Adverse effects of DBS can be:

        • surgical-related (eg, seizure, bleeding, infection)
        • device-related (eg, lead breakage, malfunction)
        • stimulation-related (eg, paresthesia, dysarthria, memory disruption, cognitive changes, psychiatric symptoms).

        The most serious risk is intracranial bleeding, which occurs in 2% to 3% of patients. Clearly, the risk-benefit ratio must be carefully considered.

        Cost and reimbursement

        Cost of treatment and potential for third-party reimbursement are important considerations for any risk-benefit analysis. Many patients who seek neuromodulation treatments will not have insurance or other coverage entitlements.28-30 Further, newer treatments are not routinely covered by insurance; however, individual case coverage may be allowed and some device manufacturers have programs to assist providers and patients obtain coverage.28-30 Even ECT, which has long been a covered treatment for major depression, is still considered investigational for other disorders. Thus, it is important to pre-certify with the patient’s health insurance provider before initiating treatment.

         

        Coverage, however, is not the only consideration when weighing cost effectiveness. Economic studies can assist with clinical and ethical decisions relating to treatment choice.31 These studies, however, need to be critically evaluated (eg, what costs were included in the analysis). Although direct costs are easier to evaluate, indirect costs—such as the patient’s ability to continue to work while receiving the treatment, caretaker availability during treatment, and whether treatment is an inpatient or outpatient procedure—are more difficult to evaluate and should be discussed with the patient. Because these specialized options have the potential to further benefit patients with depression and other neuropsychiatric disorders, it is essential to balance the pressures of cost containment with the need for more effective and better tolerated treatments.32-34

        Related Resource

        • Brunoni AR, Teng CT, Correa C, et al. Neuromodulation approaches for the treatment of major depression: challenges and recommendations from a working group meeting. Arq Neuropsiquiatr. 2010;68(3):433-451.

        Drug Brand Names

        • Lithium • Eskalith, Lithobid
        • Nortriptyline • Aventyl, Pamelor
        References

         

        1. Janicak PG, Pavuluri M, Marder S. Principles and practice of psychopharmacotherapy. 5th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 323-359. In press.

        2. Kellner CH, Knapp RG, Petrides G, et al. Continuation electroconvulsive therapy vs pharmacotherapy for relapse prevention in major depression. A multisite study from the Consortium for Research in Electroconvulsive Therapy (CORE). Arch Gen Psychiatry. 2006;63:1337-1344.

        3. Rasmussen KG, Mueller M, Rummans TA, et al. Is 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 Psychiatry. 2009;70(2):232-237.

        4. Spellman T, McClintock SM, Terrace H, et al. Differential effects of high-dose magnetic seizure therapy and electroconvulsive shock on cognitive function. Biol Psychiatry. 2008;63:1163-1170.

        5. Spellman T, Peterchev AV, Lisanby SH. Focal electrically administered seizure therapy: a novel form of ECT illustrates the roles of current directionality, polarity, and electrode configuration in seizure induction. Neuropsychopharmacology. 2009;34(8):2002-2010.

        6. Kellner CH, Knapp R, Husain MM, et al. Bifrontal, bitemporal and right unilateral electrode placement in ECT: randomised trial. Br J Psychiatry. 2010;196:226-234.

        7. 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 Stimulat. 2008;1:71-83.

        8. Sackeim HA, Rush JA, George MS, et al. Vagus nerve stimulation (VNSTM) for treatment-resistant depression: efficacy, side effects, and predictors of outcome. Neuropsychopharmacology. 2001;25(5):713-728.

        9. Schlaepfer TE, Frick C, Zobel A, et al. Vagus nerve stimulation for depression: efficacy and safety in a European study. Psychol Med. 2008;38(5):651-661.

        10. Rush AJ, Marangell LB, Sackeim HA, et al. Vagus nerve stimulation for treatment-resistant depression: a randomized controlled acute phase trial. Biol Psychiatry. 2005;58:347-354.

        11. Rush AJ, Sackeim HA, Marangell LB, et al. Effects of 12 months of vagus nerve stimulation in treatment resistant depression: a naturalistic study. Biol Psychiatry. 2005;58(5):355-363.

        12. George MS, Rush AJ, Marangell LB, et al. A one-year comparison of vagus nerve stimulation with treatment as usual for treatment-resistant depression. Biol Psychiatry. 2005;58:364-373.

        13. Schutter DJ. Antidepressant efficacy of high-frequency transcranial magnetic stimulation over the left dordolateral prefrontal cortex in double-blind sham-controlled designs: a meta-analysis. Psychol Med. 2009;39:65-75.

        14. Slotema CW, Blom JD, Hoek HW, et al. Should we expand the toolbox of psychiatric treatment methods to include repetitive transcranial magnetic stimulation (rTMS)? A meta-analysis of the efficacy of rTMS in psychiatric disorders. J Clin Psychiatry. 2010;71(7):873-884.

        15. O’Reardon JP, Solvason HB, Janicak PG, et al. Efficacy and safety of transcranial magnetic stimulation in the acute treatment of major depression: a multisite randomized controlled trial. Biol Psychiatry. 2007;62:1208-1216.

        16. 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(2):522-534.

        17. Janicak PG, O’Reardon JP, Sampson SM, et al. Transcranial magnetic stimulation in the treatment of major depressive disorder: a comprehensive summary of safety experience from acute exposure, extended exposure, and during reintroduction treatment. J Clin Psychiatry. 2008;69:222-232.

        18. George MS, Lisanby SH, Avery D, et al. Daily left prefrontal transcranial magnetic stimulation therapy for major depressive disorder: a sham controlled randomized trial. Arch Gen Psychiatry. 2010;67(5):507-516.

        19. Pilitsis JG, Bakay RAE. Deep brain stimulation for psychiatric disorders. Psychopharm Rev. 2007;42(9):67-74.

        20. Greenberg BD, Gabriels LA, Malone DA, et al. Deep brain stimulation of the ventral internal capsule/ventral striatum for obsessive-compulsive disorder: worldwide experience. Mol Psychiatry. 2010;15(1):64-79.

        21. Mallet L, Plolsan M, Jaafari N, et al. Subthalamic nucleus stimulation in severe obsessive-compulsive disorder. N Engl J Med. 2008;359:2121-2134.

        22. Goodman WK, Foote KD, Greenberg BD, et al. Deep brain stimulation for intractable obsessive compulsive disorder: pilot study using a blinded, staggered-onset design. Biol Psychiatry. 2010;67:535-542.

        23. Mayberg HS, Lozano AM, McNeely HE, et al. Deep brain stimulation for treatment-resistant depression. Neuron. 2005;45(5):651-660.

        24. Lozano AM, Mayberg HS, Giacobbe P, et al. Subcallosal cingulated gyrus deep brain stimulation for treatment-resistant depression. Biol Psychiatry. 2008;64:461-467.

        25. McNeely HE, Mayberg HS, Lozano AM, et al. Neuropsychological impact of Cg25 deep brain stimulation for treatment-resistant depression: preliminary results over 12 months. J Nerv Ment Dis. 2008;196(5):405-410.

        26. Malone DA, Dougherty DD, Rezai AR, et al. Deep brain stimulation of the ventral capsule/ventral striatum for treatment-resistant depression. Biol Psychiatry. 2009;65(4):267-275.

        27. Schlaepfer TE, Cohen MX, Frick C, et al. Deep brain stimulation to reward circuitry alleviates anhedonia in refractory major depression. Neuropsychopharmacology. 2008;33(2):368-377.

        28. Health insurance coverage NeuroStar TMS Therapy® Web site. Available at: http://www.neurostartms.com/TMSHealthInsurance/Health-Insurance-Coverage.aspx. Accessed June 2, 2010.

        29. VNS insurance information Vagus nerve stimulation therapy for treatment-resistant depression Web site. Available at: http://www.vnstherapy.com/depression/insuranceinformation/coverage.asp. Accessed June 2, 2010.

        30. Insurance coverage—DBS therapy for OCD Available at: http://www.medtronic.com/your-health/obsessive-compulsive-disorder-ocd/getting-therapy/insurance-coverage/index.htm. Accessed June 2, 2010.

        31. Simpson KN, Welch MJ, Kozel FA, et al. Cost-effectiveness of transcranial magnetic stimulation in the treatment of major depression: a health economics analysis. Adv Ther. 2009;26(3):346-368.

        32. Rado J, Dowd SM, Janicak PG. The emerging role of transcranial magnetic stimulation (TMS) for treatment of psychiatric disorders. Dir Psychiatry. 2008;28(25):215-331.

        33. Dougherty DD, Rauch SL. Somatic therapies for treatment-resistant depression: new neurotherapeutic interventions. Psychiatr Clin N Am. 2007;30:31-37.

        34. Olfson M, Marcus S, Sackeim HA, et al. Use of ECT for the inpatient treatment of recurrent major depression. Am J Psychiatry. 1998;155:22-29.

        References

         

        1. Janicak PG, Pavuluri M, Marder S. Principles and practice of psychopharmacotherapy. 5th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 323-359. In press.

        2. Kellner CH, Knapp RG, Petrides G, et al. Continuation electroconvulsive therapy vs pharmacotherapy for relapse prevention in major depression. A multisite study from the Consortium for Research in Electroconvulsive Therapy (CORE). Arch Gen Psychiatry. 2006;63:1337-1344.

        3. Rasmussen KG, Mueller M, Rummans TA, et al. Is 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 Psychiatry. 2009;70(2):232-237.

        4. Spellman T, McClintock SM, Terrace H, et al. Differential effects of high-dose magnetic seizure therapy and electroconvulsive shock on cognitive function. Biol Psychiatry. 2008;63:1163-1170.

        5. Spellman T, Peterchev AV, Lisanby SH. Focal electrically administered seizure therapy: a novel form of ECT illustrates the roles of current directionality, polarity, and electrode configuration in seizure induction. Neuropsychopharmacology. 2009;34(8):2002-2010.

        6. Kellner CH, Knapp R, Husain MM, et al. Bifrontal, bitemporal and right unilateral electrode placement in ECT: randomised trial. Br J Psychiatry. 2010;196:226-234.

        7. 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 Stimulat. 2008;1:71-83.

        8. Sackeim HA, Rush JA, George MS, et al. Vagus nerve stimulation (VNSTM) for treatment-resistant depression: efficacy, side effects, and predictors of outcome. Neuropsychopharmacology. 2001;25(5):713-728.

        9. Schlaepfer TE, Frick C, Zobel A, et al. Vagus nerve stimulation for depression: efficacy and safety in a European study. Psychol Med. 2008;38(5):651-661.

        10. Rush AJ, Marangell LB, Sackeim HA, et al. Vagus nerve stimulation for treatment-resistant depression: a randomized controlled acute phase trial. Biol Psychiatry. 2005;58:347-354.

        11. Rush AJ, Sackeim HA, Marangell LB, et al. Effects of 12 months of vagus nerve stimulation in treatment resistant depression: a naturalistic study. Biol Psychiatry. 2005;58(5):355-363.

        12. George MS, Rush AJ, Marangell LB, et al. A one-year comparison of vagus nerve stimulation with treatment as usual for treatment-resistant depression. Biol Psychiatry. 2005;58:364-373.

        13. Schutter DJ. Antidepressant efficacy of high-frequency transcranial magnetic stimulation over the left dordolateral prefrontal cortex in double-blind sham-controlled designs: a meta-analysis. Psychol Med. 2009;39:65-75.

        14. Slotema CW, Blom JD, Hoek HW, et al. Should we expand the toolbox of psychiatric treatment methods to include repetitive transcranial magnetic stimulation (rTMS)? A meta-analysis of the efficacy of rTMS in psychiatric disorders. J Clin Psychiatry. 2010;71(7):873-884.

        15. O’Reardon JP, Solvason HB, Janicak PG, et al. Efficacy and safety of transcranial magnetic stimulation in the acute treatment of major depression: a multisite randomized controlled trial. Biol Psychiatry. 2007;62:1208-1216.

        16. 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(2):522-534.

        17. Janicak PG, O’Reardon JP, Sampson SM, et al. Transcranial magnetic stimulation in the treatment of major depressive disorder: a comprehensive summary of safety experience from acute exposure, extended exposure, and during reintroduction treatment. J Clin Psychiatry. 2008;69:222-232.

        18. George MS, Lisanby SH, Avery D, et al. Daily left prefrontal transcranial magnetic stimulation therapy for major depressive disorder: a sham controlled randomized trial. Arch Gen Psychiatry. 2010;67(5):507-516.

        19. Pilitsis JG, Bakay RAE. Deep brain stimulation for psychiatric disorders. Psychopharm Rev. 2007;42(9):67-74.

        20. Greenberg BD, Gabriels LA, Malone DA, et al. Deep brain stimulation of the ventral internal capsule/ventral striatum for obsessive-compulsive disorder: worldwide experience. Mol Psychiatry. 2010;15(1):64-79.

        21. Mallet L, Plolsan M, Jaafari N, et al. Subthalamic nucleus stimulation in severe obsessive-compulsive disorder. N Engl J Med. 2008;359:2121-2134.

        22. Goodman WK, Foote KD, Greenberg BD, et al. Deep brain stimulation for intractable obsessive compulsive disorder: pilot study using a blinded, staggered-onset design. Biol Psychiatry. 2010;67:535-542.

        23. Mayberg HS, Lozano AM, McNeely HE, et al. Deep brain stimulation for treatment-resistant depression. Neuron. 2005;45(5):651-660.

        24. Lozano AM, Mayberg HS, Giacobbe P, et al. Subcallosal cingulated gyrus deep brain stimulation for treatment-resistant depression. Biol Psychiatry. 2008;64:461-467.

        25. McNeely HE, Mayberg HS, Lozano AM, et al. Neuropsychological impact of Cg25 deep brain stimulation for treatment-resistant depression: preliminary results over 12 months. J Nerv Ment Dis. 2008;196(5):405-410.

        26. Malone DA, Dougherty DD, Rezai AR, et al. Deep brain stimulation of the ventral capsule/ventral striatum for treatment-resistant depression. Biol Psychiatry. 2009;65(4):267-275.

        27. Schlaepfer TE, Cohen MX, Frick C, et al. Deep brain stimulation to reward circuitry alleviates anhedonia in refractory major depression. Neuropsychopharmacology. 2008;33(2):368-377.

        28. Health insurance coverage NeuroStar TMS Therapy® Web site. Available at: http://www.neurostartms.com/TMSHealthInsurance/Health-Insurance-Coverage.aspx. Accessed June 2, 2010.

        29. VNS insurance information Vagus nerve stimulation therapy for treatment-resistant depression Web site. Available at: http://www.vnstherapy.com/depression/insuranceinformation/coverage.asp. Accessed June 2, 2010.

        30. Insurance coverage—DBS therapy for OCD Available at: http://www.medtronic.com/your-health/obsessive-compulsive-disorder-ocd/getting-therapy/insurance-coverage/index.htm. Accessed June 2, 2010.

        31. Simpson KN, Welch MJ, Kozel FA, et al. Cost-effectiveness of transcranial magnetic stimulation in the treatment of major depression: a health economics analysis. Adv Ther. 2009;26(3):346-368.

        32. Rado J, Dowd SM, Janicak PG. The emerging role of transcranial magnetic stimulation (TMS) for treatment of psychiatric disorders. Dir Psychiatry. 2008;28(25):215-331.

        33. Dougherty DD, Rauch SL. Somatic therapies for treatment-resistant depression: new neurotherapeutic interventions. Psychiatr Clin N Am. 2007;30:31-37.

        34. Olfson M, Marcus S, Sackeim HA, et al. Use of ECT for the inpatient treatment of recurrent major depression. Am J Psychiatry. 1998;155:22-29.

        Issue
        Current Psychiatry - 09(11)
        Issue
        Current Psychiatry - 09(11)
        Page Number
        66-70,72-74
        Page Number
        66-70,72-74
        Publications
        Publications
        Topics
        Article Type
        Display Headline
        The re-emerging role of therapeutic neuromodulation
        Display Headline
        The re-emerging role of therapeutic neuromodulation
        Sections
        PURLs Copyright

        Disallow All Ads
        Alternative CME
        Use ProPublica
        Article PDF Media
        Image
        Disable zoom
        Off

        Paliperidone ER: Reformulated antipsychotic for schizophrenia Tx

        Article Type
        Changed
        Tue, 12/11/2018 - 15:07
        Display Headline
        Paliperidone ER: Reformulated antipsychotic for schizophrenia Tx

        In the 9 months since paliperidone extended-release was FDA-approved for schizophrenia, the 3 acute pivotal trials supporting its approval have been published.1-3 They join a handful of post hoc analyses of this second-generation antipsychotic (SGA) in schizophrenia subgroups, including patients over age 65, recently diagnosed patients, and those with predominant negative symptoms.

        This article discusses the evidence and paliperidone ER’s probable clinical benefits and adverse effects, with focus on its:

        • pharmacodynamics and pharmacokinetics
        • potential efficacy in schizophrenia and for specific patients and symptoms
        • safety and tolerability.

        How does paliperidone ER work?

        Paliperidone ER was approved for schizophrenia treatment in December 2006 based on three 6-week, randomized, placebo-controlled trials. Paliperidone ER is the active metabolite of risperidone (9-OH risperidone) delivered in a once-daily, time-released formulation (Table 1).

        Pharmacodynamics. Similar to risperidone, paliperidone ER has high binding affinity for dopamine (D2) and serotonin (5-HT2A) receptors, with additional affinity for histaminic (H1) and adrenergic receptors (alpha1 and alpha2) but not for muscarinic-cholinergic receptors.

        Pharmacokinetics. After oral administration, the medication is widely and rapidly distributed. The drug’s terminal half-life is about 23 hours, and steady-state concentration is reached in 4 to 5 days.4,5

        Approximately 60% of the medication is eliminated renally and 11% is eliminated in the feces unchanged, with very limited hepatic metabolism.6 As a result, paliperidone ER appears to lack enzyme-inducing or inhibiting properties and does not substantially affect drugs that undergo cytochrome P-450 metabolism in the liver.

        Thus paliperidone ER—when compared with risperidone and other antipsychotics that are metabolized primarily in the liver—is less likely to be involved in hepatic drug-drug or drug-disease interactions. However, some drugs that can induce CYP-450 enzymes—such as carbamazepine—may affect paliperidone’s metabolism.7

        Paliperidone has an osmotic controlled-release oral delivery system (OROS®) for steady medication delivery across 24 hours8 (Table 2).1-3 The tablet consists of an osmotically active tri-layer core surrounded by a semipermeable membrane. When the tablet is swallowed, the membrane controls the rate of water reaching the tablet core, which determines the rate of drug delivery.6 The result is less variation between peak and trough drug concentrations, compared with immediate-release formulations.

        Table 1

        How paliperidone ER compares with risperidone

        CharacteristicPaliperidone ERRisperidone
        FormulationOROS extended-releaseImmediate release
        Active moiety9-OH risperidoneRisperidone plus 9-OH risperidone
        MetabolismPrimarily renalPrimarily hepatic
        Drug interactionsMinimalPrimarily through cytochrome P-450 enzyme 2D6
        DosingStart at target doseTitrate to target dose
        OROS: osmotic controlled-release oral delivery system
        Table 2

        Paliperidone ER’s clinical characteristics

        Second-generation antipsychotic approved for schizophrenia
        9-OH active metabolite of risperidone
        Osmotic controlled-release system provides steady-state drug delivery over 24 hours
        Terminal half-life (time for 50% of drug to be eliminated from the body) ~23 hours
        Available in 3-mg, 6-mg, and 9-mg tablets; recommended starting dose is 6 mg/d, and labeled dose range is 3 to 12 mg/d
        Excreted primarily by the kidney; maximum recommended dose for patients with oderate to severe renal impairment is 3 mg/d
        Source: References 1-3

        Clinical use

        Paliperidone ER offers potential therapeutic benefits in treating schizophrenia patients, although not without the risk of adverse events such as extrapyramidal symptoms (EPS) (Table 3).1-3

        Patient selection. Because of its slow-release formulation and relatively stable plasma concentrations, paliperidone ER might be useful for patients who are highly sensitive to antipsychotics’ side effects. In particular, paliperidone ER might be ideal for patients who respond to but may not tolerate risperidone.

        Paliperidone ER appears to be safe in patients with liver disease. Its primary renal excretion should minimize the risk of hepatic-related drug interactions in patients taking multiple medications.

        Dosage and titration. For treating schizophrenia, the suggested starting dose of paliperidone ER is 6 mg/d taken in the morning. In the 3 pivotal trials, 6 mg was the lowest dose to show broad efficacy with minimal adverse events.9

        For many patients, the 6-mg starting dose will be the therapeutic dose. When needed, the dose may be increased in 3-mg increments every 1 to 2 weeks to a maximum 12 mg/d (a 15-mg dose was used in clinical trials, but the adverse effects out-weighed the benefits). Lower maximum doses are recommended for patients with renal impairment:

        • 6 mg/d for those with creatinine clearance ≥50 to
        • 3 mg/d for those with creatinine clearance 10 to 10
        In the pivotal trials, differences in the terminal elimination half-life between hepatically impaired and healthy patients were minimal (26.5 hours vs 23.6 hours, respectively). Unbound paliperidone levels were slightly lower in patients with hepatic impairment11 but not low enough to recommend dose adjustment.

        Safety and tolerability. Pooled data from the 3 trials indicate that adverse events (AEs) occurred during treatment in 66% to 77% of patients receiving paliperidone ER vs 66% in placebo groups. The most common AEs were headache (11% to 18%), insomnia (4% to 12%), and anxiety (6% to 9%).9

         

         

        EPS. Risk of EPS-related AEs (such as akathisia and parkinsonian symptoms) with 3-mg and 6-mg paliperidone ER doses (13% and 10%, respectively) was similar to placebo (11%) but increased with the 9-mg, 12-mg, and 15-mg doses (25%, 26%, and 24%, respectively). Should EPS occur, reduce the paliperidone ER dose or consider adding antiparkinsonian medications.

        Lab values. No clinically relevant changes were noted in blood glucose, insulin, or lipids.12 Similar to risperidone, paliperidone ER elevated prolactin levels.

        Weight gain with paliperidone ER is dose-dependent; in the clinical trials, mean body weight change for all doses was ≤1.9 kg, which is similar to the weight gain seen with risperidone and in the moderate range compared with other SGAs. When using paliperidone ER, follow the American Diabetes Association/American Psychiatric Association guidelines13 for monitoring weight gain and metabolic parameters with antipsychotics. Also monitor patients for clinical symptoms of hyperprolactinemia, and—if intolerable—adjust the dose or switch to another SGA.

        Tachycardia. Advise patients that they may experience a rapid heart rate while taking paliperidone ER. In clinical trials, tachycardia occurred in ≤14% of patients—twice the rate with placebo—but did not contribute to more serious cardiac rhythm disturbances or to discontinuation. Incidence of prolonged corrected QT interval (QTc) was 3% to 5% in the paliperidone ER group vs 3% in the placebo group.

        Cost. Paliperidone ER costs approximately $12 to $18 per daily dose, which is similar to risperidone. Cost may be a greater consideration for patients next year, when generic risperidone becomes available (see Related Resources).

        Patient education. Because of paliperidone ER’s pharmacokinetic properties, counsel patients to:

        • take 1 tablet each day in the morning
        • not chew, split, or crush the tablets but swallow whole to preserve the controlled-release delivery.
        Also inform patients that they may see the tablet’s nonabsorbable shell in their stool as undigested residue.

        Table 3

        Paliperidone ER’s potential benefits and risks in clinical practice

        Potential benefitsDetails
        EfficacyData support acute (6 weeks) and chronic (up to 24 weeks) improvement in schizophrenia symptoms, patient function, and quality of life
        PharmacokineticsPrimarily renal excretion decreases risk of hepatic drug-drug or drug-disease interactions
        Long-acting formulationOnce-daily dosing simplifies treatment and may improve adherence
        EPSRisk similar to placebo at 3-mg and 6-mg doses, but increased at higher doses
        Weight gainSimilar to risperidone
        HyperprolactinemiaSimilar to risperidone
        TachycardiaOccurred in up to 14% of patients in clinical trials (twice the rate of placebo [7%])
        QTc prolongationIncrease up to 12 msec on average, with no patients exceeding 500 msec and no clinically adverse events during trials; use paliperidone with caution in patients with arrhythmias or cardiovascular disease or who are taking other medication that can prolong the QT interval
        EPS: extrapyramidal symptoms
        Source: References 1-3

        Efficacy trials in schizophrenia

        Three 6-week trials1-3 examined paliperidone ER’s efficacy in a total of 1,692 patients with chronic schizophrenia who were hospitalized ≥14 days with acute exacerbations. The trials were double-blind, randomized, fixed-dose, parallel-group, and placebo- and active-controlled (compared with olanzapine, 10 mg/d). Patients showed no significant differences in demographic or baseline characteristics or in the use of rescue medications.

        The primary outcome measure was mean change in Positive and Negative Syndrome Scale (PANSS) total score, which quantifies positive, negative, and global psychopathologic symptom severity. Secondary outcome measures included:

        • PANSS Marder factor scores14 (derived from PANSS items that reflect positive and negative symptoms, anxiety and depression, hostility, and thought disorganization).
        • Clinical Global Impressions-Severity (CGI-S) score, which measures overall illness severity.15
        • Personal and Social Performance (PSP) scores, which rate socially useful activities, relationships, self-care, and disturbing and aggressive behaviors; improvement by 1 category (10 points) reflects a clinically meaningful change.16,17
        The first study1 was conducted at 74 U.S. centers and enrolled 444 subjects (PANSS mean baseline score 94 ± 12). Patients were randomly assigned to fixed doses of paliperidone ER, 6 mg or 12 mg; placebo; or olanzapine, 10 mg/d. The olanzapine arm confirmed assay sensitivity and was not included in the efficacy analyses. Clinical response was defined as ≥30% improvement from baseline in total PANSS score.

        A total of 43% of patients completed the study—34% taking placebo; 46% taking paliperidone ER, 6 mg; 48% taking paliperidone ER, 12 mg; and 45% taking olanzapine. Demographic and baseline characteristics of the 432 patients who received ≥1 dose were similar across all groups. Approximately 75% of patients in each group used rescue medications—primarily lorazepam—for agitation, restlessness, or insomnia for a mean of 8 days.

         

         

        Patients taking either paliperidone ER dose showed statistically significant greater improvement in PANSS total score compared with those taking placebo (6 mg, P = 0.006; 12 mg, P

        Clinical response rates were similar with the 6-mg and 12-mg paliperidone ER doses—50% and 51%, respectively—and greater than with placebo (34%). The higher response rates with paliperidone ER were statistically significant compared with placebo (6 mg, P

        Discontinuation rates for lack of efficacy were lower with paliperidone ER (6 mg, 23%; 12 mg, 14%) than with placebo (35%). A substantially lower percentage of patients taking this agent remained classified as “marked/severe/extremely severe” on the CGI-S score from baseline to endpoint, compared with the placebo group;

        • 6 mg paliperidone ER, 58% to 26%
        • 12 mg paliperidone ER, 64% to 21%
        • placebo, 60% to 45%.
        PSP scores improved in both paliperidone ER groups, but the difference compared with placebo was statistically significant only for the 6-mg dose (P

        The second study2 included U.S. and international sites and compared 3 fixed doses of paliperidone ER (6-, 9-, and 12-mg) with placebo. Among the 630 patients enrolled, 66% completed the study. Patients were randomly assigned to 6 mg, 9 mg, or 12 mg of paliperidone ER; 10 mg of olanzapine; or placebo. The number of patients who dropped out because of adverse events was comparable across the groups.

        Patient groups assigned to paliperidone ER showed significant improvement when compared with placebo (P 30% reduction in PANSS total score from baseline to endpoint included:

        • 6 mg paliperidone ER, 56%
        • 9 mg paliperidone ER, 51%
        • 12 mg paliperidone ER, 61%
        • placebo, 30%.
        Discontinuation rates for lack of efficacy also were lower in the paliperidone ER groups vs placebo (6 mg, 16%; 9 mg, 16%; 12 mg, 10%; placebo, 40%). A substantially lower percentage of patients taking the drug remained classified as “marked/severe/extremely severe” by CGI-S score, compared with placebo:

        • 6 mg paliperidone ER, 63% at baseline to 22% at endpoint
        • 9 mg paliperidone ER, 58% to 23%
        • 12 mg paliperidone ER, 64% to 16%
        • placebo, 60% to 51%.
        PSP scores improved significantly for all 3 paliperidone ER doses vs placebo.

        The third study3 was a multicenter international trial that compared 3 fixed doses of paliperidone ER (3, 9, and 15 mg) with placebo. Among the 618 randomized patients, 365 (59%) completed the study: 70 of 127 (55%) on 3-mg paliperidone ER, 78 of 125 (62%) on 9-mg paliperidone ER, 82 of 115 (71%) on 15-mg paliperidone ER, and 47 of 123 (38%) on placebo.

        All 3 paliperidone ER doses were associated with statistically significant improvements in PANSS total and Marder factor scores at endpoint compared with placebo (P

        • 3 mg paliperidone ER, 40%
        • 9 mg paliperidone ER, 46%
        • 15 mg paliperidone ER, 53%
        • placebo, 18% (P ≤0.005).
        Discontinuation rates for lack of efficacy were lower in the paliperidone ER groups vs placebo and were dose-related (3 mg, 24%; 9 mg, 18%; 15 mg, 12%; placebo, 44%). Among patients taking olanzapine, 10 mg, 13% discontinued for lack of efficacy. Substantially fewer patients in the active drug groups were classified as “marked/severe/extremely severe” from baseline to end-point on the CGI-S scale vs the placebo group:

        • 3 mg paliperidone ER, 54% to 32%
        • 9 mg paliperidone ER, 52% to 23%
        • 15 mg paliperidone ER, 57% to 17%
        • placebo, 56% to 50%.
        Finally, a statistically significant improvement in mean PSP scores from baseline to endpoint was seen for all 3 paliperidone ER doses vs placebo (3 mg, 8.3 ± 17 points; 9 mg, 7.6 ± 14 points; 15 mg, 12 ± 15.7 points; placebo, 1.5 ± 16 points [P

        Additional trial evidence

        Schizophrenia subpopulations. Post hoc analyses of data reported from the 3 pivotal trials suggest that paliperidone ER may be useful for specific groups of schizophrenia patients, including those who are recently diagnosed, age >65, or severely ill or have predominant negative symptoms or sleep problems (Table 4).18-23

        So far, these analyses have been presented as posters at meetings or in sponsored supplements but have not been published in peer-reviewed publications.

        Efficacy in delaying recurrence. Paliperidone ER’s efficacy in delaying symptom recurrence was examined in a randomized, double-blind, placebo-controlled study of 207 patients who had been stabilized on open-label, flexible-dosed paliperidone ER.24 Time to first recurrence of schizophrenia symptoms was the primary efficacy measure. Starting dose was 9 mg/d (flexible dose range 3 to 15 mg/d).

         

         

        The study was halted at a planned interim analysis because time-to-recurrence was significantly longer for patients receiving paliperidone ER compared with placebo (P

        Final analysis of the 179 patients who completed the study confirmed the interim findings. Ongoing treatment maintained improvement in patients’ acute symptoms, functioning, and quality-of-life measures.

        Table 4

        Studies of paliperidone ER in schizophrenia subpopulations

        Patient populationStudy designFindings
        Recently diagnosed413 patients diagnosed within 5 years of study entry compared with 893 patients who had been ill ≥5 years*18,19Tolerability was similar, but recently diagnosed patients were more likely to experience movement disorders and somnolence
        Age ≥65 years114 schizophrenia patients age ≥65 given paliperidone ER, 3 to 12 mg/d, or placebo in 6-week, double-blind, randomized, placebo-controlled trial20Rates of cardiovascular, cerebrovascular, neuromotor, and metabolic changes similar to placebo, except for tachycardia (16% with paliperidone vs 0% with placebo)
        Severely ill217 patients with marked to severe symptoms (baseline total PANSS score ≥105)*21Patients treated with paliperidone showed significantly greater improvement vs placebo in mean total PANSS score (–26.7 vs –5.7) and other measures
        Substantial negative symptoms299 patients with predominant negative symptoms from 3 acute efficacy trials*22Patients treated with paliperidone showed significant improvements vs placebo on primary and secondary measures of negative symptoms
        Sleep problems36 patients age 18 to 45 diagnosed with schizophrenia and schizophrenia-related insomnia*23In stable patients, paliperidone improved sleep architecture, continuity, and patient-rated sleep quality without producing or worsening daytime sleepiness
        * Studies marked with asterisks represent post hoc analyses of data from the 3 clinical trials on which the FDA based its approval of paliperidone ER.
        PANSS: Positive and Negative Syndrome Scale
        Related resources

        Drug brand names

        • Carbamazepine • Tegretol
        • Lorazepam • Ativan
        • Olanzapine • Zyprexa
        • Paliperidone ER • Invega
        • Risperidone • Risperdal
        Disclosures

        Dr. Rado and Dr. Dowd receive research support from Neuronetics, sanofi-aventis, Janssen Pharmaceutica, and Solvay.

        Dr. Janicak receives research support from Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, Neuronetics, Solvay, and sanofi-aventis. He is a consultant to Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, Neuronetics, and Solvay, and a speaker for Abbott Laboratories, Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, and Pfizer.

        References

        1. Marder S, Kramer M, Ford L, et al. Efficacy and safety of paliperidone extended-release tablets: results of a 6-week, randomized, placebo-controlled study. Biol Psychiatry 2007; Jun 27; Epub ahead of print.

        2. Kane J, Canas F, Kramer M, et al. Treatment of schizophrenia with paliperidone extended-release tablets: a 6-week placebo-controlled trial. Schizophr Res 2007;90(1-3):147-61.

        3. Davidson M, Emsley R, Kramer M, et al. Efficacy, safety and early response of paliperidone extended-release tablets (paliperidone ER): results of a 6-week, randomized, placebo-controlled study. Schizophr Res 2007;93(1-3):117-30.

        4. Rossenu SAC, Rusch S, Janssens L, et al. Extended release formulation of paliperidone shows dose proportional pharmacokinetics. Presented at: Annual Meeting of the American Association of Pharmaceutical Scientists; October 29, 2006; San Antonio, TX.

        5. Vermeir M, Boom S, Naessens I, et al. Absorption, metabolism, and excretion of a single oral dose of 14C-paliperidone 1 mg in healthy subjects. Eur Neuropsychopharmacol 2005;15(suppl):S648-9.

        6. Conley R, Gupta SK, Sathyan G. Clinical spectrum of the osmotic-controlled release oral delivery system (OROS), an advanced oral delivery form. Curr Med Res Opin 2006;22(10):1879-92.

        7. Spina E, Avenoso A, Facciola G, et al. Plasma concentrations of risperidone and 9-hydroxyrisperidone: effect of comedication with carbamazepine or valproate. Ther Drug Monit 2000;22(4):481-5.

        8. Paliperidone extended release. Prescribing information. Available at: http://www.invega.com. Accessed August 8, 2007.

        9. Meltzer H, Kramer M, Gassmann-Mayer C, et al. Efficacy and tolerability of oral paliperidone extended-release tablets in the treatment of acute schizophrenia: pooled data from three 6-week placebo controlled studies. Int J Neuropsychopharmacol 2006;9(suppl 1):S225.-

        10. Thyssen A, Cleton A, Osselae NV, et al. Effects of renal impairment on the pharmacokinetic profile of paliperidone extended-release tablets. Clin Pharmacol Ther 2007. In press.

        11. Thyssen A, Crauwels H, Cleton A, et al. Effects of hepatic impairment on the pharmacokinetics of paliperidone immediate-release. Presented at: 46th Annual Meeting of the New Clinical Drug Evaluation Unit (NCDEU); June 12-15, 2006; Boca Raton, FL.

        12. Meyer J, Kramer M, Lane R, et al. Metabolic outcomes in patients with schizophrenia treated with oral paliperidone extended release tablets: pooled analysis of three 6 week placebo-controlled studies. Int J Neuropsychopharmacol 2006;9(suppl 1):S282.-

        13. American Diabetes Association, American Psychiatric Association, American Association of Clinical Endocrinologists, North American Association for the Study of Obesity. Consensus Development Conference on Antipsychotic Drugs and Obesity and Diabetes. J Clin Psychiatry 2004;65:267-72.

        14. Marder SR, Davis JM, Chouinard G. The effects of risperidone on the five dimensions of schizophrenia derived by factor analysis: combined results of the North American trials. J Clin Psychiatry 1997;58:538-46.

        15. Guy W. Clinical Global Impressions Scale. Early clinical drug evaluation unit (ECDEU) assessment manual for psychopharmacology. Rockville, MD: National Institute of Mental Health, Department of Health, Education, and Welfare; 1976:218-22. ADM publication 76-338.

        16. Morosini PL, Magliano L, Brambilla L, et al. Development, reliability and acceptability of a new version of the DSMIV Social and Occupational Functioning Assessment Scale (SOFAS) to assess routine social functioning. Acta Psychiatr Scand 2000;101:323-9.

        17. Patrick D, Adriaenssen I, Morosini P, Rothman M. Reliability, validity and sensitivity to change of the Personal and Social Performance scale in patients with acute schizophrenia. Int J Neuropsychopharmacol 2006;9(suppl 1):S287-8.

        18. Kostic D, Bossie C, Turkoz I, et al. Paliperidone extended-release tablets in patients recently diagnosed with schizophrenia. Int J Neuropsychopharmacol 2006;9(suppl 1):S161.-

        19. Kostic D, Bossie C, Turkoz I, et al. Paliperidone extended-release tablets in patients recently diagnosed with schizophrenia. Presented at: Congress of the Collegium Internationale Neruo-Psychopharmacologicum (CINP); July 9-13, 2006; Chicago, IL.

        20. Tzimos A, Kramer M, Ford L, et al. A 6-week placebo-controlled study of the safety and tolerability of flexible doses of oral paliperidone extended release tablets in the treatment of schizophrenia in elderly patients. Int J Neuropsychopharmacol 2006;9(suppl 1):S155.-

        21. Canuso C, Youssef E, Dirks B, et al. Paliperidone extended-release in severely-ill patients with schizophrenia. Presented at: 58th Annual Institute on Psychiatric Services; October 5-8, 2006; New York, NY.

        22. Dirks B, Eerdekens M, Turkoz I, et al. Efficacy of paliperidone extended-release tablets in patients with schizophrenia and predominant negative symptoms. Int J Neuropsychopharmacol 2006;9(suppl 1):S162.-

        23. Luthringer R, Staner L, Noel N, et al. Sleep assessments in patients with schizophrenia following treatment with paliperidone extended-release tablets. Eur Neuropsychopharmacol 2006;16(suppl 4):S224.-

        24. Kramer M, Simpson G, Maciulis V, et al. Paliperidone extended-release tablets for prevention of symptom recurrence in patients with schizophrenia: a randomized double-blind, placebo-controlled study [published correction appears in J Clin Psychopharmacol. 2007;27(3):258]. J Clin Psychopharmacol 2007;27(1):6-14.

        Article PDF
        Author and Disclosure Information

        Jeffrey Rado, MD
        Assistant professor

        Sheila M. Dowd, PhD
        Assistant professor

        Philip G. Janicak, MD
        Professor

        Department of psychiatry, Rush University Medical Center, Chicago

        Issue
        Current Psychiatry - 06(09)
        Publications
        Page Number
        75-82
        Legacy Keywords
        paliperidone; Invega; schizophrenia; second-generation antipsychotic; SGA; Jeffrey Rado MD; Sheila M. Dowd PhD; Philip G. Janicak MD
        Sections
        Author and Disclosure Information

        Jeffrey Rado, MD
        Assistant professor

        Sheila M. Dowd, PhD
        Assistant professor

        Philip G. Janicak, MD
        Professor

        Department of psychiatry, Rush University Medical Center, Chicago

        Author and Disclosure Information

        Jeffrey Rado, MD
        Assistant professor

        Sheila M. Dowd, PhD
        Assistant professor

        Philip G. Janicak, MD
        Professor

        Department of psychiatry, Rush University Medical Center, Chicago

        Article PDF
        Article PDF

        In the 9 months since paliperidone extended-release was FDA-approved for schizophrenia, the 3 acute pivotal trials supporting its approval have been published.1-3 They join a handful of post hoc analyses of this second-generation antipsychotic (SGA) in schizophrenia subgroups, including patients over age 65, recently diagnosed patients, and those with predominant negative symptoms.

        This article discusses the evidence and paliperidone ER’s probable clinical benefits and adverse effects, with focus on its:

        • pharmacodynamics and pharmacokinetics
        • potential efficacy in schizophrenia and for specific patients and symptoms
        • safety and tolerability.

        How does paliperidone ER work?

        Paliperidone ER was approved for schizophrenia treatment in December 2006 based on three 6-week, randomized, placebo-controlled trials. Paliperidone ER is the active metabolite of risperidone (9-OH risperidone) delivered in a once-daily, time-released formulation (Table 1).

        Pharmacodynamics. Similar to risperidone, paliperidone ER has high binding affinity for dopamine (D2) and serotonin (5-HT2A) receptors, with additional affinity for histaminic (H1) and adrenergic receptors (alpha1 and alpha2) but not for muscarinic-cholinergic receptors.

        Pharmacokinetics. After oral administration, the medication is widely and rapidly distributed. The drug’s terminal half-life is about 23 hours, and steady-state concentration is reached in 4 to 5 days.4,5

        Approximately 60% of the medication is eliminated renally and 11% is eliminated in the feces unchanged, with very limited hepatic metabolism.6 As a result, paliperidone ER appears to lack enzyme-inducing or inhibiting properties and does not substantially affect drugs that undergo cytochrome P-450 metabolism in the liver.

        Thus paliperidone ER—when compared with risperidone and other antipsychotics that are metabolized primarily in the liver—is less likely to be involved in hepatic drug-drug or drug-disease interactions. However, some drugs that can induce CYP-450 enzymes—such as carbamazepine—may affect paliperidone’s metabolism.7

        Paliperidone has an osmotic controlled-release oral delivery system (OROS®) for steady medication delivery across 24 hours8 (Table 2).1-3 The tablet consists of an osmotically active tri-layer core surrounded by a semipermeable membrane. When the tablet is swallowed, the membrane controls the rate of water reaching the tablet core, which determines the rate of drug delivery.6 The result is less variation between peak and trough drug concentrations, compared with immediate-release formulations.

        Table 1

        How paliperidone ER compares with risperidone

        CharacteristicPaliperidone ERRisperidone
        FormulationOROS extended-releaseImmediate release
        Active moiety9-OH risperidoneRisperidone plus 9-OH risperidone
        MetabolismPrimarily renalPrimarily hepatic
        Drug interactionsMinimalPrimarily through cytochrome P-450 enzyme 2D6
        DosingStart at target doseTitrate to target dose
        OROS: osmotic controlled-release oral delivery system
        Table 2

        Paliperidone ER’s clinical characteristics

        Second-generation antipsychotic approved for schizophrenia
        9-OH active metabolite of risperidone
        Osmotic controlled-release system provides steady-state drug delivery over 24 hours
        Terminal half-life (time for 50% of drug to be eliminated from the body) ~23 hours
        Available in 3-mg, 6-mg, and 9-mg tablets; recommended starting dose is 6 mg/d, and labeled dose range is 3 to 12 mg/d
        Excreted primarily by the kidney; maximum recommended dose for patients with oderate to severe renal impairment is 3 mg/d
        Source: References 1-3

        Clinical use

        Paliperidone ER offers potential therapeutic benefits in treating schizophrenia patients, although not without the risk of adverse events such as extrapyramidal symptoms (EPS) (Table 3).1-3

        Patient selection. Because of its slow-release formulation and relatively stable plasma concentrations, paliperidone ER might be useful for patients who are highly sensitive to antipsychotics’ side effects. In particular, paliperidone ER might be ideal for patients who respond to but may not tolerate risperidone.

        Paliperidone ER appears to be safe in patients with liver disease. Its primary renal excretion should minimize the risk of hepatic-related drug interactions in patients taking multiple medications.

        Dosage and titration. For treating schizophrenia, the suggested starting dose of paliperidone ER is 6 mg/d taken in the morning. In the 3 pivotal trials, 6 mg was the lowest dose to show broad efficacy with minimal adverse events.9

        For many patients, the 6-mg starting dose will be the therapeutic dose. When needed, the dose may be increased in 3-mg increments every 1 to 2 weeks to a maximum 12 mg/d (a 15-mg dose was used in clinical trials, but the adverse effects out-weighed the benefits). Lower maximum doses are recommended for patients with renal impairment:

        • 6 mg/d for those with creatinine clearance ≥50 to
        • 3 mg/d for those with creatinine clearance 10 to 10
        In the pivotal trials, differences in the terminal elimination half-life between hepatically impaired and healthy patients were minimal (26.5 hours vs 23.6 hours, respectively). Unbound paliperidone levels were slightly lower in patients with hepatic impairment11 but not low enough to recommend dose adjustment.

        Safety and tolerability. Pooled data from the 3 trials indicate that adverse events (AEs) occurred during treatment in 66% to 77% of patients receiving paliperidone ER vs 66% in placebo groups. The most common AEs were headache (11% to 18%), insomnia (4% to 12%), and anxiety (6% to 9%).9

         

         

        EPS. Risk of EPS-related AEs (such as akathisia and parkinsonian symptoms) with 3-mg and 6-mg paliperidone ER doses (13% and 10%, respectively) was similar to placebo (11%) but increased with the 9-mg, 12-mg, and 15-mg doses (25%, 26%, and 24%, respectively). Should EPS occur, reduce the paliperidone ER dose or consider adding antiparkinsonian medications.

        Lab values. No clinically relevant changes were noted in blood glucose, insulin, or lipids.12 Similar to risperidone, paliperidone ER elevated prolactin levels.

        Weight gain with paliperidone ER is dose-dependent; in the clinical trials, mean body weight change for all doses was ≤1.9 kg, which is similar to the weight gain seen with risperidone and in the moderate range compared with other SGAs. When using paliperidone ER, follow the American Diabetes Association/American Psychiatric Association guidelines13 for monitoring weight gain and metabolic parameters with antipsychotics. Also monitor patients for clinical symptoms of hyperprolactinemia, and—if intolerable—adjust the dose or switch to another SGA.

        Tachycardia. Advise patients that they may experience a rapid heart rate while taking paliperidone ER. In clinical trials, tachycardia occurred in ≤14% of patients—twice the rate with placebo—but did not contribute to more serious cardiac rhythm disturbances or to discontinuation. Incidence of prolonged corrected QT interval (QTc) was 3% to 5% in the paliperidone ER group vs 3% in the placebo group.

        Cost. Paliperidone ER costs approximately $12 to $18 per daily dose, which is similar to risperidone. Cost may be a greater consideration for patients next year, when generic risperidone becomes available (see Related Resources).

        Patient education. Because of paliperidone ER’s pharmacokinetic properties, counsel patients to:

        • take 1 tablet each day in the morning
        • not chew, split, or crush the tablets but swallow whole to preserve the controlled-release delivery.
        Also inform patients that they may see the tablet’s nonabsorbable shell in their stool as undigested residue.

        Table 3

        Paliperidone ER’s potential benefits and risks in clinical practice

        Potential benefitsDetails
        EfficacyData support acute (6 weeks) and chronic (up to 24 weeks) improvement in schizophrenia symptoms, patient function, and quality of life
        PharmacokineticsPrimarily renal excretion decreases risk of hepatic drug-drug or drug-disease interactions
        Long-acting formulationOnce-daily dosing simplifies treatment and may improve adherence
        EPSRisk similar to placebo at 3-mg and 6-mg doses, but increased at higher doses
        Weight gainSimilar to risperidone
        HyperprolactinemiaSimilar to risperidone
        TachycardiaOccurred in up to 14% of patients in clinical trials (twice the rate of placebo [7%])
        QTc prolongationIncrease up to 12 msec on average, with no patients exceeding 500 msec and no clinically adverse events during trials; use paliperidone with caution in patients with arrhythmias or cardiovascular disease or who are taking other medication that can prolong the QT interval
        EPS: extrapyramidal symptoms
        Source: References 1-3

        Efficacy trials in schizophrenia

        Three 6-week trials1-3 examined paliperidone ER’s efficacy in a total of 1,692 patients with chronic schizophrenia who were hospitalized ≥14 days with acute exacerbations. The trials were double-blind, randomized, fixed-dose, parallel-group, and placebo- and active-controlled (compared with olanzapine, 10 mg/d). Patients showed no significant differences in demographic or baseline characteristics or in the use of rescue medications.

        The primary outcome measure was mean change in Positive and Negative Syndrome Scale (PANSS) total score, which quantifies positive, negative, and global psychopathologic symptom severity. Secondary outcome measures included:

        • PANSS Marder factor scores14 (derived from PANSS items that reflect positive and negative symptoms, anxiety and depression, hostility, and thought disorganization).
        • Clinical Global Impressions-Severity (CGI-S) score, which measures overall illness severity.15
        • Personal and Social Performance (PSP) scores, which rate socially useful activities, relationships, self-care, and disturbing and aggressive behaviors; improvement by 1 category (10 points) reflects a clinically meaningful change.16,17
        The first study1 was conducted at 74 U.S. centers and enrolled 444 subjects (PANSS mean baseline score 94 ± 12). Patients were randomly assigned to fixed doses of paliperidone ER, 6 mg or 12 mg; placebo; or olanzapine, 10 mg/d. The olanzapine arm confirmed assay sensitivity and was not included in the efficacy analyses. Clinical response was defined as ≥30% improvement from baseline in total PANSS score.

        A total of 43% of patients completed the study—34% taking placebo; 46% taking paliperidone ER, 6 mg; 48% taking paliperidone ER, 12 mg; and 45% taking olanzapine. Demographic and baseline characteristics of the 432 patients who received ≥1 dose were similar across all groups. Approximately 75% of patients in each group used rescue medications—primarily lorazepam—for agitation, restlessness, or insomnia for a mean of 8 days.

         

         

        Patients taking either paliperidone ER dose showed statistically significant greater improvement in PANSS total score compared with those taking placebo (6 mg, P = 0.006; 12 mg, P

        Clinical response rates were similar with the 6-mg and 12-mg paliperidone ER doses—50% and 51%, respectively—and greater than with placebo (34%). The higher response rates with paliperidone ER were statistically significant compared with placebo (6 mg, P

        Discontinuation rates for lack of efficacy were lower with paliperidone ER (6 mg, 23%; 12 mg, 14%) than with placebo (35%). A substantially lower percentage of patients taking this agent remained classified as “marked/severe/extremely severe” on the CGI-S score from baseline to endpoint, compared with the placebo group;

        • 6 mg paliperidone ER, 58% to 26%
        • 12 mg paliperidone ER, 64% to 21%
        • placebo, 60% to 45%.
        PSP scores improved in both paliperidone ER groups, but the difference compared with placebo was statistically significant only for the 6-mg dose (P

        The second study2 included U.S. and international sites and compared 3 fixed doses of paliperidone ER (6-, 9-, and 12-mg) with placebo. Among the 630 patients enrolled, 66% completed the study. Patients were randomly assigned to 6 mg, 9 mg, or 12 mg of paliperidone ER; 10 mg of olanzapine; or placebo. The number of patients who dropped out because of adverse events was comparable across the groups.

        Patient groups assigned to paliperidone ER showed significant improvement when compared with placebo (P 30% reduction in PANSS total score from baseline to endpoint included:

        • 6 mg paliperidone ER, 56%
        • 9 mg paliperidone ER, 51%
        • 12 mg paliperidone ER, 61%
        • placebo, 30%.
        Discontinuation rates for lack of efficacy also were lower in the paliperidone ER groups vs placebo (6 mg, 16%; 9 mg, 16%; 12 mg, 10%; placebo, 40%). A substantially lower percentage of patients taking the drug remained classified as “marked/severe/extremely severe” by CGI-S score, compared with placebo:

        • 6 mg paliperidone ER, 63% at baseline to 22% at endpoint
        • 9 mg paliperidone ER, 58% to 23%
        • 12 mg paliperidone ER, 64% to 16%
        • placebo, 60% to 51%.
        PSP scores improved significantly for all 3 paliperidone ER doses vs placebo.

        The third study3 was a multicenter international trial that compared 3 fixed doses of paliperidone ER (3, 9, and 15 mg) with placebo. Among the 618 randomized patients, 365 (59%) completed the study: 70 of 127 (55%) on 3-mg paliperidone ER, 78 of 125 (62%) on 9-mg paliperidone ER, 82 of 115 (71%) on 15-mg paliperidone ER, and 47 of 123 (38%) on placebo.

        All 3 paliperidone ER doses were associated with statistically significant improvements in PANSS total and Marder factor scores at endpoint compared with placebo (P

        • 3 mg paliperidone ER, 40%
        • 9 mg paliperidone ER, 46%
        • 15 mg paliperidone ER, 53%
        • placebo, 18% (P ≤0.005).
        Discontinuation rates for lack of efficacy were lower in the paliperidone ER groups vs placebo and were dose-related (3 mg, 24%; 9 mg, 18%; 15 mg, 12%; placebo, 44%). Among patients taking olanzapine, 10 mg, 13% discontinued for lack of efficacy. Substantially fewer patients in the active drug groups were classified as “marked/severe/extremely severe” from baseline to end-point on the CGI-S scale vs the placebo group:

        • 3 mg paliperidone ER, 54% to 32%
        • 9 mg paliperidone ER, 52% to 23%
        • 15 mg paliperidone ER, 57% to 17%
        • placebo, 56% to 50%.
        Finally, a statistically significant improvement in mean PSP scores from baseline to endpoint was seen for all 3 paliperidone ER doses vs placebo (3 mg, 8.3 ± 17 points; 9 mg, 7.6 ± 14 points; 15 mg, 12 ± 15.7 points; placebo, 1.5 ± 16 points [P

        Additional trial evidence

        Schizophrenia subpopulations. Post hoc analyses of data reported from the 3 pivotal trials suggest that paliperidone ER may be useful for specific groups of schizophrenia patients, including those who are recently diagnosed, age >65, or severely ill or have predominant negative symptoms or sleep problems (Table 4).18-23

        So far, these analyses have been presented as posters at meetings or in sponsored supplements but have not been published in peer-reviewed publications.

        Efficacy in delaying recurrence. Paliperidone ER’s efficacy in delaying symptom recurrence was examined in a randomized, double-blind, placebo-controlled study of 207 patients who had been stabilized on open-label, flexible-dosed paliperidone ER.24 Time to first recurrence of schizophrenia symptoms was the primary efficacy measure. Starting dose was 9 mg/d (flexible dose range 3 to 15 mg/d).

         

         

        The study was halted at a planned interim analysis because time-to-recurrence was significantly longer for patients receiving paliperidone ER compared with placebo (P

        Final analysis of the 179 patients who completed the study confirmed the interim findings. Ongoing treatment maintained improvement in patients’ acute symptoms, functioning, and quality-of-life measures.

        Table 4

        Studies of paliperidone ER in schizophrenia subpopulations

        Patient populationStudy designFindings
        Recently diagnosed413 patients diagnosed within 5 years of study entry compared with 893 patients who had been ill ≥5 years*18,19Tolerability was similar, but recently diagnosed patients were more likely to experience movement disorders and somnolence
        Age ≥65 years114 schizophrenia patients age ≥65 given paliperidone ER, 3 to 12 mg/d, or placebo in 6-week, double-blind, randomized, placebo-controlled trial20Rates of cardiovascular, cerebrovascular, neuromotor, and metabolic changes similar to placebo, except for tachycardia (16% with paliperidone vs 0% with placebo)
        Severely ill217 patients with marked to severe symptoms (baseline total PANSS score ≥105)*21Patients treated with paliperidone showed significantly greater improvement vs placebo in mean total PANSS score (–26.7 vs –5.7) and other measures
        Substantial negative symptoms299 patients with predominant negative symptoms from 3 acute efficacy trials*22Patients treated with paliperidone showed significant improvements vs placebo on primary and secondary measures of negative symptoms
        Sleep problems36 patients age 18 to 45 diagnosed with schizophrenia and schizophrenia-related insomnia*23In stable patients, paliperidone improved sleep architecture, continuity, and patient-rated sleep quality without producing or worsening daytime sleepiness
        * Studies marked with asterisks represent post hoc analyses of data from the 3 clinical trials on which the FDA based its approval of paliperidone ER.
        PANSS: Positive and Negative Syndrome Scale
        Related resources

        Drug brand names

        • Carbamazepine • Tegretol
        • Lorazepam • Ativan
        • Olanzapine • Zyprexa
        • Paliperidone ER • Invega
        • Risperidone • Risperdal
        Disclosures

        Dr. Rado and Dr. Dowd receive research support from Neuronetics, sanofi-aventis, Janssen Pharmaceutica, and Solvay.

        Dr. Janicak receives research support from Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, Neuronetics, Solvay, and sanofi-aventis. He is a consultant to Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, Neuronetics, and Solvay, and a speaker for Abbott Laboratories, Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, and Pfizer.

        In the 9 months since paliperidone extended-release was FDA-approved for schizophrenia, the 3 acute pivotal trials supporting its approval have been published.1-3 They join a handful of post hoc analyses of this second-generation antipsychotic (SGA) in schizophrenia subgroups, including patients over age 65, recently diagnosed patients, and those with predominant negative symptoms.

        This article discusses the evidence and paliperidone ER’s probable clinical benefits and adverse effects, with focus on its:

        • pharmacodynamics and pharmacokinetics
        • potential efficacy in schizophrenia and for specific patients and symptoms
        • safety and tolerability.

        How does paliperidone ER work?

        Paliperidone ER was approved for schizophrenia treatment in December 2006 based on three 6-week, randomized, placebo-controlled trials. Paliperidone ER is the active metabolite of risperidone (9-OH risperidone) delivered in a once-daily, time-released formulation (Table 1).

        Pharmacodynamics. Similar to risperidone, paliperidone ER has high binding affinity for dopamine (D2) and serotonin (5-HT2A) receptors, with additional affinity for histaminic (H1) and adrenergic receptors (alpha1 and alpha2) but not for muscarinic-cholinergic receptors.

        Pharmacokinetics. After oral administration, the medication is widely and rapidly distributed. The drug’s terminal half-life is about 23 hours, and steady-state concentration is reached in 4 to 5 days.4,5

        Approximately 60% of the medication is eliminated renally and 11% is eliminated in the feces unchanged, with very limited hepatic metabolism.6 As a result, paliperidone ER appears to lack enzyme-inducing or inhibiting properties and does not substantially affect drugs that undergo cytochrome P-450 metabolism in the liver.

        Thus paliperidone ER—when compared with risperidone and other antipsychotics that are metabolized primarily in the liver—is less likely to be involved in hepatic drug-drug or drug-disease interactions. However, some drugs that can induce CYP-450 enzymes—such as carbamazepine—may affect paliperidone’s metabolism.7

        Paliperidone has an osmotic controlled-release oral delivery system (OROS®) for steady medication delivery across 24 hours8 (Table 2).1-3 The tablet consists of an osmotically active tri-layer core surrounded by a semipermeable membrane. When the tablet is swallowed, the membrane controls the rate of water reaching the tablet core, which determines the rate of drug delivery.6 The result is less variation between peak and trough drug concentrations, compared with immediate-release formulations.

        Table 1

        How paliperidone ER compares with risperidone

        CharacteristicPaliperidone ERRisperidone
        FormulationOROS extended-releaseImmediate release
        Active moiety9-OH risperidoneRisperidone plus 9-OH risperidone
        MetabolismPrimarily renalPrimarily hepatic
        Drug interactionsMinimalPrimarily through cytochrome P-450 enzyme 2D6
        DosingStart at target doseTitrate to target dose
        OROS: osmotic controlled-release oral delivery system
        Table 2

        Paliperidone ER’s clinical characteristics

        Second-generation antipsychotic approved for schizophrenia
        9-OH active metabolite of risperidone
        Osmotic controlled-release system provides steady-state drug delivery over 24 hours
        Terminal half-life (time for 50% of drug to be eliminated from the body) ~23 hours
        Available in 3-mg, 6-mg, and 9-mg tablets; recommended starting dose is 6 mg/d, and labeled dose range is 3 to 12 mg/d
        Excreted primarily by the kidney; maximum recommended dose for patients with oderate to severe renal impairment is 3 mg/d
        Source: References 1-3

        Clinical use

        Paliperidone ER offers potential therapeutic benefits in treating schizophrenia patients, although not without the risk of adverse events such as extrapyramidal symptoms (EPS) (Table 3).1-3

        Patient selection. Because of its slow-release formulation and relatively stable plasma concentrations, paliperidone ER might be useful for patients who are highly sensitive to antipsychotics’ side effects. In particular, paliperidone ER might be ideal for patients who respond to but may not tolerate risperidone.

        Paliperidone ER appears to be safe in patients with liver disease. Its primary renal excretion should minimize the risk of hepatic-related drug interactions in patients taking multiple medications.

        Dosage and titration. For treating schizophrenia, the suggested starting dose of paliperidone ER is 6 mg/d taken in the morning. In the 3 pivotal trials, 6 mg was the lowest dose to show broad efficacy with minimal adverse events.9

        For many patients, the 6-mg starting dose will be the therapeutic dose. When needed, the dose may be increased in 3-mg increments every 1 to 2 weeks to a maximum 12 mg/d (a 15-mg dose was used in clinical trials, but the adverse effects out-weighed the benefits). Lower maximum doses are recommended for patients with renal impairment:

        • 6 mg/d for those with creatinine clearance ≥50 to
        • 3 mg/d for those with creatinine clearance 10 to 10
        In the pivotal trials, differences in the terminal elimination half-life between hepatically impaired and healthy patients were minimal (26.5 hours vs 23.6 hours, respectively). Unbound paliperidone levels were slightly lower in patients with hepatic impairment11 but not low enough to recommend dose adjustment.

        Safety and tolerability. Pooled data from the 3 trials indicate that adverse events (AEs) occurred during treatment in 66% to 77% of patients receiving paliperidone ER vs 66% in placebo groups. The most common AEs were headache (11% to 18%), insomnia (4% to 12%), and anxiety (6% to 9%).9

         

         

        EPS. Risk of EPS-related AEs (such as akathisia and parkinsonian symptoms) with 3-mg and 6-mg paliperidone ER doses (13% and 10%, respectively) was similar to placebo (11%) but increased with the 9-mg, 12-mg, and 15-mg doses (25%, 26%, and 24%, respectively). Should EPS occur, reduce the paliperidone ER dose or consider adding antiparkinsonian medications.

        Lab values. No clinically relevant changes were noted in blood glucose, insulin, or lipids.12 Similar to risperidone, paliperidone ER elevated prolactin levels.

        Weight gain with paliperidone ER is dose-dependent; in the clinical trials, mean body weight change for all doses was ≤1.9 kg, which is similar to the weight gain seen with risperidone and in the moderate range compared with other SGAs. When using paliperidone ER, follow the American Diabetes Association/American Psychiatric Association guidelines13 for monitoring weight gain and metabolic parameters with antipsychotics. Also monitor patients for clinical symptoms of hyperprolactinemia, and—if intolerable—adjust the dose or switch to another SGA.

        Tachycardia. Advise patients that they may experience a rapid heart rate while taking paliperidone ER. In clinical trials, tachycardia occurred in ≤14% of patients—twice the rate with placebo—but did not contribute to more serious cardiac rhythm disturbances or to discontinuation. Incidence of prolonged corrected QT interval (QTc) was 3% to 5% in the paliperidone ER group vs 3% in the placebo group.

        Cost. Paliperidone ER costs approximately $12 to $18 per daily dose, which is similar to risperidone. Cost may be a greater consideration for patients next year, when generic risperidone becomes available (see Related Resources).

        Patient education. Because of paliperidone ER’s pharmacokinetic properties, counsel patients to:

        • take 1 tablet each day in the morning
        • not chew, split, or crush the tablets but swallow whole to preserve the controlled-release delivery.
        Also inform patients that they may see the tablet’s nonabsorbable shell in their stool as undigested residue.

        Table 3

        Paliperidone ER’s potential benefits and risks in clinical practice

        Potential benefitsDetails
        EfficacyData support acute (6 weeks) and chronic (up to 24 weeks) improvement in schizophrenia symptoms, patient function, and quality of life
        PharmacokineticsPrimarily renal excretion decreases risk of hepatic drug-drug or drug-disease interactions
        Long-acting formulationOnce-daily dosing simplifies treatment and may improve adherence
        EPSRisk similar to placebo at 3-mg and 6-mg doses, but increased at higher doses
        Weight gainSimilar to risperidone
        HyperprolactinemiaSimilar to risperidone
        TachycardiaOccurred in up to 14% of patients in clinical trials (twice the rate of placebo [7%])
        QTc prolongationIncrease up to 12 msec on average, with no patients exceeding 500 msec and no clinically adverse events during trials; use paliperidone with caution in patients with arrhythmias or cardiovascular disease or who are taking other medication that can prolong the QT interval
        EPS: extrapyramidal symptoms
        Source: References 1-3

        Efficacy trials in schizophrenia

        Three 6-week trials1-3 examined paliperidone ER’s efficacy in a total of 1,692 patients with chronic schizophrenia who were hospitalized ≥14 days with acute exacerbations. The trials were double-blind, randomized, fixed-dose, parallel-group, and placebo- and active-controlled (compared with olanzapine, 10 mg/d). Patients showed no significant differences in demographic or baseline characteristics or in the use of rescue medications.

        The primary outcome measure was mean change in Positive and Negative Syndrome Scale (PANSS) total score, which quantifies positive, negative, and global psychopathologic symptom severity. Secondary outcome measures included:

        • PANSS Marder factor scores14 (derived from PANSS items that reflect positive and negative symptoms, anxiety and depression, hostility, and thought disorganization).
        • Clinical Global Impressions-Severity (CGI-S) score, which measures overall illness severity.15
        • Personal and Social Performance (PSP) scores, which rate socially useful activities, relationships, self-care, and disturbing and aggressive behaviors; improvement by 1 category (10 points) reflects a clinically meaningful change.16,17
        The first study1 was conducted at 74 U.S. centers and enrolled 444 subjects (PANSS mean baseline score 94 ± 12). Patients were randomly assigned to fixed doses of paliperidone ER, 6 mg or 12 mg; placebo; or olanzapine, 10 mg/d. The olanzapine arm confirmed assay sensitivity and was not included in the efficacy analyses. Clinical response was defined as ≥30% improvement from baseline in total PANSS score.

        A total of 43% of patients completed the study—34% taking placebo; 46% taking paliperidone ER, 6 mg; 48% taking paliperidone ER, 12 mg; and 45% taking olanzapine. Demographic and baseline characteristics of the 432 patients who received ≥1 dose were similar across all groups. Approximately 75% of patients in each group used rescue medications—primarily lorazepam—for agitation, restlessness, or insomnia for a mean of 8 days.

         

         

        Patients taking either paliperidone ER dose showed statistically significant greater improvement in PANSS total score compared with those taking placebo (6 mg, P = 0.006; 12 mg, P

        Clinical response rates were similar with the 6-mg and 12-mg paliperidone ER doses—50% and 51%, respectively—and greater than with placebo (34%). The higher response rates with paliperidone ER were statistically significant compared with placebo (6 mg, P

        Discontinuation rates for lack of efficacy were lower with paliperidone ER (6 mg, 23%; 12 mg, 14%) than with placebo (35%). A substantially lower percentage of patients taking this agent remained classified as “marked/severe/extremely severe” on the CGI-S score from baseline to endpoint, compared with the placebo group;

        • 6 mg paliperidone ER, 58% to 26%
        • 12 mg paliperidone ER, 64% to 21%
        • placebo, 60% to 45%.
        PSP scores improved in both paliperidone ER groups, but the difference compared with placebo was statistically significant only for the 6-mg dose (P

        The second study2 included U.S. and international sites and compared 3 fixed doses of paliperidone ER (6-, 9-, and 12-mg) with placebo. Among the 630 patients enrolled, 66% completed the study. Patients were randomly assigned to 6 mg, 9 mg, or 12 mg of paliperidone ER; 10 mg of olanzapine; or placebo. The number of patients who dropped out because of adverse events was comparable across the groups.

        Patient groups assigned to paliperidone ER showed significant improvement when compared with placebo (P 30% reduction in PANSS total score from baseline to endpoint included:

        • 6 mg paliperidone ER, 56%
        • 9 mg paliperidone ER, 51%
        • 12 mg paliperidone ER, 61%
        • placebo, 30%.
        Discontinuation rates for lack of efficacy also were lower in the paliperidone ER groups vs placebo (6 mg, 16%; 9 mg, 16%; 12 mg, 10%; placebo, 40%). A substantially lower percentage of patients taking the drug remained classified as “marked/severe/extremely severe” by CGI-S score, compared with placebo:

        • 6 mg paliperidone ER, 63% at baseline to 22% at endpoint
        • 9 mg paliperidone ER, 58% to 23%
        • 12 mg paliperidone ER, 64% to 16%
        • placebo, 60% to 51%.
        PSP scores improved significantly for all 3 paliperidone ER doses vs placebo.

        The third study3 was a multicenter international trial that compared 3 fixed doses of paliperidone ER (3, 9, and 15 mg) with placebo. Among the 618 randomized patients, 365 (59%) completed the study: 70 of 127 (55%) on 3-mg paliperidone ER, 78 of 125 (62%) on 9-mg paliperidone ER, 82 of 115 (71%) on 15-mg paliperidone ER, and 47 of 123 (38%) on placebo.

        All 3 paliperidone ER doses were associated with statistically significant improvements in PANSS total and Marder factor scores at endpoint compared with placebo (P

        • 3 mg paliperidone ER, 40%
        • 9 mg paliperidone ER, 46%
        • 15 mg paliperidone ER, 53%
        • placebo, 18% (P ≤0.005).
        Discontinuation rates for lack of efficacy were lower in the paliperidone ER groups vs placebo and were dose-related (3 mg, 24%; 9 mg, 18%; 15 mg, 12%; placebo, 44%). Among patients taking olanzapine, 10 mg, 13% discontinued for lack of efficacy. Substantially fewer patients in the active drug groups were classified as “marked/severe/extremely severe” from baseline to end-point on the CGI-S scale vs the placebo group:

        • 3 mg paliperidone ER, 54% to 32%
        • 9 mg paliperidone ER, 52% to 23%
        • 15 mg paliperidone ER, 57% to 17%
        • placebo, 56% to 50%.
        Finally, a statistically significant improvement in mean PSP scores from baseline to endpoint was seen for all 3 paliperidone ER doses vs placebo (3 mg, 8.3 ± 17 points; 9 mg, 7.6 ± 14 points; 15 mg, 12 ± 15.7 points; placebo, 1.5 ± 16 points [P

        Additional trial evidence

        Schizophrenia subpopulations. Post hoc analyses of data reported from the 3 pivotal trials suggest that paliperidone ER may be useful for specific groups of schizophrenia patients, including those who are recently diagnosed, age >65, or severely ill or have predominant negative symptoms or sleep problems (Table 4).18-23

        So far, these analyses have been presented as posters at meetings or in sponsored supplements but have not been published in peer-reviewed publications.

        Efficacy in delaying recurrence. Paliperidone ER’s efficacy in delaying symptom recurrence was examined in a randomized, double-blind, placebo-controlled study of 207 patients who had been stabilized on open-label, flexible-dosed paliperidone ER.24 Time to first recurrence of schizophrenia symptoms was the primary efficacy measure. Starting dose was 9 mg/d (flexible dose range 3 to 15 mg/d).

         

         

        The study was halted at a planned interim analysis because time-to-recurrence was significantly longer for patients receiving paliperidone ER compared with placebo (P

        Final analysis of the 179 patients who completed the study confirmed the interim findings. Ongoing treatment maintained improvement in patients’ acute symptoms, functioning, and quality-of-life measures.

        Table 4

        Studies of paliperidone ER in schizophrenia subpopulations

        Patient populationStudy designFindings
        Recently diagnosed413 patients diagnosed within 5 years of study entry compared with 893 patients who had been ill ≥5 years*18,19Tolerability was similar, but recently diagnosed patients were more likely to experience movement disorders and somnolence
        Age ≥65 years114 schizophrenia patients age ≥65 given paliperidone ER, 3 to 12 mg/d, or placebo in 6-week, double-blind, randomized, placebo-controlled trial20Rates of cardiovascular, cerebrovascular, neuromotor, and metabolic changes similar to placebo, except for tachycardia (16% with paliperidone vs 0% with placebo)
        Severely ill217 patients with marked to severe symptoms (baseline total PANSS score ≥105)*21Patients treated with paliperidone showed significantly greater improvement vs placebo in mean total PANSS score (–26.7 vs –5.7) and other measures
        Substantial negative symptoms299 patients with predominant negative symptoms from 3 acute efficacy trials*22Patients treated with paliperidone showed significant improvements vs placebo on primary and secondary measures of negative symptoms
        Sleep problems36 patients age 18 to 45 diagnosed with schizophrenia and schizophrenia-related insomnia*23In stable patients, paliperidone improved sleep architecture, continuity, and patient-rated sleep quality without producing or worsening daytime sleepiness
        * Studies marked with asterisks represent post hoc analyses of data from the 3 clinical trials on which the FDA based its approval of paliperidone ER.
        PANSS: Positive and Negative Syndrome Scale
        Related resources

        Drug brand names

        • Carbamazepine • Tegretol
        • Lorazepam • Ativan
        • Olanzapine • Zyprexa
        • Paliperidone ER • Invega
        • Risperidone • Risperdal
        Disclosures

        Dr. Rado and Dr. Dowd receive research support from Neuronetics, sanofi-aventis, Janssen Pharmaceutica, and Solvay.

        Dr. Janicak receives research support from Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, Neuronetics, Solvay, and sanofi-aventis. He is a consultant to Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, Neuronetics, and Solvay, and a speaker for Abbott Laboratories, Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, and Pfizer.

        References

        1. Marder S, Kramer M, Ford L, et al. Efficacy and safety of paliperidone extended-release tablets: results of a 6-week, randomized, placebo-controlled study. Biol Psychiatry 2007; Jun 27; Epub ahead of print.

        2. Kane J, Canas F, Kramer M, et al. Treatment of schizophrenia with paliperidone extended-release tablets: a 6-week placebo-controlled trial. Schizophr Res 2007;90(1-3):147-61.

        3. Davidson M, Emsley R, Kramer M, et al. Efficacy, safety and early response of paliperidone extended-release tablets (paliperidone ER): results of a 6-week, randomized, placebo-controlled study. Schizophr Res 2007;93(1-3):117-30.

        4. Rossenu SAC, Rusch S, Janssens L, et al. Extended release formulation of paliperidone shows dose proportional pharmacokinetics. Presented at: Annual Meeting of the American Association of Pharmaceutical Scientists; October 29, 2006; San Antonio, TX.

        5. Vermeir M, Boom S, Naessens I, et al. Absorption, metabolism, and excretion of a single oral dose of 14C-paliperidone 1 mg in healthy subjects. Eur Neuropsychopharmacol 2005;15(suppl):S648-9.

        6. Conley R, Gupta SK, Sathyan G. Clinical spectrum of the osmotic-controlled release oral delivery system (OROS), an advanced oral delivery form. Curr Med Res Opin 2006;22(10):1879-92.

        7. Spina E, Avenoso A, Facciola G, et al. Plasma concentrations of risperidone and 9-hydroxyrisperidone: effect of comedication with carbamazepine or valproate. Ther Drug Monit 2000;22(4):481-5.

        8. Paliperidone extended release. Prescribing information. Available at: http://www.invega.com. Accessed August 8, 2007.

        9. Meltzer H, Kramer M, Gassmann-Mayer C, et al. Efficacy and tolerability of oral paliperidone extended-release tablets in the treatment of acute schizophrenia: pooled data from three 6-week placebo controlled studies. Int J Neuropsychopharmacol 2006;9(suppl 1):S225.-

        10. Thyssen A, Cleton A, Osselae NV, et al. Effects of renal impairment on the pharmacokinetic profile of paliperidone extended-release tablets. Clin Pharmacol Ther 2007. In press.

        11. Thyssen A, Crauwels H, Cleton A, et al. Effects of hepatic impairment on the pharmacokinetics of paliperidone immediate-release. Presented at: 46th Annual Meeting of the New Clinical Drug Evaluation Unit (NCDEU); June 12-15, 2006; Boca Raton, FL.

        12. Meyer J, Kramer M, Lane R, et al. Metabolic outcomes in patients with schizophrenia treated with oral paliperidone extended release tablets: pooled analysis of three 6 week placebo-controlled studies. Int J Neuropsychopharmacol 2006;9(suppl 1):S282.-

        13. American Diabetes Association, American Psychiatric Association, American Association of Clinical Endocrinologists, North American Association for the Study of Obesity. Consensus Development Conference on Antipsychotic Drugs and Obesity and Diabetes. J Clin Psychiatry 2004;65:267-72.

        14. Marder SR, Davis JM, Chouinard G. The effects of risperidone on the five dimensions of schizophrenia derived by factor analysis: combined results of the North American trials. J Clin Psychiatry 1997;58:538-46.

        15. Guy W. Clinical Global Impressions Scale. Early clinical drug evaluation unit (ECDEU) assessment manual for psychopharmacology. Rockville, MD: National Institute of Mental Health, Department of Health, Education, and Welfare; 1976:218-22. ADM publication 76-338.

        16. Morosini PL, Magliano L, Brambilla L, et al. Development, reliability and acceptability of a new version of the DSMIV Social and Occupational Functioning Assessment Scale (SOFAS) to assess routine social functioning. Acta Psychiatr Scand 2000;101:323-9.

        17. Patrick D, Adriaenssen I, Morosini P, Rothman M. Reliability, validity and sensitivity to change of the Personal and Social Performance scale in patients with acute schizophrenia. Int J Neuropsychopharmacol 2006;9(suppl 1):S287-8.

        18. Kostic D, Bossie C, Turkoz I, et al. Paliperidone extended-release tablets in patients recently diagnosed with schizophrenia. Int J Neuropsychopharmacol 2006;9(suppl 1):S161.-

        19. Kostic D, Bossie C, Turkoz I, et al. Paliperidone extended-release tablets in patients recently diagnosed with schizophrenia. Presented at: Congress of the Collegium Internationale Neruo-Psychopharmacologicum (CINP); July 9-13, 2006; Chicago, IL.

        20. Tzimos A, Kramer M, Ford L, et al. A 6-week placebo-controlled study of the safety and tolerability of flexible doses of oral paliperidone extended release tablets in the treatment of schizophrenia in elderly patients. Int J Neuropsychopharmacol 2006;9(suppl 1):S155.-

        21. Canuso C, Youssef E, Dirks B, et al. Paliperidone extended-release in severely-ill patients with schizophrenia. Presented at: 58th Annual Institute on Psychiatric Services; October 5-8, 2006; New York, NY.

        22. Dirks B, Eerdekens M, Turkoz I, et al. Efficacy of paliperidone extended-release tablets in patients with schizophrenia and predominant negative symptoms. Int J Neuropsychopharmacol 2006;9(suppl 1):S162.-

        23. Luthringer R, Staner L, Noel N, et al. Sleep assessments in patients with schizophrenia following treatment with paliperidone extended-release tablets. Eur Neuropsychopharmacol 2006;16(suppl 4):S224.-

        24. Kramer M, Simpson G, Maciulis V, et al. Paliperidone extended-release tablets for prevention of symptom recurrence in patients with schizophrenia: a randomized double-blind, placebo-controlled study [published correction appears in J Clin Psychopharmacol. 2007;27(3):258]. J Clin Psychopharmacol 2007;27(1):6-14.

        References

        1. Marder S, Kramer M, Ford L, et al. Efficacy and safety of paliperidone extended-release tablets: results of a 6-week, randomized, placebo-controlled study. Biol Psychiatry 2007; Jun 27; Epub ahead of print.

        2. Kane J, Canas F, Kramer M, et al. Treatment of schizophrenia with paliperidone extended-release tablets: a 6-week placebo-controlled trial. Schizophr Res 2007;90(1-3):147-61.

        3. Davidson M, Emsley R, Kramer M, et al. Efficacy, safety and early response of paliperidone extended-release tablets (paliperidone ER): results of a 6-week, randomized, placebo-controlled study. Schizophr Res 2007;93(1-3):117-30.

        4. Rossenu SAC, Rusch S, Janssens L, et al. Extended release formulation of paliperidone shows dose proportional pharmacokinetics. Presented at: Annual Meeting of the American Association of Pharmaceutical Scientists; October 29, 2006; San Antonio, TX.

        5. Vermeir M, Boom S, Naessens I, et al. Absorption, metabolism, and excretion of a single oral dose of 14C-paliperidone 1 mg in healthy subjects. Eur Neuropsychopharmacol 2005;15(suppl):S648-9.

        6. Conley R, Gupta SK, Sathyan G. Clinical spectrum of the osmotic-controlled release oral delivery system (OROS), an advanced oral delivery form. Curr Med Res Opin 2006;22(10):1879-92.

        7. Spina E, Avenoso A, Facciola G, et al. Plasma concentrations of risperidone and 9-hydroxyrisperidone: effect of comedication with carbamazepine or valproate. Ther Drug Monit 2000;22(4):481-5.

        8. Paliperidone extended release. Prescribing information. Available at: http://www.invega.com. Accessed August 8, 2007.

        9. Meltzer H, Kramer M, Gassmann-Mayer C, et al. Efficacy and tolerability of oral paliperidone extended-release tablets in the treatment of acute schizophrenia: pooled data from three 6-week placebo controlled studies. Int J Neuropsychopharmacol 2006;9(suppl 1):S225.-

        10. Thyssen A, Cleton A, Osselae NV, et al. Effects of renal impairment on the pharmacokinetic profile of paliperidone extended-release tablets. Clin Pharmacol Ther 2007. In press.

        11. Thyssen A, Crauwels H, Cleton A, et al. Effects of hepatic impairment on the pharmacokinetics of paliperidone immediate-release. Presented at: 46th Annual Meeting of the New Clinical Drug Evaluation Unit (NCDEU); June 12-15, 2006; Boca Raton, FL.

        12. Meyer J, Kramer M, Lane R, et al. Metabolic outcomes in patients with schizophrenia treated with oral paliperidone extended release tablets: pooled analysis of three 6 week placebo-controlled studies. Int J Neuropsychopharmacol 2006;9(suppl 1):S282.-

        13. American Diabetes Association, American Psychiatric Association, American Association of Clinical Endocrinologists, North American Association for the Study of Obesity. Consensus Development Conference on Antipsychotic Drugs and Obesity and Diabetes. J Clin Psychiatry 2004;65:267-72.

        14. Marder SR, Davis JM, Chouinard G. The effects of risperidone on the five dimensions of schizophrenia derived by factor analysis: combined results of the North American trials. J Clin Psychiatry 1997;58:538-46.

        15. Guy W. Clinical Global Impressions Scale. Early clinical drug evaluation unit (ECDEU) assessment manual for psychopharmacology. Rockville, MD: National Institute of Mental Health, Department of Health, Education, and Welfare; 1976:218-22. ADM publication 76-338.

        16. Morosini PL, Magliano L, Brambilla L, et al. Development, reliability and acceptability of a new version of the DSMIV Social and Occupational Functioning Assessment Scale (SOFAS) to assess routine social functioning. Acta Psychiatr Scand 2000;101:323-9.

        17. Patrick D, Adriaenssen I, Morosini P, Rothman M. Reliability, validity and sensitivity to change of the Personal and Social Performance scale in patients with acute schizophrenia. Int J Neuropsychopharmacol 2006;9(suppl 1):S287-8.

        18. Kostic D, Bossie C, Turkoz I, et al. Paliperidone extended-release tablets in patients recently diagnosed with schizophrenia. Int J Neuropsychopharmacol 2006;9(suppl 1):S161.-

        19. Kostic D, Bossie C, Turkoz I, et al. Paliperidone extended-release tablets in patients recently diagnosed with schizophrenia. Presented at: Congress of the Collegium Internationale Neruo-Psychopharmacologicum (CINP); July 9-13, 2006; Chicago, IL.

        20. Tzimos A, Kramer M, Ford L, et al. A 6-week placebo-controlled study of the safety and tolerability of flexible doses of oral paliperidone extended release tablets in the treatment of schizophrenia in elderly patients. Int J Neuropsychopharmacol 2006;9(suppl 1):S155.-

        21. Canuso C, Youssef E, Dirks B, et al. Paliperidone extended-release in severely-ill patients with schizophrenia. Presented at: 58th Annual Institute on Psychiatric Services; October 5-8, 2006; New York, NY.

        22. Dirks B, Eerdekens M, Turkoz I, et al. Efficacy of paliperidone extended-release tablets in patients with schizophrenia and predominant negative symptoms. Int J Neuropsychopharmacol 2006;9(suppl 1):S162.-

        23. Luthringer R, Staner L, Noel N, et al. Sleep assessments in patients with schizophrenia following treatment with paliperidone extended-release tablets. Eur Neuropsychopharmacol 2006;16(suppl 4):S224.-

        24. Kramer M, Simpson G, Maciulis V, et al. Paliperidone extended-release tablets for prevention of symptom recurrence in patients with schizophrenia: a randomized double-blind, placebo-controlled study [published correction appears in J Clin Psychopharmacol. 2007;27(3):258]. J Clin Psychopharmacol 2007;27(1):6-14.

        Issue
        Current Psychiatry - 06(09)
        Issue
        Current Psychiatry - 06(09)
        Page Number
        75-82
        Page Number
        75-82
        Publications
        Publications
        Article Type
        Display Headline
        Paliperidone ER: Reformulated antipsychotic for schizophrenia Tx
        Display Headline
        Paliperidone ER: Reformulated antipsychotic for schizophrenia Tx
        Legacy Keywords
        paliperidone; Invega; schizophrenia; second-generation antipsychotic; SGA; Jeffrey Rado MD; Sheila M. Dowd PhD; Philip G. Janicak MD
        Legacy Keywords
        paliperidone; Invega; schizophrenia; second-generation antipsychotic; SGA; Jeffrey Rado MD; Sheila M. Dowd PhD; Philip G. Janicak MD
        Sections
        Article Source

        PURLs Copyright

        Inside the Article

        Article PDF Media
        Image
        Disable zoom
        Off

        Vagus nerve stimulation

        Article Type
        Changed
        Tue, 12/11/2018 - 15:37
        Display Headline
        Vagus nerve stimulation

        What is vagus nerve stimulation’s (VNS) role in treating chronic or recurrent depression? Which patients would benefit from this implant, now FDA-approved for depression as well as epilepsy?

        Drawing from the evidence, this article discusses which patients with depression may be candidates for VNS, how it works, and its potential benefits and side effects.

        Clinical Applicability

        VNS is indicated for patients with chronic or recurrent treatment-resistant depression during an episode that has not responded to ≥4 adequate antidepressant treatment trials (defined as ≥3 on the Antidepressant Treatment History Form [ATHF]) (Table 1). Implantation theoretically promotes 100% adherence and reduces drug-drug interaction risk. Interactions between VNS and nonpsychotropics are possible but unlikely.

        Paradoxically, data suggest that patients with low to moderate resistance to antidepressant treatment (≤3 antidepressant trial failures) are most likely to benefit from VNS.1 Patients who had never received electroconvulsive therapy (ECT) (indicating relatively low treatment resistance) were nearly four times more likely than ECT-treated patients to respond to VNS.2 Conversely, 13 subjects who had not responded to ≥ 7 adequate treatment trials (indicating relatively severe treatment resistance) did not respond to VNS.2

        Table 1

        Vagus nerve stimulation device: Fast facts

         

        Brand name:
        Cyberonics Vagus Nerve Stimulation (VNS) Therapy System
        FDA-approved indications:
        Treatment-resistant depression (previously approved for treatment-refractory epilepsy)
        Manufacturer:
        Cyberonics
        Recommended use:
        Treating depressive episode that has not responded to ≥4 antidepressant trials or electroconvulsive therapy in a patient with chronic or recurrent depression
        Information on VNS remote device training:
        1-877-NOW-4-VNS (669-4867) or www.vnstherapy.com

        How VNS Works

        The vagus (10th cranial) nerve is a main efferent outflow tract for parasympathetic innervation of the abdomen and chest, regulating heart rate, acid secretion, and bowel motility.

        The largest component of the left vagus nerve—approximately 80%—conducts information about pain, hunger, and satiety. These fibers are also believed to contribute to VNS’ antidepressant effects by carrying information to the solitary nucleus of the medulla. From there, fibers project to the median raphe nucleus and locus coeruleus, key areas of serotonergic and noradrenergic innervation relevant to depression.

        Positron emission tomography studies suggest that VNS also increases blood flow to the thalamus, hypothalamus, and insula—brain areas considered relevant to mood disorders.3

        VNS requires subcutaneous implantation of a pacemaker-like pulse generator into the upper left chest. The generator is 6.9 mm thick and weighs 25 grams. Wires extend from the device into the left vagus nerve in the neck (Figure). A neurosurgeon usually performs the 1- to 2-hour outpatient procedure, although ENT, vascular, and general surgeons may also do the implant.

        The device sends electric pulses to the left vagus nerve every few seconds (Table 2). Using an accompanying hand-held device and a computer, the clinician programs the implant and adjusts stimulation parameters to ensure the correct amount of stimulation.

        FDA approved VNS in 1997 for refractory epilepsy. Clinical observations that VNS improved epilepsy patients’ mood spurred interest in its antidepressant effects.4 Preliminary data suggest VNS also could help manage anxiety disorders, obesity, pain syndromes, and Alzheimer’s disease.5

         

        Figure How VNS device works


        RTEmagicC_9be250daef.jpg.jpg

        Pacemaker-like VNSdevice is implanted into the upper left chest. Wires extending from the device transport electric pulses into the left vagus nerve in the neck, which carries information to areas of serotonergic and noradrenergic innervation relevant to depression.Table 2

        VNS stimulation parameters

         

        Frequency: 20 to 30 Hz
        Intensity: 0.25 mA (0.25 to 3.0 mA)
        Pulse width: 250 to 500 μs
        Duty cycle: 30 seconds on/5 minutes off

        Cost

        VNS implantation costs approximately $25,000, including the device, surgeon’s fee, and facility charge. Psychiatrists generally would initiate the referral process.

        Follow-up management fees for epilepsy are $150 to $250 per visit. Several follow-up visits are required after stimulation is started to verify the device is working, evaluate treatment response and tolerability, and adjust stimulation as needed. Thereafter, periodic visits are appropriate.

        Generally, insurers cover VNS as an epilepsy treatment; whether private insurers and Medicare will cover VNS for depression remains to be seen. Case mangers at Cyberonics, the device’s manufacturer, are on call to assist with VNS coverage, coding, and reimbursement issues (see Related resources).

        Because the internal implant’s battery life is 6 to 11 years, VNS therapy will likely be cost-effective for many patients, although follow-up surgery would be required to replace the battery. Costs of using VNS have not been compared with other antidepressant modalities.

        VNS’ Efficacy In Depression

        In an open-label trial, 60 patients ages 20 to 63 received VNS with no placebo or active comparator.2 Thirty had completed an open-label pilot study that showed VNS’ potential antidepressant effects.6 Before implantation, all subjects had:

         

         

         

        • a major depressive episode lasting >2 years or >4 lifetime major depressive episodes
        • nonresponse to ECT or ≥2 adequate antidepressant trials (ATHF scores >3) during their current major depressive episode (median duration: 4.7 years)
        • DSM-IV diagnosis of major depressive disorder or bipolar type I or II disorder depressed phase.
        • baseline scores ≥20 on the 28-item Hamilton Rating Scale for Depression (HRSD-28) and ≤50 on the Global Assessment of Functioning (GAF) scale.

        Two weeks after implantation, the stimulator was turned on and adjusted for another 2 weeks to the maximum tolerable dose. Patients then received 8 weeks of fixed-dose stimulation. Participants who had been taking an antidepressant, mood stabilizer, second-generation antipsychotic, or other psychotropic at the same dosages for ≥4 weeks before the study could continue their medications during the VNS trial (median concurrent treatments: 4).

        Three months after implantation, 18 of 59 subjects (30.5%) showed clinical response (≥50% improvement in HRSD-28 scores over baseline). Nine patients (15.3%) showed depression remission (HRSD-28 score ≤10). Median time to first response was 45.5 days.

        Twenty participants (34%) showed a ≥50% reduction in baseline Montgomery-Asberg Depression Rating Scale (MADRS) scores, and 22 (37%) showed Clinical Global Impression-Improvement Scale (CGI-I) scores improving to 1 or 2.

        Therapeutic effects did not differ among patients with unipolar and bipolar depression. Participants with mild to moderate depression (defined as 2 to 3 failed adequate trials) showed higher response rates (50% vs. 29.1%) than did those with more-severe depression (defined as ≥4 failed adequate trials).2

        Among 28 patients followed for 1 year, 13 (46%) met HRSD-28 response criteria (≥ 50% score reduction) and 8 (29%) met remission criteria (score ≤ 10), showing gradual improvement.1 After 2 years, 44% of patients met HDRS-28 response criteria, and 22% met remission criteria, showing sustained benefit.7 How many subjects were taking one or more concomitant psychotropics is unknown.

        In a double-blind controlled trial, 235 subjects ages 18 to 80 received VNS or a sham comparator.8 Treatment response and remission were defined as ≥50% reduction from baseline and ≤9, respectively, on the 24-item HRSD (HRSD-24). Patient selection criteria were similar to those of the open-label study.

        All patients received VNS implants, which were inactive the first 2 weeks. Patients were then randomly assigned to active treatment (stimulator turned on) or sham control (stimulator left off). After 10 weeks of treatment, HRSD-24, CGI-I, and MADRS scores were similar between the VNS and sham groups, but Inventory of Depressive Symptomatology Self Report (IDS-SR) scores improved much more in the active treatment group (P<0.03). Patients in the sham group then had their stimulators turned on.

        After 1 year of active treatment for both groups, response and remission rates more than doubled among 205 evaluable subjects (response: 14.4% to 29.8%; remission: 7.3% to 17.1%). MADRS and IDS-SR scores also improved. Three percent of subjects dropped out because of adverse events.

        Another analysis of these data revealed significant improvement among the VNS treatment group vs. a comparator-matched control group of treatment-resistant patients across 2 years.8

        Depression treatment among patients in the comparator group followed standard clinical practice.

        Side Effects

        Voice alteration or hoarseness was most commonly reported after 12 weeks in the open-label trial (55% of subjects). Headache (22%), cough (17%), shortness of breath (15%), neck pain (17%), dysphagia (20%), and pain (15%) were also reported.2 These effects emerge or increase with stimulation intensity and may be ameliorated by reducing the dose.

        Small risks of infection (1%) and nerve damage (1%) were reported. Leaving the stimulator off for 14 days after implantation decreases nerve damage risk. Pain at the incision site (experienced by 30%) resolved after 1 to 2 weeks.2 Other adverse events included:

         

        • hypomania in one bipolar patient; this was resolved by adjusting medication and reducing stimulation
        • leg pain in 2 subjects
        • worsened depression in 5 patients (2 of these may have been related to stimulation)
        • emesis and diarrhea in 1 subject.

        One patient with multiple cardiac risk factors developed a myocardial infarction but completed the trial after angioplasty and stent placement.2

        After 1 year in the open-label trial, no subjects dropped out because of adverse events. Common side events included voice alteration (21%), shortness of breath (7%), and neck pain (7%). More-serious adverse events reported between the acute trial and 12-month follow-up included hypomania (2 episodes), one deep venous thrombophlebitits episode, and one episode each of back pain and appendicitis.1 No cognitive effects have been reported.

        In the double-blind controlled trial, 31 of 235 subjects (13%) experienced worsening of depression, and 25 of the 31 depressed subjects attempted suicide.9 Whether these effects were related to the depression or VNS stimulation is unclear. Side effects reported more frequently in the active treatment group than in the sham control group included voice alteration (68% vs. 38%), cough (29% vs. 9%), shortness of breath (23% vs. 14%), dysphagia (21% vs. 10%), and neck pain (21% vs. 10%).

         

         

        If VNS Is Intolerable

        Patients may deactivate the device with a magnet if they are uncomfortable. Pulse stimulation stops when a magnet is held against the left upper chest and resumes when the magnet is removed.

        Training

        Cyberonics plans to offer free VNS training to psychiatrists who practice at selected centers that accept treatment-resistant depression case referrals from primary care physicians, community psychiatrists, and other providers. Community psychiatrists who see treatment-resistant patients also are eligible for free training. For information, see Related resources.

        Related resources

         

        Disclosure

        The authors receive grant support from Neuronetics. They report no proprietary interest in the technology discussed in this article.

        References

         

        1. Marangell LB, Rush AJ, George MS, et al. Vagus nerve stimulation (VNS) for major depressive episodes: one year outcomes. Biol Psychiatry 2002;51:280-7.

        2. Sackeim HA, Rush AJ, George MS, et al. Vagus nerve stimulation (VNS) for treatment-resistant depression: efficacy, side effects, and predictors of outcome. Neuropsychopharmacology 2001;25(5):713-28.

        3. Henry TR, Bakay RA, Votaw JR, et al. Brain blood flow alterations induced in partial epilepsy I: acute effects at high and low levels of stimulation. Epilepsia 1998;39(9):983-90.

        4. Elger G, Hoppe C, Falkai P, et al. Vagus nerve stimulation is associated with mood improvements in epilepsy patients. Epilepsy Res 2000;42(2):203-10.

        5. George MS, Nahas Z, Bohning DE, et al. Vagus nerve stimulation therapy: a research update. Neurology 2002;59(6 suppl 4):S56-61.

        6. Rush AJ, George MS, Sackeim HA, et al. Vagus nerve stimulation (VNS) for treatment-resistant depression: a multicenter study. Biol Psychiatry 2000;47:276-86.

        7. Rush AJ, George MS, Sackeim HA, et al. Continuing benefit of VNS therapy over 2 years for treatment-resistant depression. San Juan, Puerto Rico: American College of Neuropsychopharmacology annual meeting, 2002.

        8. Cyberonics premarket approval application supplement (D-02/D-04 clinical report, PMA-S), submitted to FDA October 2003.

        9. Zwillich T. FDA panel recommends device for depression. WebMD Medical News June 17, 2004. Available at: http://my.webmd.com/content/article/89/100114.htm. Accessed August 9, 2005.

        Article PDF
        Author and Disclosure Information

         

        Jeffrey Rado, MD, MPH
        Instructor
        Philip G. Janicak, MD
        Professor

        Department of psychiatry, Rush University Medical Center, Chicago, IL

        Issue
        Current Psychiatry - 04(09)
        Publications
        Page Number
        78-82
        Sections
        Author and Disclosure Information

         

        Jeffrey Rado, MD, MPH
        Instructor
        Philip G. Janicak, MD
        Professor

        Department of psychiatry, Rush University Medical Center, Chicago, IL

        Author and Disclosure Information

         

        Jeffrey Rado, MD, MPH
        Instructor
        Philip G. Janicak, MD
        Professor

        Department of psychiatry, Rush University Medical Center, Chicago, IL

        Article PDF
        Article PDF

        What is vagus nerve stimulation’s (VNS) role in treating chronic or recurrent depression? Which patients would benefit from this implant, now FDA-approved for depression as well as epilepsy?

        Drawing from the evidence, this article discusses which patients with depression may be candidates for VNS, how it works, and its potential benefits and side effects.

        Clinical Applicability

        VNS is indicated for patients with chronic or recurrent treatment-resistant depression during an episode that has not responded to ≥4 adequate antidepressant treatment trials (defined as ≥3 on the Antidepressant Treatment History Form [ATHF]) (Table 1). Implantation theoretically promotes 100% adherence and reduces drug-drug interaction risk. Interactions between VNS and nonpsychotropics are possible but unlikely.

        Paradoxically, data suggest that patients with low to moderate resistance to antidepressant treatment (≤3 antidepressant trial failures) are most likely to benefit from VNS.1 Patients who had never received electroconvulsive therapy (ECT) (indicating relatively low treatment resistance) were nearly four times more likely than ECT-treated patients to respond to VNS.2 Conversely, 13 subjects who had not responded to ≥ 7 adequate treatment trials (indicating relatively severe treatment resistance) did not respond to VNS.2

        Table 1

        Vagus nerve stimulation device: Fast facts

         

        Brand name:
        Cyberonics Vagus Nerve Stimulation (VNS) Therapy System
        FDA-approved indications:
        Treatment-resistant depression (previously approved for treatment-refractory epilepsy)
        Manufacturer:
        Cyberonics
        Recommended use:
        Treating depressive episode that has not responded to ≥4 antidepressant trials or electroconvulsive therapy in a patient with chronic or recurrent depression
        Information on VNS remote device training:
        1-877-NOW-4-VNS (669-4867) or www.vnstherapy.com

        How VNS Works

        The vagus (10th cranial) nerve is a main efferent outflow tract for parasympathetic innervation of the abdomen and chest, regulating heart rate, acid secretion, and bowel motility.

        The largest component of the left vagus nerve—approximately 80%—conducts information about pain, hunger, and satiety. These fibers are also believed to contribute to VNS’ antidepressant effects by carrying information to the solitary nucleus of the medulla. From there, fibers project to the median raphe nucleus and locus coeruleus, key areas of serotonergic and noradrenergic innervation relevant to depression.

        Positron emission tomography studies suggest that VNS also increases blood flow to the thalamus, hypothalamus, and insula—brain areas considered relevant to mood disorders.3

        VNS requires subcutaneous implantation of a pacemaker-like pulse generator into the upper left chest. The generator is 6.9 mm thick and weighs 25 grams. Wires extend from the device into the left vagus nerve in the neck (Figure). A neurosurgeon usually performs the 1- to 2-hour outpatient procedure, although ENT, vascular, and general surgeons may also do the implant.

        The device sends electric pulses to the left vagus nerve every few seconds (Table 2). Using an accompanying hand-held device and a computer, the clinician programs the implant and adjusts stimulation parameters to ensure the correct amount of stimulation.

        FDA approved VNS in 1997 for refractory epilepsy. Clinical observations that VNS improved epilepsy patients’ mood spurred interest in its antidepressant effects.4 Preliminary data suggest VNS also could help manage anxiety disorders, obesity, pain syndromes, and Alzheimer’s disease.5

         

        Figure How VNS device works


        RTEmagicC_9be250daef.jpg.jpg

        Pacemaker-like VNSdevice is implanted into the upper left chest. Wires extending from the device transport electric pulses into the left vagus nerve in the neck, which carries information to areas of serotonergic and noradrenergic innervation relevant to depression.Table 2

        VNS stimulation parameters

         

        Frequency: 20 to 30 Hz
        Intensity: 0.25 mA (0.25 to 3.0 mA)
        Pulse width: 250 to 500 μs
        Duty cycle: 30 seconds on/5 minutes off

        Cost

        VNS implantation costs approximately $25,000, including the device, surgeon’s fee, and facility charge. Psychiatrists generally would initiate the referral process.

        Follow-up management fees for epilepsy are $150 to $250 per visit. Several follow-up visits are required after stimulation is started to verify the device is working, evaluate treatment response and tolerability, and adjust stimulation as needed. Thereafter, periodic visits are appropriate.

        Generally, insurers cover VNS as an epilepsy treatment; whether private insurers and Medicare will cover VNS for depression remains to be seen. Case mangers at Cyberonics, the device’s manufacturer, are on call to assist with VNS coverage, coding, and reimbursement issues (see Related resources).

        Because the internal implant’s battery life is 6 to 11 years, VNS therapy will likely be cost-effective for many patients, although follow-up surgery would be required to replace the battery. Costs of using VNS have not been compared with other antidepressant modalities.

        VNS’ Efficacy In Depression

        In an open-label trial, 60 patients ages 20 to 63 received VNS with no placebo or active comparator.2 Thirty had completed an open-label pilot study that showed VNS’ potential antidepressant effects.6 Before implantation, all subjects had:

         

         

         

        • a major depressive episode lasting >2 years or >4 lifetime major depressive episodes
        • nonresponse to ECT or ≥2 adequate antidepressant trials (ATHF scores >3) during their current major depressive episode (median duration: 4.7 years)
        • DSM-IV diagnosis of major depressive disorder or bipolar type I or II disorder depressed phase.
        • baseline scores ≥20 on the 28-item Hamilton Rating Scale for Depression (HRSD-28) and ≤50 on the Global Assessment of Functioning (GAF) scale.

        Two weeks after implantation, the stimulator was turned on and adjusted for another 2 weeks to the maximum tolerable dose. Patients then received 8 weeks of fixed-dose stimulation. Participants who had been taking an antidepressant, mood stabilizer, second-generation antipsychotic, or other psychotropic at the same dosages for ≥4 weeks before the study could continue their medications during the VNS trial (median concurrent treatments: 4).

        Three months after implantation, 18 of 59 subjects (30.5%) showed clinical response (≥50% improvement in HRSD-28 scores over baseline). Nine patients (15.3%) showed depression remission (HRSD-28 score ≤10). Median time to first response was 45.5 days.

        Twenty participants (34%) showed a ≥50% reduction in baseline Montgomery-Asberg Depression Rating Scale (MADRS) scores, and 22 (37%) showed Clinical Global Impression-Improvement Scale (CGI-I) scores improving to 1 or 2.

        Therapeutic effects did not differ among patients with unipolar and bipolar depression. Participants with mild to moderate depression (defined as 2 to 3 failed adequate trials) showed higher response rates (50% vs. 29.1%) than did those with more-severe depression (defined as ≥4 failed adequate trials).2

        Among 28 patients followed for 1 year, 13 (46%) met HRSD-28 response criteria (≥ 50% score reduction) and 8 (29%) met remission criteria (score ≤ 10), showing gradual improvement.1 After 2 years, 44% of patients met HDRS-28 response criteria, and 22% met remission criteria, showing sustained benefit.7 How many subjects were taking one or more concomitant psychotropics is unknown.

        In a double-blind controlled trial, 235 subjects ages 18 to 80 received VNS or a sham comparator.8 Treatment response and remission were defined as ≥50% reduction from baseline and ≤9, respectively, on the 24-item HRSD (HRSD-24). Patient selection criteria were similar to those of the open-label study.

        All patients received VNS implants, which were inactive the first 2 weeks. Patients were then randomly assigned to active treatment (stimulator turned on) or sham control (stimulator left off). After 10 weeks of treatment, HRSD-24, CGI-I, and MADRS scores were similar between the VNS and sham groups, but Inventory of Depressive Symptomatology Self Report (IDS-SR) scores improved much more in the active treatment group (P<0.03). Patients in the sham group then had their stimulators turned on.

        After 1 year of active treatment for both groups, response and remission rates more than doubled among 205 evaluable subjects (response: 14.4% to 29.8%; remission: 7.3% to 17.1%). MADRS and IDS-SR scores also improved. Three percent of subjects dropped out because of adverse events.

        Another analysis of these data revealed significant improvement among the VNS treatment group vs. a comparator-matched control group of treatment-resistant patients across 2 years.8

        Depression treatment among patients in the comparator group followed standard clinical practice.

        Side Effects

        Voice alteration or hoarseness was most commonly reported after 12 weeks in the open-label trial (55% of subjects). Headache (22%), cough (17%), shortness of breath (15%), neck pain (17%), dysphagia (20%), and pain (15%) were also reported.2 These effects emerge or increase with stimulation intensity and may be ameliorated by reducing the dose.

        Small risks of infection (1%) and nerve damage (1%) were reported. Leaving the stimulator off for 14 days after implantation decreases nerve damage risk. Pain at the incision site (experienced by 30%) resolved after 1 to 2 weeks.2 Other adverse events included:

         

        • hypomania in one bipolar patient; this was resolved by adjusting medication and reducing stimulation
        • leg pain in 2 subjects
        • worsened depression in 5 patients (2 of these may have been related to stimulation)
        • emesis and diarrhea in 1 subject.

        One patient with multiple cardiac risk factors developed a myocardial infarction but completed the trial after angioplasty and stent placement.2

        After 1 year in the open-label trial, no subjects dropped out because of adverse events. Common side events included voice alteration (21%), shortness of breath (7%), and neck pain (7%). More-serious adverse events reported between the acute trial and 12-month follow-up included hypomania (2 episodes), one deep venous thrombophlebitits episode, and one episode each of back pain and appendicitis.1 No cognitive effects have been reported.

        In the double-blind controlled trial, 31 of 235 subjects (13%) experienced worsening of depression, and 25 of the 31 depressed subjects attempted suicide.9 Whether these effects were related to the depression or VNS stimulation is unclear. Side effects reported more frequently in the active treatment group than in the sham control group included voice alteration (68% vs. 38%), cough (29% vs. 9%), shortness of breath (23% vs. 14%), dysphagia (21% vs. 10%), and neck pain (21% vs. 10%).

         

         

        If VNS Is Intolerable

        Patients may deactivate the device with a magnet if they are uncomfortable. Pulse stimulation stops when a magnet is held against the left upper chest and resumes when the magnet is removed.

        Training

        Cyberonics plans to offer free VNS training to psychiatrists who practice at selected centers that accept treatment-resistant depression case referrals from primary care physicians, community psychiatrists, and other providers. Community psychiatrists who see treatment-resistant patients also are eligible for free training. For information, see Related resources.

        Related resources

         

        Disclosure

        The authors receive grant support from Neuronetics. They report no proprietary interest in the technology discussed in this article.

        What is vagus nerve stimulation’s (VNS) role in treating chronic or recurrent depression? Which patients would benefit from this implant, now FDA-approved for depression as well as epilepsy?

        Drawing from the evidence, this article discusses which patients with depression may be candidates for VNS, how it works, and its potential benefits and side effects.

        Clinical Applicability

        VNS is indicated for patients with chronic or recurrent treatment-resistant depression during an episode that has not responded to ≥4 adequate antidepressant treatment trials (defined as ≥3 on the Antidepressant Treatment History Form [ATHF]) (Table 1). Implantation theoretically promotes 100% adherence and reduces drug-drug interaction risk. Interactions between VNS and nonpsychotropics are possible but unlikely.

        Paradoxically, data suggest that patients with low to moderate resistance to antidepressant treatment (≤3 antidepressant trial failures) are most likely to benefit from VNS.1 Patients who had never received electroconvulsive therapy (ECT) (indicating relatively low treatment resistance) were nearly four times more likely than ECT-treated patients to respond to VNS.2 Conversely, 13 subjects who had not responded to ≥ 7 adequate treatment trials (indicating relatively severe treatment resistance) did not respond to VNS.2

        Table 1

        Vagus nerve stimulation device: Fast facts

         

        Brand name:
        Cyberonics Vagus Nerve Stimulation (VNS) Therapy System
        FDA-approved indications:
        Treatment-resistant depression (previously approved for treatment-refractory epilepsy)
        Manufacturer:
        Cyberonics
        Recommended use:
        Treating depressive episode that has not responded to ≥4 antidepressant trials or electroconvulsive therapy in a patient with chronic or recurrent depression
        Information on VNS remote device training:
        1-877-NOW-4-VNS (669-4867) or www.vnstherapy.com

        How VNS Works

        The vagus (10th cranial) nerve is a main efferent outflow tract for parasympathetic innervation of the abdomen and chest, regulating heart rate, acid secretion, and bowel motility.

        The largest component of the left vagus nerve—approximately 80%—conducts information about pain, hunger, and satiety. These fibers are also believed to contribute to VNS’ antidepressant effects by carrying information to the solitary nucleus of the medulla. From there, fibers project to the median raphe nucleus and locus coeruleus, key areas of serotonergic and noradrenergic innervation relevant to depression.

        Positron emission tomography studies suggest that VNS also increases blood flow to the thalamus, hypothalamus, and insula—brain areas considered relevant to mood disorders.3

        VNS requires subcutaneous implantation of a pacemaker-like pulse generator into the upper left chest. The generator is 6.9 mm thick and weighs 25 grams. Wires extend from the device into the left vagus nerve in the neck (Figure). A neurosurgeon usually performs the 1- to 2-hour outpatient procedure, although ENT, vascular, and general surgeons may also do the implant.

        The device sends electric pulses to the left vagus nerve every few seconds (Table 2). Using an accompanying hand-held device and a computer, the clinician programs the implant and adjusts stimulation parameters to ensure the correct amount of stimulation.

        FDA approved VNS in 1997 for refractory epilepsy. Clinical observations that VNS improved epilepsy patients’ mood spurred interest in its antidepressant effects.4 Preliminary data suggest VNS also could help manage anxiety disorders, obesity, pain syndromes, and Alzheimer’s disease.5

         

        Figure How VNS device works


        RTEmagicC_9be250daef.jpg.jpg

        Pacemaker-like VNSdevice is implanted into the upper left chest. Wires extending from the device transport electric pulses into the left vagus nerve in the neck, which carries information to areas of serotonergic and noradrenergic innervation relevant to depression.Table 2

        VNS stimulation parameters

         

        Frequency: 20 to 30 Hz
        Intensity: 0.25 mA (0.25 to 3.0 mA)
        Pulse width: 250 to 500 μs
        Duty cycle: 30 seconds on/5 minutes off

        Cost

        VNS implantation costs approximately $25,000, including the device, surgeon’s fee, and facility charge. Psychiatrists generally would initiate the referral process.

        Follow-up management fees for epilepsy are $150 to $250 per visit. Several follow-up visits are required after stimulation is started to verify the device is working, evaluate treatment response and tolerability, and adjust stimulation as needed. Thereafter, periodic visits are appropriate.

        Generally, insurers cover VNS as an epilepsy treatment; whether private insurers and Medicare will cover VNS for depression remains to be seen. Case mangers at Cyberonics, the device’s manufacturer, are on call to assist with VNS coverage, coding, and reimbursement issues (see Related resources).

        Because the internal implant’s battery life is 6 to 11 years, VNS therapy will likely be cost-effective for many patients, although follow-up surgery would be required to replace the battery. Costs of using VNS have not been compared with other antidepressant modalities.

        VNS’ Efficacy In Depression

        In an open-label trial, 60 patients ages 20 to 63 received VNS with no placebo or active comparator.2 Thirty had completed an open-label pilot study that showed VNS’ potential antidepressant effects.6 Before implantation, all subjects had:

         

         

         

        • a major depressive episode lasting >2 years or >4 lifetime major depressive episodes
        • nonresponse to ECT or ≥2 adequate antidepressant trials (ATHF scores >3) during their current major depressive episode (median duration: 4.7 years)
        • DSM-IV diagnosis of major depressive disorder or bipolar type I or II disorder depressed phase.
        • baseline scores ≥20 on the 28-item Hamilton Rating Scale for Depression (HRSD-28) and ≤50 on the Global Assessment of Functioning (GAF) scale.

        Two weeks after implantation, the stimulator was turned on and adjusted for another 2 weeks to the maximum tolerable dose. Patients then received 8 weeks of fixed-dose stimulation. Participants who had been taking an antidepressant, mood stabilizer, second-generation antipsychotic, or other psychotropic at the same dosages for ≥4 weeks before the study could continue their medications during the VNS trial (median concurrent treatments: 4).

        Three months after implantation, 18 of 59 subjects (30.5%) showed clinical response (≥50% improvement in HRSD-28 scores over baseline). Nine patients (15.3%) showed depression remission (HRSD-28 score ≤10). Median time to first response was 45.5 days.

        Twenty participants (34%) showed a ≥50% reduction in baseline Montgomery-Asberg Depression Rating Scale (MADRS) scores, and 22 (37%) showed Clinical Global Impression-Improvement Scale (CGI-I) scores improving to 1 or 2.

        Therapeutic effects did not differ among patients with unipolar and bipolar depression. Participants with mild to moderate depression (defined as 2 to 3 failed adequate trials) showed higher response rates (50% vs. 29.1%) than did those with more-severe depression (defined as ≥4 failed adequate trials).2

        Among 28 patients followed for 1 year, 13 (46%) met HRSD-28 response criteria (≥ 50% score reduction) and 8 (29%) met remission criteria (score ≤ 10), showing gradual improvement.1 After 2 years, 44% of patients met HDRS-28 response criteria, and 22% met remission criteria, showing sustained benefit.7 How many subjects were taking one or more concomitant psychotropics is unknown.

        In a double-blind controlled trial, 235 subjects ages 18 to 80 received VNS or a sham comparator.8 Treatment response and remission were defined as ≥50% reduction from baseline and ≤9, respectively, on the 24-item HRSD (HRSD-24). Patient selection criteria were similar to those of the open-label study.

        All patients received VNS implants, which were inactive the first 2 weeks. Patients were then randomly assigned to active treatment (stimulator turned on) or sham control (stimulator left off). After 10 weeks of treatment, HRSD-24, CGI-I, and MADRS scores were similar between the VNS and sham groups, but Inventory of Depressive Symptomatology Self Report (IDS-SR) scores improved much more in the active treatment group (P<0.03). Patients in the sham group then had their stimulators turned on.

        After 1 year of active treatment for both groups, response and remission rates more than doubled among 205 evaluable subjects (response: 14.4% to 29.8%; remission: 7.3% to 17.1%). MADRS and IDS-SR scores also improved. Three percent of subjects dropped out because of adverse events.

        Another analysis of these data revealed significant improvement among the VNS treatment group vs. a comparator-matched control group of treatment-resistant patients across 2 years.8

        Depression treatment among patients in the comparator group followed standard clinical practice.

        Side Effects

        Voice alteration or hoarseness was most commonly reported after 12 weeks in the open-label trial (55% of subjects). Headache (22%), cough (17%), shortness of breath (15%), neck pain (17%), dysphagia (20%), and pain (15%) were also reported.2 These effects emerge or increase with stimulation intensity and may be ameliorated by reducing the dose.

        Small risks of infection (1%) and nerve damage (1%) were reported. Leaving the stimulator off for 14 days after implantation decreases nerve damage risk. Pain at the incision site (experienced by 30%) resolved after 1 to 2 weeks.2 Other adverse events included:

         

        • hypomania in one bipolar patient; this was resolved by adjusting medication and reducing stimulation
        • leg pain in 2 subjects
        • worsened depression in 5 patients (2 of these may have been related to stimulation)
        • emesis and diarrhea in 1 subject.

        One patient with multiple cardiac risk factors developed a myocardial infarction but completed the trial after angioplasty and stent placement.2

        After 1 year in the open-label trial, no subjects dropped out because of adverse events. Common side events included voice alteration (21%), shortness of breath (7%), and neck pain (7%). More-serious adverse events reported between the acute trial and 12-month follow-up included hypomania (2 episodes), one deep venous thrombophlebitits episode, and one episode each of back pain and appendicitis.1 No cognitive effects have been reported.

        In the double-blind controlled trial, 31 of 235 subjects (13%) experienced worsening of depression, and 25 of the 31 depressed subjects attempted suicide.9 Whether these effects were related to the depression or VNS stimulation is unclear. Side effects reported more frequently in the active treatment group than in the sham control group included voice alteration (68% vs. 38%), cough (29% vs. 9%), shortness of breath (23% vs. 14%), dysphagia (21% vs. 10%), and neck pain (21% vs. 10%).

         

         

        If VNS Is Intolerable

        Patients may deactivate the device with a magnet if they are uncomfortable. Pulse stimulation stops when a magnet is held against the left upper chest and resumes when the magnet is removed.

        Training

        Cyberonics plans to offer free VNS training to psychiatrists who practice at selected centers that accept treatment-resistant depression case referrals from primary care physicians, community psychiatrists, and other providers. Community psychiatrists who see treatment-resistant patients also are eligible for free training. For information, see Related resources.

        Related resources

         

        Disclosure

        The authors receive grant support from Neuronetics. They report no proprietary interest in the technology discussed in this article.

        References

         

        1. Marangell LB, Rush AJ, George MS, et al. Vagus nerve stimulation (VNS) for major depressive episodes: one year outcomes. Biol Psychiatry 2002;51:280-7.

        2. Sackeim HA, Rush AJ, George MS, et al. Vagus nerve stimulation (VNS) for treatment-resistant depression: efficacy, side effects, and predictors of outcome. Neuropsychopharmacology 2001;25(5):713-28.

        3. Henry TR, Bakay RA, Votaw JR, et al. Brain blood flow alterations induced in partial epilepsy I: acute effects at high and low levels of stimulation. Epilepsia 1998;39(9):983-90.

        4. Elger G, Hoppe C, Falkai P, et al. Vagus nerve stimulation is associated with mood improvements in epilepsy patients. Epilepsy Res 2000;42(2):203-10.

        5. George MS, Nahas Z, Bohning DE, et al. Vagus nerve stimulation therapy: a research update. Neurology 2002;59(6 suppl 4):S56-61.

        6. Rush AJ, George MS, Sackeim HA, et al. Vagus nerve stimulation (VNS) for treatment-resistant depression: a multicenter study. Biol Psychiatry 2000;47:276-86.

        7. Rush AJ, George MS, Sackeim HA, et al. Continuing benefit of VNS therapy over 2 years for treatment-resistant depression. San Juan, Puerto Rico: American College of Neuropsychopharmacology annual meeting, 2002.

        8. Cyberonics premarket approval application supplement (D-02/D-04 clinical report, PMA-S), submitted to FDA October 2003.

        9. Zwillich T. FDA panel recommends device for depression. WebMD Medical News June 17, 2004. Available at: http://my.webmd.com/content/article/89/100114.htm. Accessed August 9, 2005.

        References

         

        1. Marangell LB, Rush AJ, George MS, et al. Vagus nerve stimulation (VNS) for major depressive episodes: one year outcomes. Biol Psychiatry 2002;51:280-7.

        2. Sackeim HA, Rush AJ, George MS, et al. Vagus nerve stimulation (VNS) for treatment-resistant depression: efficacy, side effects, and predictors of outcome. Neuropsychopharmacology 2001;25(5):713-28.

        3. Henry TR, Bakay RA, Votaw JR, et al. Brain blood flow alterations induced in partial epilepsy I: acute effects at high and low levels of stimulation. Epilepsia 1998;39(9):983-90.

        4. Elger G, Hoppe C, Falkai P, et al. Vagus nerve stimulation is associated with mood improvements in epilepsy patients. Epilepsy Res 2000;42(2):203-10.

        5. George MS, Nahas Z, Bohning DE, et al. Vagus nerve stimulation therapy: a research update. Neurology 2002;59(6 suppl 4):S56-61.

        6. Rush AJ, George MS, Sackeim HA, et al. Vagus nerve stimulation (VNS) for treatment-resistant depression: a multicenter study. Biol Psychiatry 2000;47:276-86.

        7. Rush AJ, George MS, Sackeim HA, et al. Continuing benefit of VNS therapy over 2 years for treatment-resistant depression. San Juan, Puerto Rico: American College of Neuropsychopharmacology annual meeting, 2002.

        8. Cyberonics premarket approval application supplement (D-02/D-04 clinical report, PMA-S), submitted to FDA October 2003.

        9. Zwillich T. FDA panel recommends device for depression. WebMD Medical News June 17, 2004. Available at: http://my.webmd.com/content/article/89/100114.htm. Accessed August 9, 2005.

        Issue
        Current Psychiatry - 04(09)
        Issue
        Current Psychiatry - 04(09)
        Page Number
        78-82
        Page Number
        78-82
        Publications
        Publications
        Article Type
        Display Headline
        Vagus nerve stimulation
        Display Headline
        Vagus nerve stimulation
        Sections
        Disallow All Ads
        Alternative CME
        Article PDF Media
        Image
        Disable zoom
        Off

        Therapy-resistant major depression The attraction of magnetism: How effective—and safe—is rTMS?

        Article Type
        Changed
        Tue, 12/11/2018 - 15:11
        Display Headline
        Therapy-resistant major depression The attraction of magnetism: How effective—and safe—is rTMS?

        Using magnets to improve health is sometimes hawked in dubious classified ads and “infomercials.” However, a legitimate use of magnetism—repetitive transcranial magnetic stimulation (rTMS)—is showing promise in treating severe depression (Box) 1-4 and other psychiatric disorders.

        Patients or their families are likely to ask psychiatrists about rTMS as more becomes known about this investigational technology. Drawing from our experience and the evidence, we offer an update on whether rTMS may be an alternative for treating depression and address issues that must be resolved before it could be used in clinical practice.

        WHAT IS RTMS?

        rTMS consists of a series of magnetic pulses produced by a stimulator, which can be adjusted for:

        • coil type and placement
        • stimulation site, intensity, frequency, and number
        • amount of time between stimulations
        • treatment duration.

        Box

        rTMS: A new antidepressant technology?

        In 1985, Barker and colleagues developed single-pulse transcranial magnetic stimulation to examine motor cortex function.1 The single-pulse mechanism they discovered was subsequently adapted to deliver repetitive pulses and is referred to as repetitive transcranial magnetic stimulation (rTMS).

        How rTMS works. Transcranial magnetic stimulation uses an electromagnetic coil applied to the head to produce an intense, localized, fluctuating magnetic field that passes unimpeded into a small area of the brain, inducing an electrical current. This results in neuronal depolarization in a localized area under the coil, and possibly distal effects as well.2 During the neurophysiological studies, it was discovered that subjects also experienced a change in mood.

        Antidepressant effects. Similar physiologic effects induced by rTMS, electroconvulsive therapy (ECT), and antidepressants on the endocrine system, sleep parameters, and biochemical measures suggest antidepressant properties.3 In 1993, the first published study examining rTMS in psychiatric patients reported reduced depressive symptoms in two subjects.4 Since then, several clinical trials have examined rTMS’ antidepressive effects. In 2001, Canada’s Health Ministry approved rTMS for treating major depression. In the United States, rTMS remains investigational and is FDA-approved only for clinical trials.

        Coil type and placement. Initial studies involved stimulation—typically low-frequency—over the vertex, but most subsequent rTMS trials in depression have stimulated the left dorsolateral prefrontal cortex. Neuroimaging studies have shown prefrontal functioning abnormalities in depressed subjects, and it is hypothesized that stimulating this area (plus possible distal effects) may produce an antidepressant effect.5

        Various configurations have been used, but circular and figure-eight-shaped coils are most common. These flat coils are made of tightly wound ferromagnetic material such as copper, enclosed in a heavy plastic cover. With the figure-eight coil, the intersection of the two loops produces the strongest magnetic field.

        Stimulation site. Stimulation intensity depends on the individual’s motor threshold, and the site can be determined visually or electrophysiologically.

        • With the visual method, the motor threshold over the left primary motor cortex site for the first dorsal interosseous muscle (FDI) or the abductor pollius brevis (APB) is determined by iteration. This involves placing the coil at a progression of sites and increasing stimulation intensity until reliable (in 5 of 10 stimulations) contractions are seen in the right FDI or APB.
        • Similarly, the electrophysiologic method uses 5 of 10 motorevoked potentials of 50 microvolts to locate the site.

        The only small trial that compared visual and electrophysiologic site determination showed similar results with both methods.6 The most common stimulation site is the left dorsolateral prefrontal cortex, 5 cm anterior and parasagittal to the FDI or APB motor cortex. Alternately, frameless stereotactic systems or the international 10-20 proportional system used in EEG labs have been recommended to target sites more accurately.

        Stimulus intensity. Each individual’s motor threshold determines stimulus intensity. Using functional MRI studies, researchers from the Medical University of South Carolina concluded that higher stimulation intensity relative to the motor threshold may have a more robust effect, as the magnetic field declines with distance from the coil.7 However, intensities >120% of the motor threshold are generally avoided because of possible increased seizure risk.9

        Frequency of stimulation. Most researchers apply frequencies of 1 to 20 Hz over the left dorsolateral prefrontal cortex, but also use lower frequencies (<1 Hz) over the right dorsolateral prefrontal cortex. Using higher frequencies in major depression is attractive in theory because of:

        • the reported association of decreased regional cerebral blood flow with hypometabolism in the left dorsolateral prefrontal cortex
        • higher-frequency stimulation’s ability to produce temporary excitation and neuronal depolarization.

        Number of stimulations. The number of stimulations is determined by frequency (Hz) and stimulation train duration (for example, 10 Hz for 5 seconds equals 50 stimulations). A typical treatment session incorporates 10 to 30 stimulation trains several seconds apart (the inter-train interval). Thus, a typical session delivers 1,000 to 1,200 stimulations. In studies of unmedicated depressed patients, the total number of stimulations has varied from 8,000 to 32,000 per treatment course.

         

         

        Duration between two stimulation trains. Chen et al have demonstrated that shorter (<1 second) inter-train intervals increase seizure risk with higher frequencies (such as 20 Hz) and intensities (>100% of motor threshold) of stimulation.9 Based on their studies with healthy volunteers, they recommended several “safe” ranges (such as 5 seconds at 110% of motor threshold). Most trials use 30- to 60-second inter-train intervals.

        Most treatments continue 2 to 4 weeks, Monday through Friday, although more frequent treatments are being studied.

        EFFICACY FOR DEPRESSION

        Most studies of rTMS in depression have compared real rTMS to a sham control or electroconvulsive therapy (ECT).

        In earlier studies, the sham procedure typically involved tilting the coil away from the skull. This method has been questioned, however, because of evidence of neuronal depolarization.10

        More recent sham coils mimic the real coils’ sound and sensation, without magnetic stimulation.

        Despite these methodologic problems and some mixed results, depressed patients receiving rTMS show more favorable results than those receiving sham rTMS.11,12 Several meta-analyses have attempted to quantify rTMS’ efficacy for depression:

        • Holtzheimer et al concluded that rTMS was statistically superior to sham rTMS, but the clinical significance of these findings was modest in a population of mostly outpatients with less-severe depression.13
        • Burt et al found a statistically strong antidepressant effect, but its magnitude varied and few of the studies yielded a substantial clinical response or remission. The team also noted that rTMS’ long-term efficacy or adverse effects are unknown.14
        • Kozel et al concluded that left prefrontal rTMS rendered a significant antidepressant effect with measurable clinical improvement.15
        • Gershon et al16 supported an antidepressant effect for rTMS when compared with sham rTMS or ECT.

        Ongoing rTMS research includes subjects with many types of mild to severe psychiatric illnesses, including major depression, obsessive-compulsive disorder, and psychosis. Typically, patients referred for experimental approaches have not responded to or tolerated available treatments. Exclusion criteria used by most rTMS studies are listed in the Table.

        Table

        Medical conditions that preclude use of rTMS

        Serious medical conditions
         History of seizures
         Increased intracranial pressure
         Serious head trauma
        Myocardial infarction within the past 6 months
        Pregnancy or childbearing potential (unless reliable contraception is being used)
        Intracranial metallic implants
        Pacemakers or other implanted devices

        rTMS vs. ECT. Four randomized, controlled trials have compared rTMS with ECT for treating severely ill, often medication-resistant patients.17-20 Although their methodologies differed, all four studies concluded that rTMS and ECT offer similar efficacy, except that rTMS may be less effective for treating psychotic depression.

        One study found ECT more effective than rTMS for psychotic depression, although the patients who received ECT were also treated with antipsychotics and/or antidepressants.17 Our study,19 which did not use these agents, has not corroborated this observation. Preliminary data also indicate comparable relapse rates following acute ECT and rTMS when subjects are followed on maintenance medication.21

        ADVERSE EFFECTS

        The potential adverse effects of new treatments must always be considered. Thus far, rTMS appears to produce minimal, relatively benign complications, including:

        • mild discomfort at the stimulation site
        • localized muscle twitching during stimulation
        • mild post-treatment headaches—believed caused by muscle contractions—which usually respond to aspirin or acetaminophen
        • treatment stimulation-related seizures (rarely).8

        The rTMS device makes a loud clicking noise, and subjects wear protective ear plugs during treatment.

        Patient experience. The first rTMS session—during which the patient’s motor threshold is determined—can last up to 45 minutes. Subsequent sessions are usually 15 to 20 minutes. Patients are typically apprehensive before the first session but become more relaxed with experience and tolerate the treatments easily.

        During the procedure, many patients describe a tapping sensation on the forehead, and some experience slight muscle twitching around the eye or corner of the mouth. As the coil warms, the skin it touches sometimes flushes pink, although this does not seem to bother our patients. They can return to their daily routines immediately after a session.

        rTMS for major depression. In our experience, rTMS may help patients with major depression. For example, one patient diagnosed with a major depressive episode with psychotic features was referred to our study comparing rTMS with ECT.19 Her depression had lasted several months, with partial response to ECT treatments. She signed informed consent and was randomly assigned to receive rTMS treatment.

        At study admission, the patient’s Hamilton Depression Rating Scale (HDRS) score was 48, indicating moderate to severe depression. Following 10 rTMS sessions, her HDRS score had dropped to 2, with remission of symptoms. No follow-up results were documented.

        Cognitive effects. Whereas mood disorders are associated with medication-independent neuropsychological deficits, most studies have found no adverse cognitive effects with rTMS.22 Indeed, some of our rTMS patients have improved in certain cognitive tests, although this may be explained by test-retest effects or better attention and concentration associated with mood improvement.

         

         

        Figure Potential roles for rTMS in treating major depression


        RTEmagicC_af5a338892.jpg.jpg

        Solid lines represent current standards of practice. Dotted lines represent hypothetical roles for rTMS.

        Source: Adapted and reprinted with permission from Dowd et al. Is repetitive transcranial magnetic stimulation an alternative to ECTfor the treatment of depression? Contemp Psychiatry 2002;1:1-10.

        POTENTIAL ROLE FOR rTMS

        Today’s standard treatment of major depressive episodes begins with an antidepressant (plus an antipsychotic, if necessary) and proceeds to augmentation strategies if response is insufficient. rTMS may one day become an augmentation or monotherapy option for patients who do not respond sufficiently to standard treatments (Figure).

        ECT treatment may be initiated if a patient has had a prior good response to ECT, is intolerant to medication, or prefers ECT. In that case, rTMS may be used as an alternate initial treatment or with ECT. Thus, rTMS may be used:

        • to augment antidepressants
        • as an alternative to antidepressants or ECT
        • or sequentially with ECT.

        Before that can happen, however, optimal treatment parameters need to be clarified by larger, well-designed, controlled studies comparing rTMS to a valid sham treatment, antidepressants, and ECT.

        Related resources

        • International Society for Transcranial Stimulation. www.ists.unibe.ch/
        • Repetitive Transcranial Magnetic Stimulation Research Clinic at Yale-New Haven Psychiatric Hospital.

        Disclosure

        The authors report that they have no proprietary interest in the technology discussed in this article.

        References

        1. Barker A, Jalinous R, Freeston I. Non-invasive magnetic stimulation of human motor cortex. Lancet 1985;1:1106-7.

        2. Lisanby SH, Datto CJ, Szuba MP. ECT and rTMS: past, present, and future. Depress Anxiety 2000;12:115-17.

        3. Post A, Keck PE, Jr. Transcranial magnetic stimulation as a therapeutic tool in psychiatry: what do we know about the neurobiological mechanisms? J Psychiatr Res 2001;35:193-215.

        4. Holfich G, Kasper S, Hufnagel A, et al. Application of transcranial magnetic stimulation in treatment of drug resistant major depression—a report of two cases. Human Psychopharmacol 1993;8:361-5.

        5. George MS, Nahas Z, Speer AM, et al. Transcranial magnetic stimulation—a new method for investigating the neuroanatomy of depression. In: Ebert D, Ebmeier K (eds). New models for depression. New York: Karger, 1998;94-122.

        6. Pridmore A, Americo Fernandes Filho J, Nahas Z, et al. Motor threshold in transcranial magnetic stimulation: a comparison of a neurophysiological method and a visualization of movement method. J ECT 1998;14(1):25-7.

        7. Kozel FA, Nahas Z, deBrux C, et al. How coil-cortex distance relates to age, motor threshold, and antidepressant response to repetitive transcranial magnetic stimulation. J Neuropsychiatry Clin Neurosci 2000;13:376-84.

        8. Wassermann EM. Risk and safety of repetitive transcranial magnetic stimulation: report and suggested guidelines from the International Workshop on the Safety of Repetitive Transcranial Magnetic Stimulation, 1996. Electroencephalogr Clin Neurophysiol 1998;108:1-16.

        9. Chen R, Gerloff C, Classen J, et al. Safety of different inter-train intervals for repetitive transcranial magnetic stimulation and recommendations for safe ranges of stimulation parameters. Electroencephalogr Clin Neurophysiol 1997;105:415-21.

        10. Loo CK, Taylor JL, Gandevia SC, et al. Transcranial magnetic stimulation in controlled treatment studies: Are some “sham” forms active? Biol Psychiatry. 2000;47:325-31.

        11. George MS, Nahas Z, Molloy M, et al. A controlled trial of daily left prefrontal cortex TMS for treating depression. Biol Psychiatry 2000;48:962-70.

        12. Berman RM, Narasimhan M, Sanacora G, et al. A randomized clinical trial of repetitive transcranial magnetic stimulation in the treatment of major depression. Biol Psychiatry 2000;47:332-7.

        13. Holtzheimer PE, Russo J, Avery D. A meta-analysis of repetitive transcranial magnetic stimulation in the treatment of depression. Psychopharmacol Bull 2001;35:149-69.

        14. Burt T, Lisanby SH, Sackeim HA. Neuropsychiatric applications of transcranial magnetic stimulation: a meta-analysis. Int J Neuropsychopharmacol 2002;5:73-103.

        15. Kozel FE, George MS. Meta-analysis of left prefrontal repetitive transcranial magnetic stimulation (rTMS) to treat depression. J Psychiatr Pract 2002;8:270-5.

        16. Gershon AA, Dannon PN, Grunhaus L. Transcranial magnetic stimulation in the treatment of depression. Am JPsychiatry 2003;160(5):835-45.

        17. Grunhaus L, Dannon PN, Schreiber S, et al. Repetitive transcranial magnetic stimulation is as effective as electroconvulsive therapy in the treatment of nondelusional major depressive disorder: an open study. Biol Psychiatry 2000;47:314-24.

        18. Pridmore S, Bruno R, Turnier-Shea Y, et al. Comparison of unlimited numbers of rapid transcranial magnetic stimulation and ECT treatment sessions in major depression episodes. Int J Neuropsychopharmacol 2000;3:129-34.

        19. Janicak PG, Dowd SM, Martis B, et al. Repetitive transcranial magnetic stimulation versus electroconvulsive therapy for major depression: preliminary results of a randomized trial. Biol Psychiatry 2002;51:659-67

        20. Grunhaus L, Schreiber S, Dolberg OT, et al. A randomized controlled comparison of electroconvulsive therapy and repetitive transcranial magnetic stimulation in severe and resistant nonpsychotic major depression. Biol Psychiatry 2003;53:324-31.

        21. Dannon PH, Dolberg OT, Schreiber S, Grunhaus L. Three and six month outcome following courses of either ECT or rTMS in a population of severely depressed individuals—preliminary report. Biol Psychiatry 2002;15:687-90.

        22. Martis B, Alam D, Dowd SM, et al. Neurocognitive effects of repetitive transcranial magnetic stimulation in severe major depression. Clin Neurophysiology (in press).

        Author and Disclosure Information

        Sheila M. Dowd, PhD
        Assistant professor Department of psychiatry University of Illinois at Chicago

        Philip G. Janicak, MD
        Professor of psychiatry and pharmacology Medical director, Psychiatric Clinical Research Center University of Illinois at Chicago

        Issue
        Current Psychiatry - 02(06)
        Publications
        Topics
        Page Number
        59-66
        Sections
        Author and Disclosure Information

        Sheila M. Dowd, PhD
        Assistant professor Department of psychiatry University of Illinois at Chicago

        Philip G. Janicak, MD
        Professor of psychiatry and pharmacology Medical director, Psychiatric Clinical Research Center University of Illinois at Chicago

        Author and Disclosure Information

        Sheila M. Dowd, PhD
        Assistant professor Department of psychiatry University of Illinois at Chicago

        Philip G. Janicak, MD
        Professor of psychiatry and pharmacology Medical director, Psychiatric Clinical Research Center University of Illinois at Chicago

        Using magnets to improve health is sometimes hawked in dubious classified ads and “infomercials.” However, a legitimate use of magnetism—repetitive transcranial magnetic stimulation (rTMS)—is showing promise in treating severe depression (Box) 1-4 and other psychiatric disorders.

        Patients or their families are likely to ask psychiatrists about rTMS as more becomes known about this investigational technology. Drawing from our experience and the evidence, we offer an update on whether rTMS may be an alternative for treating depression and address issues that must be resolved before it could be used in clinical practice.

        WHAT IS RTMS?

        rTMS consists of a series of magnetic pulses produced by a stimulator, which can be adjusted for:

        • coil type and placement
        • stimulation site, intensity, frequency, and number
        • amount of time between stimulations
        • treatment duration.

        Box

        rTMS: A new antidepressant technology?

        In 1985, Barker and colleagues developed single-pulse transcranial magnetic stimulation to examine motor cortex function.1 The single-pulse mechanism they discovered was subsequently adapted to deliver repetitive pulses and is referred to as repetitive transcranial magnetic stimulation (rTMS).

        How rTMS works. Transcranial magnetic stimulation uses an electromagnetic coil applied to the head to produce an intense, localized, fluctuating magnetic field that passes unimpeded into a small area of the brain, inducing an electrical current. This results in neuronal depolarization in a localized area under the coil, and possibly distal effects as well.2 During the neurophysiological studies, it was discovered that subjects also experienced a change in mood.

        Antidepressant effects. Similar physiologic effects induced by rTMS, electroconvulsive therapy (ECT), and antidepressants on the endocrine system, sleep parameters, and biochemical measures suggest antidepressant properties.3 In 1993, the first published study examining rTMS in psychiatric patients reported reduced depressive symptoms in two subjects.4 Since then, several clinical trials have examined rTMS’ antidepressive effects. In 2001, Canada’s Health Ministry approved rTMS for treating major depression. In the United States, rTMS remains investigational and is FDA-approved only for clinical trials.

        Coil type and placement. Initial studies involved stimulation—typically low-frequency—over the vertex, but most subsequent rTMS trials in depression have stimulated the left dorsolateral prefrontal cortex. Neuroimaging studies have shown prefrontal functioning abnormalities in depressed subjects, and it is hypothesized that stimulating this area (plus possible distal effects) may produce an antidepressant effect.5

        Various configurations have been used, but circular and figure-eight-shaped coils are most common. These flat coils are made of tightly wound ferromagnetic material such as copper, enclosed in a heavy plastic cover. With the figure-eight coil, the intersection of the two loops produces the strongest magnetic field.

        Stimulation site. Stimulation intensity depends on the individual’s motor threshold, and the site can be determined visually or electrophysiologically.

        • With the visual method, the motor threshold over the left primary motor cortex site for the first dorsal interosseous muscle (FDI) or the abductor pollius brevis (APB) is determined by iteration. This involves placing the coil at a progression of sites and increasing stimulation intensity until reliable (in 5 of 10 stimulations) contractions are seen in the right FDI or APB.
        • Similarly, the electrophysiologic method uses 5 of 10 motorevoked potentials of 50 microvolts to locate the site.

        The only small trial that compared visual and electrophysiologic site determination showed similar results with both methods.6 The most common stimulation site is the left dorsolateral prefrontal cortex, 5 cm anterior and parasagittal to the FDI or APB motor cortex. Alternately, frameless stereotactic systems or the international 10-20 proportional system used in EEG labs have been recommended to target sites more accurately.

        Stimulus intensity. Each individual’s motor threshold determines stimulus intensity. Using functional MRI studies, researchers from the Medical University of South Carolina concluded that higher stimulation intensity relative to the motor threshold may have a more robust effect, as the magnetic field declines with distance from the coil.7 However, intensities >120% of the motor threshold are generally avoided because of possible increased seizure risk.9

        Frequency of stimulation. Most researchers apply frequencies of 1 to 20 Hz over the left dorsolateral prefrontal cortex, but also use lower frequencies (<1 Hz) over the right dorsolateral prefrontal cortex. Using higher frequencies in major depression is attractive in theory because of:

        • the reported association of decreased regional cerebral blood flow with hypometabolism in the left dorsolateral prefrontal cortex
        • higher-frequency stimulation’s ability to produce temporary excitation and neuronal depolarization.

        Number of stimulations. The number of stimulations is determined by frequency (Hz) and stimulation train duration (for example, 10 Hz for 5 seconds equals 50 stimulations). A typical treatment session incorporates 10 to 30 stimulation trains several seconds apart (the inter-train interval). Thus, a typical session delivers 1,000 to 1,200 stimulations. In studies of unmedicated depressed patients, the total number of stimulations has varied from 8,000 to 32,000 per treatment course.

         

         

        Duration between two stimulation trains. Chen et al have demonstrated that shorter (<1 second) inter-train intervals increase seizure risk with higher frequencies (such as 20 Hz) and intensities (>100% of motor threshold) of stimulation.9 Based on their studies with healthy volunteers, they recommended several “safe” ranges (such as 5 seconds at 110% of motor threshold). Most trials use 30- to 60-second inter-train intervals.

        Most treatments continue 2 to 4 weeks, Monday through Friday, although more frequent treatments are being studied.

        EFFICACY FOR DEPRESSION

        Most studies of rTMS in depression have compared real rTMS to a sham control or electroconvulsive therapy (ECT).

        In earlier studies, the sham procedure typically involved tilting the coil away from the skull. This method has been questioned, however, because of evidence of neuronal depolarization.10

        More recent sham coils mimic the real coils’ sound and sensation, without magnetic stimulation.

        Despite these methodologic problems and some mixed results, depressed patients receiving rTMS show more favorable results than those receiving sham rTMS.11,12 Several meta-analyses have attempted to quantify rTMS’ efficacy for depression:

        • Holtzheimer et al concluded that rTMS was statistically superior to sham rTMS, but the clinical significance of these findings was modest in a population of mostly outpatients with less-severe depression.13
        • Burt et al found a statistically strong antidepressant effect, but its magnitude varied and few of the studies yielded a substantial clinical response or remission. The team also noted that rTMS’ long-term efficacy or adverse effects are unknown.14
        • Kozel et al concluded that left prefrontal rTMS rendered a significant antidepressant effect with measurable clinical improvement.15
        • Gershon et al16 supported an antidepressant effect for rTMS when compared with sham rTMS or ECT.

        Ongoing rTMS research includes subjects with many types of mild to severe psychiatric illnesses, including major depression, obsessive-compulsive disorder, and psychosis. Typically, patients referred for experimental approaches have not responded to or tolerated available treatments. Exclusion criteria used by most rTMS studies are listed in the Table.

        Table

        Medical conditions that preclude use of rTMS

        Serious medical conditions
         History of seizures
         Increased intracranial pressure
         Serious head trauma
        Myocardial infarction within the past 6 months
        Pregnancy or childbearing potential (unless reliable contraception is being used)
        Intracranial metallic implants
        Pacemakers or other implanted devices

        rTMS vs. ECT. Four randomized, controlled trials have compared rTMS with ECT for treating severely ill, often medication-resistant patients.17-20 Although their methodologies differed, all four studies concluded that rTMS and ECT offer similar efficacy, except that rTMS may be less effective for treating psychotic depression.

        One study found ECT more effective than rTMS for psychotic depression, although the patients who received ECT were also treated with antipsychotics and/or antidepressants.17 Our study,19 which did not use these agents, has not corroborated this observation. Preliminary data also indicate comparable relapse rates following acute ECT and rTMS when subjects are followed on maintenance medication.21

        ADVERSE EFFECTS

        The potential adverse effects of new treatments must always be considered. Thus far, rTMS appears to produce minimal, relatively benign complications, including:

        • mild discomfort at the stimulation site
        • localized muscle twitching during stimulation
        • mild post-treatment headaches—believed caused by muscle contractions—which usually respond to aspirin or acetaminophen
        • treatment stimulation-related seizures (rarely).8

        The rTMS device makes a loud clicking noise, and subjects wear protective ear plugs during treatment.

        Patient experience. The first rTMS session—during which the patient’s motor threshold is determined—can last up to 45 minutes. Subsequent sessions are usually 15 to 20 minutes. Patients are typically apprehensive before the first session but become more relaxed with experience and tolerate the treatments easily.

        During the procedure, many patients describe a tapping sensation on the forehead, and some experience slight muscle twitching around the eye or corner of the mouth. As the coil warms, the skin it touches sometimes flushes pink, although this does not seem to bother our patients. They can return to their daily routines immediately after a session.

        rTMS for major depression. In our experience, rTMS may help patients with major depression. For example, one patient diagnosed with a major depressive episode with psychotic features was referred to our study comparing rTMS with ECT.19 Her depression had lasted several months, with partial response to ECT treatments. She signed informed consent and was randomly assigned to receive rTMS treatment.

        At study admission, the patient’s Hamilton Depression Rating Scale (HDRS) score was 48, indicating moderate to severe depression. Following 10 rTMS sessions, her HDRS score had dropped to 2, with remission of symptoms. No follow-up results were documented.

        Cognitive effects. Whereas mood disorders are associated with medication-independent neuropsychological deficits, most studies have found no adverse cognitive effects with rTMS.22 Indeed, some of our rTMS patients have improved in certain cognitive tests, although this may be explained by test-retest effects or better attention and concentration associated with mood improvement.

         

         

        Figure Potential roles for rTMS in treating major depression


        RTEmagicC_af5a338892.jpg.jpg

        Solid lines represent current standards of practice. Dotted lines represent hypothetical roles for rTMS.

        Source: Adapted and reprinted with permission from Dowd et al. Is repetitive transcranial magnetic stimulation an alternative to ECTfor the treatment of depression? Contemp Psychiatry 2002;1:1-10.

        POTENTIAL ROLE FOR rTMS

        Today’s standard treatment of major depressive episodes begins with an antidepressant (plus an antipsychotic, if necessary) and proceeds to augmentation strategies if response is insufficient. rTMS may one day become an augmentation or monotherapy option for patients who do not respond sufficiently to standard treatments (Figure).

        ECT treatment may be initiated if a patient has had a prior good response to ECT, is intolerant to medication, or prefers ECT. In that case, rTMS may be used as an alternate initial treatment or with ECT. Thus, rTMS may be used:

        • to augment antidepressants
        • as an alternative to antidepressants or ECT
        • or sequentially with ECT.

        Before that can happen, however, optimal treatment parameters need to be clarified by larger, well-designed, controlled studies comparing rTMS to a valid sham treatment, antidepressants, and ECT.

        Related resources

        • International Society for Transcranial Stimulation. www.ists.unibe.ch/
        • Repetitive Transcranial Magnetic Stimulation Research Clinic at Yale-New Haven Psychiatric Hospital.

        Disclosure

        The authors report that they have no proprietary interest in the technology discussed in this article.

        Using magnets to improve health is sometimes hawked in dubious classified ads and “infomercials.” However, a legitimate use of magnetism—repetitive transcranial magnetic stimulation (rTMS)—is showing promise in treating severe depression (Box) 1-4 and other psychiatric disorders.

        Patients or their families are likely to ask psychiatrists about rTMS as more becomes known about this investigational technology. Drawing from our experience and the evidence, we offer an update on whether rTMS may be an alternative for treating depression and address issues that must be resolved before it could be used in clinical practice.

        WHAT IS RTMS?

        rTMS consists of a series of magnetic pulses produced by a stimulator, which can be adjusted for:

        • coil type and placement
        • stimulation site, intensity, frequency, and number
        • amount of time between stimulations
        • treatment duration.

        Box

        rTMS: A new antidepressant technology?

        In 1985, Barker and colleagues developed single-pulse transcranial magnetic stimulation to examine motor cortex function.1 The single-pulse mechanism they discovered was subsequently adapted to deliver repetitive pulses and is referred to as repetitive transcranial magnetic stimulation (rTMS).

        How rTMS works. Transcranial magnetic stimulation uses an electromagnetic coil applied to the head to produce an intense, localized, fluctuating magnetic field that passes unimpeded into a small area of the brain, inducing an electrical current. This results in neuronal depolarization in a localized area under the coil, and possibly distal effects as well.2 During the neurophysiological studies, it was discovered that subjects also experienced a change in mood.

        Antidepressant effects. Similar physiologic effects induced by rTMS, electroconvulsive therapy (ECT), and antidepressants on the endocrine system, sleep parameters, and biochemical measures suggest antidepressant properties.3 In 1993, the first published study examining rTMS in psychiatric patients reported reduced depressive symptoms in two subjects.4 Since then, several clinical trials have examined rTMS’ antidepressive effects. In 2001, Canada’s Health Ministry approved rTMS for treating major depression. In the United States, rTMS remains investigational and is FDA-approved only for clinical trials.

        Coil type and placement. Initial studies involved stimulation—typically low-frequency—over the vertex, but most subsequent rTMS trials in depression have stimulated the left dorsolateral prefrontal cortex. Neuroimaging studies have shown prefrontal functioning abnormalities in depressed subjects, and it is hypothesized that stimulating this area (plus possible distal effects) may produce an antidepressant effect.5

        Various configurations have been used, but circular and figure-eight-shaped coils are most common. These flat coils are made of tightly wound ferromagnetic material such as copper, enclosed in a heavy plastic cover. With the figure-eight coil, the intersection of the two loops produces the strongest magnetic field.

        Stimulation site. Stimulation intensity depends on the individual’s motor threshold, and the site can be determined visually or electrophysiologically.

        • With the visual method, the motor threshold over the left primary motor cortex site for the first dorsal interosseous muscle (FDI) or the abductor pollius brevis (APB) is determined by iteration. This involves placing the coil at a progression of sites and increasing stimulation intensity until reliable (in 5 of 10 stimulations) contractions are seen in the right FDI or APB.
        • Similarly, the electrophysiologic method uses 5 of 10 motorevoked potentials of 50 microvolts to locate the site.

        The only small trial that compared visual and electrophysiologic site determination showed similar results with both methods.6 The most common stimulation site is the left dorsolateral prefrontal cortex, 5 cm anterior and parasagittal to the FDI or APB motor cortex. Alternately, frameless stereotactic systems or the international 10-20 proportional system used in EEG labs have been recommended to target sites more accurately.

        Stimulus intensity. Each individual’s motor threshold determines stimulus intensity. Using functional MRI studies, researchers from the Medical University of South Carolina concluded that higher stimulation intensity relative to the motor threshold may have a more robust effect, as the magnetic field declines with distance from the coil.7 However, intensities >120% of the motor threshold are generally avoided because of possible increased seizure risk.9

        Frequency of stimulation. Most researchers apply frequencies of 1 to 20 Hz over the left dorsolateral prefrontal cortex, but also use lower frequencies (<1 Hz) over the right dorsolateral prefrontal cortex. Using higher frequencies in major depression is attractive in theory because of:

        • the reported association of decreased regional cerebral blood flow with hypometabolism in the left dorsolateral prefrontal cortex
        • higher-frequency stimulation’s ability to produce temporary excitation and neuronal depolarization.

        Number of stimulations. The number of stimulations is determined by frequency (Hz) and stimulation train duration (for example, 10 Hz for 5 seconds equals 50 stimulations). A typical treatment session incorporates 10 to 30 stimulation trains several seconds apart (the inter-train interval). Thus, a typical session delivers 1,000 to 1,200 stimulations. In studies of unmedicated depressed patients, the total number of stimulations has varied from 8,000 to 32,000 per treatment course.

         

         

        Duration between two stimulation trains. Chen et al have demonstrated that shorter (<1 second) inter-train intervals increase seizure risk with higher frequencies (such as 20 Hz) and intensities (>100% of motor threshold) of stimulation.9 Based on their studies with healthy volunteers, they recommended several “safe” ranges (such as 5 seconds at 110% of motor threshold). Most trials use 30- to 60-second inter-train intervals.

        Most treatments continue 2 to 4 weeks, Monday through Friday, although more frequent treatments are being studied.

        EFFICACY FOR DEPRESSION

        Most studies of rTMS in depression have compared real rTMS to a sham control or electroconvulsive therapy (ECT).

        In earlier studies, the sham procedure typically involved tilting the coil away from the skull. This method has been questioned, however, because of evidence of neuronal depolarization.10

        More recent sham coils mimic the real coils’ sound and sensation, without magnetic stimulation.

        Despite these methodologic problems and some mixed results, depressed patients receiving rTMS show more favorable results than those receiving sham rTMS.11,12 Several meta-analyses have attempted to quantify rTMS’ efficacy for depression:

        • Holtzheimer et al concluded that rTMS was statistically superior to sham rTMS, but the clinical significance of these findings was modest in a population of mostly outpatients with less-severe depression.13
        • Burt et al found a statistically strong antidepressant effect, but its magnitude varied and few of the studies yielded a substantial clinical response or remission. The team also noted that rTMS’ long-term efficacy or adverse effects are unknown.14
        • Kozel et al concluded that left prefrontal rTMS rendered a significant antidepressant effect with measurable clinical improvement.15
        • Gershon et al16 supported an antidepressant effect for rTMS when compared with sham rTMS or ECT.

        Ongoing rTMS research includes subjects with many types of mild to severe psychiatric illnesses, including major depression, obsessive-compulsive disorder, and psychosis. Typically, patients referred for experimental approaches have not responded to or tolerated available treatments. Exclusion criteria used by most rTMS studies are listed in the Table.

        Table

        Medical conditions that preclude use of rTMS

        Serious medical conditions
         History of seizures
         Increased intracranial pressure
         Serious head trauma
        Myocardial infarction within the past 6 months
        Pregnancy or childbearing potential (unless reliable contraception is being used)
        Intracranial metallic implants
        Pacemakers or other implanted devices

        rTMS vs. ECT. Four randomized, controlled trials have compared rTMS with ECT for treating severely ill, often medication-resistant patients.17-20 Although their methodologies differed, all four studies concluded that rTMS and ECT offer similar efficacy, except that rTMS may be less effective for treating psychotic depression.

        One study found ECT more effective than rTMS for psychotic depression, although the patients who received ECT were also treated with antipsychotics and/or antidepressants.17 Our study,19 which did not use these agents, has not corroborated this observation. Preliminary data also indicate comparable relapse rates following acute ECT and rTMS when subjects are followed on maintenance medication.21

        ADVERSE EFFECTS

        The potential adverse effects of new treatments must always be considered. Thus far, rTMS appears to produce minimal, relatively benign complications, including:

        • mild discomfort at the stimulation site
        • localized muscle twitching during stimulation
        • mild post-treatment headaches—believed caused by muscle contractions—which usually respond to aspirin or acetaminophen
        • treatment stimulation-related seizures (rarely).8

        The rTMS device makes a loud clicking noise, and subjects wear protective ear plugs during treatment.

        Patient experience. The first rTMS session—during which the patient’s motor threshold is determined—can last up to 45 minutes. Subsequent sessions are usually 15 to 20 minutes. Patients are typically apprehensive before the first session but become more relaxed with experience and tolerate the treatments easily.

        During the procedure, many patients describe a tapping sensation on the forehead, and some experience slight muscle twitching around the eye or corner of the mouth. As the coil warms, the skin it touches sometimes flushes pink, although this does not seem to bother our patients. They can return to their daily routines immediately after a session.

        rTMS for major depression. In our experience, rTMS may help patients with major depression. For example, one patient diagnosed with a major depressive episode with psychotic features was referred to our study comparing rTMS with ECT.19 Her depression had lasted several months, with partial response to ECT treatments. She signed informed consent and was randomly assigned to receive rTMS treatment.

        At study admission, the patient’s Hamilton Depression Rating Scale (HDRS) score was 48, indicating moderate to severe depression. Following 10 rTMS sessions, her HDRS score had dropped to 2, with remission of symptoms. No follow-up results were documented.

        Cognitive effects. Whereas mood disorders are associated with medication-independent neuropsychological deficits, most studies have found no adverse cognitive effects with rTMS.22 Indeed, some of our rTMS patients have improved in certain cognitive tests, although this may be explained by test-retest effects or better attention and concentration associated with mood improvement.

         

         

        Figure Potential roles for rTMS in treating major depression


        RTEmagicC_af5a338892.jpg.jpg

        Solid lines represent current standards of practice. Dotted lines represent hypothetical roles for rTMS.

        Source: Adapted and reprinted with permission from Dowd et al. Is repetitive transcranial magnetic stimulation an alternative to ECTfor the treatment of depression? Contemp Psychiatry 2002;1:1-10.

        POTENTIAL ROLE FOR rTMS

        Today’s standard treatment of major depressive episodes begins with an antidepressant (plus an antipsychotic, if necessary) and proceeds to augmentation strategies if response is insufficient. rTMS may one day become an augmentation or monotherapy option for patients who do not respond sufficiently to standard treatments (Figure).

        ECT treatment may be initiated if a patient has had a prior good response to ECT, is intolerant to medication, or prefers ECT. In that case, rTMS may be used as an alternate initial treatment or with ECT. Thus, rTMS may be used:

        • to augment antidepressants
        • as an alternative to antidepressants or ECT
        • or sequentially with ECT.

        Before that can happen, however, optimal treatment parameters need to be clarified by larger, well-designed, controlled studies comparing rTMS to a valid sham treatment, antidepressants, and ECT.

        Related resources

        • International Society for Transcranial Stimulation. www.ists.unibe.ch/
        • Repetitive Transcranial Magnetic Stimulation Research Clinic at Yale-New Haven Psychiatric Hospital.

        Disclosure

        The authors report that they have no proprietary interest in the technology discussed in this article.

        References

        1. Barker A, Jalinous R, Freeston I. Non-invasive magnetic stimulation of human motor cortex. Lancet 1985;1:1106-7.

        2. Lisanby SH, Datto CJ, Szuba MP. ECT and rTMS: past, present, and future. Depress Anxiety 2000;12:115-17.

        3. Post A, Keck PE, Jr. Transcranial magnetic stimulation as a therapeutic tool in psychiatry: what do we know about the neurobiological mechanisms? J Psychiatr Res 2001;35:193-215.

        4. Holfich G, Kasper S, Hufnagel A, et al. Application of transcranial magnetic stimulation in treatment of drug resistant major depression—a report of two cases. Human Psychopharmacol 1993;8:361-5.

        5. George MS, Nahas Z, Speer AM, et al. Transcranial magnetic stimulation—a new method for investigating the neuroanatomy of depression. In: Ebert D, Ebmeier K (eds). New models for depression. New York: Karger, 1998;94-122.

        6. Pridmore A, Americo Fernandes Filho J, Nahas Z, et al. Motor threshold in transcranial magnetic stimulation: a comparison of a neurophysiological method and a visualization of movement method. J ECT 1998;14(1):25-7.

        7. Kozel FA, Nahas Z, deBrux C, et al. How coil-cortex distance relates to age, motor threshold, and antidepressant response to repetitive transcranial magnetic stimulation. J Neuropsychiatry Clin Neurosci 2000;13:376-84.

        8. Wassermann EM. Risk and safety of repetitive transcranial magnetic stimulation: report and suggested guidelines from the International Workshop on the Safety of Repetitive Transcranial Magnetic Stimulation, 1996. Electroencephalogr Clin Neurophysiol 1998;108:1-16.

        9. Chen R, Gerloff C, Classen J, et al. Safety of different inter-train intervals for repetitive transcranial magnetic stimulation and recommendations for safe ranges of stimulation parameters. Electroencephalogr Clin Neurophysiol 1997;105:415-21.

        10. Loo CK, Taylor JL, Gandevia SC, et al. Transcranial magnetic stimulation in controlled treatment studies: Are some “sham” forms active? Biol Psychiatry. 2000;47:325-31.

        11. George MS, Nahas Z, Molloy M, et al. A controlled trial of daily left prefrontal cortex TMS for treating depression. Biol Psychiatry 2000;48:962-70.

        12. Berman RM, Narasimhan M, Sanacora G, et al. A randomized clinical trial of repetitive transcranial magnetic stimulation in the treatment of major depression. Biol Psychiatry 2000;47:332-7.

        13. Holtzheimer PE, Russo J, Avery D. A meta-analysis of repetitive transcranial magnetic stimulation in the treatment of depression. Psychopharmacol Bull 2001;35:149-69.

        14. Burt T, Lisanby SH, Sackeim HA. Neuropsychiatric applications of transcranial magnetic stimulation: a meta-analysis. Int J Neuropsychopharmacol 2002;5:73-103.

        15. Kozel FE, George MS. Meta-analysis of left prefrontal repetitive transcranial magnetic stimulation (rTMS) to treat depression. J Psychiatr Pract 2002;8:270-5.

        16. Gershon AA, Dannon PN, Grunhaus L. Transcranial magnetic stimulation in the treatment of depression. Am JPsychiatry 2003;160(5):835-45.

        17. Grunhaus L, Dannon PN, Schreiber S, et al. Repetitive transcranial magnetic stimulation is as effective as electroconvulsive therapy in the treatment of nondelusional major depressive disorder: an open study. Biol Psychiatry 2000;47:314-24.

        18. Pridmore S, Bruno R, Turnier-Shea Y, et al. Comparison of unlimited numbers of rapid transcranial magnetic stimulation and ECT treatment sessions in major depression episodes. Int J Neuropsychopharmacol 2000;3:129-34.

        19. Janicak PG, Dowd SM, Martis B, et al. Repetitive transcranial magnetic stimulation versus electroconvulsive therapy for major depression: preliminary results of a randomized trial. Biol Psychiatry 2002;51:659-67

        20. Grunhaus L, Schreiber S, Dolberg OT, et al. A randomized controlled comparison of electroconvulsive therapy and repetitive transcranial magnetic stimulation in severe and resistant nonpsychotic major depression. Biol Psychiatry 2003;53:324-31.

        21. Dannon PH, Dolberg OT, Schreiber S, Grunhaus L. Three and six month outcome following courses of either ECT or rTMS in a population of severely depressed individuals—preliminary report. Biol Psychiatry 2002;15:687-90.

        22. Martis B, Alam D, Dowd SM, et al. Neurocognitive effects of repetitive transcranial magnetic stimulation in severe major depression. Clin Neurophysiology (in press).

        References

        1. Barker A, Jalinous R, Freeston I. Non-invasive magnetic stimulation of human motor cortex. Lancet 1985;1:1106-7.

        2. Lisanby SH, Datto CJ, Szuba MP. ECT and rTMS: past, present, and future. Depress Anxiety 2000;12:115-17.

        3. Post A, Keck PE, Jr. Transcranial magnetic stimulation as a therapeutic tool in psychiatry: what do we know about the neurobiological mechanisms? J Psychiatr Res 2001;35:193-215.

        4. Holfich G, Kasper S, Hufnagel A, et al. Application of transcranial magnetic stimulation in treatment of drug resistant major depression—a report of two cases. Human Psychopharmacol 1993;8:361-5.

        5. George MS, Nahas Z, Speer AM, et al. Transcranial magnetic stimulation—a new method for investigating the neuroanatomy of depression. In: Ebert D, Ebmeier K (eds). New models for depression. New York: Karger, 1998;94-122.

        6. Pridmore A, Americo Fernandes Filho J, Nahas Z, et al. Motor threshold in transcranial magnetic stimulation: a comparison of a neurophysiological method and a visualization of movement method. J ECT 1998;14(1):25-7.

        7. Kozel FA, Nahas Z, deBrux C, et al. How coil-cortex distance relates to age, motor threshold, and antidepressant response to repetitive transcranial magnetic stimulation. J Neuropsychiatry Clin Neurosci 2000;13:376-84.

        8. Wassermann EM. Risk and safety of repetitive transcranial magnetic stimulation: report and suggested guidelines from the International Workshop on the Safety of Repetitive Transcranial Magnetic Stimulation, 1996. Electroencephalogr Clin Neurophysiol 1998;108:1-16.

        9. Chen R, Gerloff C, Classen J, et al. Safety of different inter-train intervals for repetitive transcranial magnetic stimulation and recommendations for safe ranges of stimulation parameters. Electroencephalogr Clin Neurophysiol 1997;105:415-21.

        10. Loo CK, Taylor JL, Gandevia SC, et al. Transcranial magnetic stimulation in controlled treatment studies: Are some “sham” forms active? Biol Psychiatry. 2000;47:325-31.

        11. George MS, Nahas Z, Molloy M, et al. A controlled trial of daily left prefrontal cortex TMS for treating depression. Biol Psychiatry 2000;48:962-70.

        12. Berman RM, Narasimhan M, Sanacora G, et al. A randomized clinical trial of repetitive transcranial magnetic stimulation in the treatment of major depression. Biol Psychiatry 2000;47:332-7.

        13. Holtzheimer PE, Russo J, Avery D. A meta-analysis of repetitive transcranial magnetic stimulation in the treatment of depression. Psychopharmacol Bull 2001;35:149-69.

        14. Burt T, Lisanby SH, Sackeim HA. Neuropsychiatric applications of transcranial magnetic stimulation: a meta-analysis. Int J Neuropsychopharmacol 2002;5:73-103.

        15. Kozel FE, George MS. Meta-analysis of left prefrontal repetitive transcranial magnetic stimulation (rTMS) to treat depression. J Psychiatr Pract 2002;8:270-5.

        16. Gershon AA, Dannon PN, Grunhaus L. Transcranial magnetic stimulation in the treatment of depression. Am JPsychiatry 2003;160(5):835-45.

        17. Grunhaus L, Dannon PN, Schreiber S, et al. Repetitive transcranial magnetic stimulation is as effective as electroconvulsive therapy in the treatment of nondelusional major depressive disorder: an open study. Biol Psychiatry 2000;47:314-24.

        18. Pridmore S, Bruno R, Turnier-Shea Y, et al. Comparison of unlimited numbers of rapid transcranial magnetic stimulation and ECT treatment sessions in major depression episodes. Int J Neuropsychopharmacol 2000;3:129-34.

        19. Janicak PG, Dowd SM, Martis B, et al. Repetitive transcranial magnetic stimulation versus electroconvulsive therapy for major depression: preliminary results of a randomized trial. Biol Psychiatry 2002;51:659-67

        20. Grunhaus L, Schreiber S, Dolberg OT, et al. A randomized controlled comparison of electroconvulsive therapy and repetitive transcranial magnetic stimulation in severe and resistant nonpsychotic major depression. Biol Psychiatry 2003;53:324-31.

        21. Dannon PH, Dolberg OT, Schreiber S, Grunhaus L. Three and six month outcome following courses of either ECT or rTMS in a population of severely depressed individuals—preliminary report. Biol Psychiatry 2002;15:687-90.

        22. Martis B, Alam D, Dowd SM, et al. Neurocognitive effects of repetitive transcranial magnetic stimulation in severe major depression. Clin Neurophysiology (in press).

        Issue
        Current Psychiatry - 02(06)
        Issue
        Current Psychiatry - 02(06)
        Page Number
        59-66
        Page Number
        59-66
        Publications
        Publications
        Topics
        Article Type
        Display Headline
        Therapy-resistant major depression The attraction of magnetism: How effective—and safe—is rTMS?
        Display Headline
        Therapy-resistant major depression The attraction of magnetism: How effective—and safe—is rTMS?
        Sections
        Article Source

        PURLs Copyright

        Inside the Article

        Antipsychotics and mood disorders: A complicated alliance

        Article Type
        Changed
        Mon, 03/04/2019 - 15:49
        Display Headline
        Antipsychotics and mood disorders: A complicated alliance

        Major mood disorders are challenging to diagnose and often difficult to treat. They entail unipolar depression; bipolar disorder, which includes manic, depressed, or mixed episodes; and schizoaffective disorder, which includes both depressed and bipolar subtypes. Antidepressants and mood stabilizers are the primary pharmacological treatments. They may be insufficient, however, for patients with more severe episodes, often characterized by psychosis and treatment resistance.

        In these patients, antipsychotics have played an important but controversial part in management, primarily as oral or parenteral adjuncts. Literature and clinical experience now support another, unique role for the current generation of novel agents.

        Compared to earlier antipsychotics, these agents produce substantially fewer neurological adverse effects, including acute extrapyramidal and tardive syndromes, and can augment antidepressants and mood stabilizers. In addition, they may:

         

        • Possess a better antipsychotic profile, with enhanced therapeutic effects on positive, negative, cognitive, and mood symptoms
        • Have a role in the acute and long-term management of these disorders when anticipated parenteral formulations become available (e.g., acute intramuscular olanzapine and ziprasidone—and long-acting intramuscular risperidone)
        • Possess inherent thymoleptic properties (see “Unresolved issues with antipsychotics,” below).

         

        UNRESOLVED ISSUES WITH ANTIPSYCHOTICS

         

        1. Defining what constitutes a mood stabilizer.1 Proposed definitions suggest that the drug must entail the following:
        2. Clarifying the mechanisms underlying the apparent mood-regulating effects of novel agents
        3. Ascertaining both acute and maintenance efficacy
        4. Clarifying the propensity of some agents to switch depressed patients into mania
        5. Increasing the number of well-designed studies with sufficient sample sizes, including comparison trials assessing the relative efficacy of different novel agents
        6. Reducing the tendency to publish only positive reports when new drugs are first available
        7. Introducing parenteral formulations of novel agents
        8. Resolving concerns about weight gain, new-onset diabetes, QT c prolongation, and sedation
        9. Rectifying the current level of substantially greater costs

        Management of unipolar depression

        Neuroleptics Delusions and hallucinations indicate a more severe form of depressive disorder, with poor short- and long-term outcomes in comparison to those without psychosis. To illustrate, Table 1 lists a summary of response rates in psychotic and nonpsychotic depressed patients given a tricyclic antidepressant (TCA). The data indicate that patients suffering from psychotic depression typically do not benefit from antidepressant monotherapy and usually require a combination of antidepressant and antipsychotic or, alternatively, electroconvulsive therapy (ECT).

        There is, however, some limited clinical and neuroimaging evidence that amoxapine can be used as an effective monotherapy in this group. Amoxapine is an antidepressant whose primary active metabolite, 8-hydroxy amoxapine, may have antipsychotic properties.1 With the possible exception of amoxapine, combined antipsychotic-antidepressant treatment is the rule.

        Table 1

        Psychotic and nonpsychotic depressed patients’ response to monotherapy with a tricyclic antidepressant

         

         PsychoticNonpsychotic 
         Responders (%) (n=127)Nonresponders (%) (n=236)Responders (%) (n=464)Nonresponders (%) (n=227)Difference
        13 studies35%65%67%33%32%
        Adapted from Chan CH, Janicak PG, Davis JM, et al. Response of psychotic and nonpsychotic depressed patients to tricyclic antidepressants. J Clin Psychiatry. 1987;48:197-200.

        Historically, studies have also evaluated neuroleptic monotherapy for depressed patients. While some reported superiority over a placebo, none found conventional antipsychotics superior to imipramine. Indeed, patients with schizophrenia who are treated with a neuroleptic often develop symptoms that are difficult to distinguish from depression (e.g., secondary negative symptoms). These often improve when the neuroleptic is discontinued or the patients are switched to a novel antipsychotic such as risperidone, olanzapine, or ziprasidone, all of which have putative antidepressant effects.

        When employing an antipsychotic in depressed patients, the dosage and duration of treatment are two critical considerations. To minimize neuromotor adverse effects, use low doses of a neuroleptic (e.g., haloperidol, 1 to 5 mg/d) in conjunction with the primary antidepressant therapy. The neuroleptic should then be tapered gradually after psychotic symptoms have been controlled, usually during the acute phase of treatment. Ideally, patients would then take antidepressant monotherapy through the continuation phase and, if necessary, the maintenance phase of treatment. If psychosis recurs, re-introduce the antipsychotic intermittently.

        Novel antipsychotics In contrast to neuroleptics, novel antipsychotics have been reported to improve depression in various psychotic and mood disorders.

        For example, ziprasidone has serotonin and noradrenergic reuptake blocking effects comparable to such classic TCAs as imipramine and amitriptyline, as well as high binding affinity at the 5-HT1A, 5-HT1D, and 5-HT2C receptors. This neuroreceptor profile indicates possible antidepressant effects.

        While randomized, controlled trials with mood-disordered patients are few, there have been promising preliminary reports of augmentation of antidepressants with risperidone and olanzapine in both psychotic and nonpsychotic depressed patients.

        Ostroff and Nelson2 reported the results of an open-label study of eight SSRI-nonresponsive patients (mean treatment 7.3 weeks). These patients had no psychotic features and had a dramatic reduction in depressive symptoms, as well as some improvement in sexual dysfunction, with the addition of 0.5 mg to 1.0 mg risperidone. The clinicians suggested that risperidone’s 5-HT2A antagonism might explain its augmentation of the partial SSRI response.

         

         

        Olanzapine alone (n=3) or combined with an antidepressant (n=12) has also been reported to improve both depression and psychosis.3 In a double-blind, amitriptyline-controlled trial, Svestka and Synek4 found that olanzapine demonstrated antidepressant efficacy in 33 unipolar and seven bipolar depressed patients. Thirteen of these patients also had psychotic symptoms.

        Shelton et al5 reported the results of a two-center, 8-week, double-blind comparison of olanzapine alone, fluoxetine alone, or their combination in 28 patients suffering from treatment-resistant, non-bipolar disorder without psychosis. They found that the combination was superior to either drug alone based on improvement in the Hamilton Depression Rating Scale (HDRS) total score. From their preliminary data, it also appears that the doses required were relatively low, reducing the risk of side effects.

        Their findings, however, need to be replicated in more controlled studies with combinations, addressing possible adverse effects, the potential for clinically relevant drug interactions, decreased compliance rates, and increased cost of treatment. Earlier reports raised concern about the potential of these agents to increase switching to hypomania or mania. But in more recent reports, this has not emerged as a significant problem.7

        Finally, several case reports and case series indicate that agents such as clozapine and risperidone may augment ECT in particularly severe, treatment-resistant depressive episodes.7

        Management of bipolar and schizoaffective depressed episodes

        Neuroleptics Antipsychotics are frequently used to manage more severe, usually psychotic episodes of bipolar and schizoaffective depression. Reports indicate that affectively ill patients receiving neuroleptics may be more prone to develop neuromotor adverse effects than are those suffering from schizophrenia. Thus, their use for such patients must be well justified, limited in dosage and duration, and carefully monitored for the emergence of acute and tardive neurological events.

        Novel antipsychotics Novel antipsychotics have demonstrated fewer propensities than have neuroleptics in worsening depression or negative symptoms in schizophrenic patients, and have possible antidepressant effects. In support of this hypothesis, and reminiscent of data from earlier risperidone and olanzapine trials, ziprasidone was observed to improve the Montgomery Asberg Rating Scale (MADRS) and Brief Psychotic Rating Scale (BPRS) depressive cluster scores in three clinical trials with schizophrenic and schizoaffective patients.8,9

        Vieta et al reported the efficacy and safety of risperidone add-on therapy for treating various episodes of bipolar (n=358) and schizoaffective (n=183) disorders.6 In this multicenter, open study, 33 patients (6.1%) suffered a depressed episode and received a mean risperidone dose of 1.6 (± 2.3) mg/d added to their ongoing but ineffective drug regimen. Mean HDRS declined significantly over the 6-month course. Further, switch rates were low and in the expected range for spontaneous fluctuations seen in these disorders.

        The results of a 6-week, double-blind, controlled trial of risperidone versus haloperidol in 62 patients with schizoaffective disorder, bipolar or depressed subtype, were published.10 Risperidone (average dose of 5.5 mg/d) was comparable to haloperidol (average dose of 10.8 mg/d) in reducing the mean in the Positive and Negative Syndrome Scale and Clinician-Administered Rating Scale for Mania change scores.

        In those patients with baseline HDRS scores ≥ 20, risperidone produced a significantly greater reduction in mean change scores than did haloperidol. In addition, patients had no mood switches with risperidone or haloperidol; there was a significantly higher incidence of patients who had extra-pyramidal symptoms with haloperidol than among those taking risperidone; and six patients in the group taking haloperidol dropped out after experiencing adverse effects. None of the patients taking risperidone dropped out.

        Table 2

        Lithium versus antipsychotics for acute mania

         

         LithiumAntipsychotics 
         Responders (%) (n=64)Nonresponders (%) (n=10)Responders (%) (n=38)Nonresponders (%) (n=33)Difference
        5 studies89%11%54%46%35%
        Adapted from Janicak PG, Newman RH, Davis JM. Advances in the treatment of mania and related disorders: a reappraisal. Psychiatric Ann. 1992;22(2):94.

        Management of bipolar manic or mixed episodes

        Up to 80% of all bipolar patients receive an antipsychotic drug during the acute and/or maintenance phase of their illness, even though loading doses of valproate and benzodiazepines may also be used during an exacerbation and pose much less risk, especially in terms of adverse neurological effects.

        Neuroleptics Shortly after their introduction, neuroleptics were found to reduce mortality secondary to dehydration and exhaustion in many highly agitated patients during an acute manic episode such as lethal catatonia.7

        While earlier controlled studies found these agents to be effective in the treatment of acute mania, they are clearly less efficacious than lithium for core manic symptoms.11Table 2 demonstrates a meta-analysis of five well-controlled, double-blind studies documenting the statistical superiority of lithium over neuroleptics. These agents, however, offer the advantage of a more rapid onset of action, particularly when given in the acute parenteral formulation, and are superior to lithium in the initial control of agitation. Further, long-acting depot formulations of neuroleptics may be the only viable strategy for chronic, recurrent, noncompliant patients.

         

         

        As with psychotic depression, dosing and duration of neuroleptic treatment are important concerns. In this context, Rifkin et al demonstrated that 10 mg of haloperidol per day had comparable efficacy but fewer adverse effects than did 30 or 80 mg per day in a group of acutely manic patients.12 Despite such data, high chlorpromazine-equivalent doses are often administered acutely and maintained for sustained periods. This can be a significant problem given the apparent great sensitivity of bipolar patients to the neurological sequelae of these antipsychotic agents.

        Novel antipsychotics Early case series reports indicated that clozapine may benefit treatment-refractory bipolar patients. Given the inherent drawbacks of clozapine (e.g., agranulocytosis and seizure induction), attention now focuses on other novel agents with more benign adverse effect profiles than clopazine. Controlled trials with olanzapine and risperidone serve to reinforce the usefulness of these as well as other novel agents.

        Tohen et al published the results of a 3-week, double-blind, placebo-controlled trial of olanzapine in 139 patients experiencing an acute bipolar manic or mixed episode.13 Olanzapine produced a statistically greater mean improvement than did the placebo on the Young Mania Rating Scale (YMRS) change scores. Further, 49% of the olanzapine-treated group (n=70) met the a priori criteria for response versus only 24% of the placebo-treated group (n=69). A second study using a higher starting dose of olanzapine, less rescue medication, and longer treatment duration than the first study resulted in a similar outcome.14

        Sachs et al reported on the results of a 3-week, double-blind, placebo-controlled trial involving 156 patients with bipolar manic or mixed subtype who received a mood stabilizer (lithium or valproate) plus a placebo, risperidone (1 to 6 mg/d), or haloperidol (2 to 12 mg/d).15 The clinicians concluded that risperidone plus a mood stabilizer was statistically superior to a placebo plus a mood stabilizer, and produced more rapid reduction in manic symptoms, regardless of whether psychosis was present.

        Sajatovic et al16 published the results of a prospective, open trial with quetiapine (mean dose = 203 ± 124 mg/d) as add-on therapy in 20 patients (10 bipolar, 10 schizoaffective; 19 male, 1 female) insufficiently responsive to their mood stabilizer or antipsychotic. Pre-post assessments indicated significant improvement in the BPRS, Mania Rating Scale (MRS), and HDRS scores. While the combination was generally well tolerated, there was a mean weight gain of 4.9 kg (10.8 lb). This raises the specter of complications associated with substantial weight gain produced by several of the novel antipsychotics.

        A recent report indicates that ziprasidone may also be an effective antimanic agent. In a randomized, double-blind, placebo-controlled, multicenter trial involving 210 bipolar (manic or mixed episodes) patients, ziprasidone (80 to 160 mg/d; n=140) was compared to a placebo (n=70) for 3 weeks.17 By day 2 and all subsequent time points, ziprasidone was superior in terms of mean change scores from the baseline MRS; produced a more rapid and significantly greater improvement in overall psychopathology in both positive and negative symptoms; and did not produce significant adverse effects (including relevant ECG parameter changes) when compared with the placebo. Similar trials are being conducted for risperidone, aripiprazole, and iloperidone.

        Finally, Meehan et al18 reported on the results of an acute parenteral formulation of olanzapine used to manage agitation in an acute manic or mixed episode. This was a 24-hour, double-blind, placebo-controlled trial comparing intramuscular olanzapine to intramuscular lorazepam. The following results were indicated:

         

        • Olanzapine (doses of 5 to 10 mg) produced a significantly greater reduction in excitation than did the placebo or lorazepam at 30 minutes post-injection.
        • Twice as many patients receiving lorazepam or a placebo versus olanzapine required more than one injection.
        • Except for olanzapine-induced tachycardia in one patient, there were no significant changes in vital signs, ECG parameters, or laboratory assays among the three groups.
        • Somnolence (13%) and dizziness (9%) were the most frequent side effects in the olanzapine group.

        Treatment strategies for depression and mania

        Considering the existing research data, clinical experience, and the risk/benefit ratio, treatment strategies that emphasize the role of antipsychotics in managing severe mood disorders are presented in the algorithms in Figures 1 and 2.

        Figure 1 emphasizes the role of antipsychotics in the pharmacological management of patients with major depression. For unipolar depression with psychotic symptoms, options include an antidepressant plus an antipsychotic; amoxapine monotherapy; and possibly monotherapy with a novel agent such as ziprasidone. For bipolar depression with psychosis or schizoaffective disorder with depression, combining a mood stabilizer such as lithium plus an antipsychotic may be sufficient, but often an antidepressant must also be added. If the response is insufficient, consider switching to a novel antipsychotic (e.g., olanzapine or risperidone) plus a mood stabilizer (± antidepressant). In more serious exacerbations (e.g., high suicidality), ECT may be most appropriate. Secondary choices include clozapine with or without an antidepressant or novel antipsychotic such as risperidone combined with ECT.

         

         

        Figure 2 describes the use of antipsychotics in patients with mania. If response to a primary mood stabilizer such as lithium, valproate, or their combination in the context of a bipolar or schizoaffective disorder is insufficient—or if patients have severe manic or psychotic symptoms—an antipsychotic may be added to the primary mood stabilizer.

        Alternatively, when mood stabilizers are not tolerated or a clinical situation such as pregnancy precludes their use, a novel agent such as olanzapine or risperidone may be given as monotherapy. While the safety of these agents in pregnancy is not clearly established, clinical experience thus far indicates they may be safer than agents such as valproate or carbamazepine. These agents would be the first choice given their diminished propensity for extrapyramidal symptoms; absence of clozapine-related adverse effects such as agranulocytosis and seizures; and growing evidence of possible mood stabilizing effects.

         

        Figure 1 ANTIPSYCHOTICS IN THE TREATMENT OF MAJOR DEPRESSION


        RTEmagicC_9ae32bde6f.jpg.jpg

        Figure 2 ANTIPSYCHOTICS IN THE TREATMENT OF MANIA


        RTEmagicC_860ac76e3c.jpg.jpg

        For patients who remain nonresponsive, clozapine should be considered either as monotherapy or combined with valproate and/or lithium. Combining this agent with carbamazepine is not recommended because of the possibility of an increased risk of hematotoxicity.

        Electroconvulsive therapy may be used safely and effectively in patients who are severely ill (e.g., those with manic delirium); pose an immediate danger because of their potential for violence; are in medical crisis; or have medical contraindications to pharmacotherapy. There is preliminary evidence that ECT can be safely administered with novel antipsychotics such as clozapine, risperidone, or olanzapine to produce additional benefit in patients insufficiently responsive to either therapy alone.

        Related resources

         

        Drug brand names

         

        • Amitriptyline • Elavil
        • Amoxapine • Asendin
        • Aripiprazole • (in development)
        • Carbamazepine • Tegretol, Epitol
        • Clozapine • Clozaril
        • Haloperidol • Haldol
        • Iloperidone • (in development)
        • Imipramine • Tofranil
        • Lorazepam • Ativan
        • Olanzapine • Zyprexa
        • Quetiapine • Seroquel
        • Risperidone • Risperdal
        • Valproate sodium • Depacon
        • Ziprasidone • Geodon

        Disclosure

        The author reports that he receives research/grant support from, serves as a consultant for, and on the speaker’s bureau of Janssen Pharmaceutica. He also receives research/grant support from Genentech Inc. and Bristol-Myers Squibb Co.; serves as a consultant for Pfizer Inc., Sepracor, and Novartis Pharmaceuticals Corp.; and is on the speaker’s bureau of Abbott Laboratories, Eli Lilly and Co., Pfizer Inc., Forest Pharmaceuticals, Bristol-Myers Squibb Co., and Wyeth-Ayerst Pharmaceuticals.

        References

         

        1. Kapur S, Cho R, Jones C, et al. Is amoxapine an atypical antipsychotic? Positronemission tomography investigation of its dopamine2 and serotonin2 occupancy. Biol Psychiatry. 1999;45:1217-1220.

        2. Ostroff RB, Nelson JC. Risperidone augmentation of selective serotonin reuptake inhibitors in major depression. J Clin Psychiatry. 1999;60:256-259.

        3. Rothschild AJ, Bates KS, Boehringer KL, Syed A. Olanzapine response in psychotic depression. J Clin Psychiatry. 1999;60:116-118.

        4. Svestka J, Synek O. Does olanzapine have antidepressant effect? A double-blind amitriptyline-controlled study [abstract]. Int J Neuropsychopharmacol. 2000;3(suppl 1):S251.-

        5. Shelton RC, Tollefson GD, Tohen M, et al. A novel augmentation strategy for treating resistant major depression. Am J Psychiatry. 2001;158:131-134.

        6. Vieta E, Goikolea JM, Corbella B, et al. Risperidone safety and efficacy in the treatment of bipolar and schizoaffective disorders: results from a 5-month, multicenter, open study. J Clin Psychiatry. 2001;62(10):818.-

        7. Janicak PG, Davis JM, et al. Principles and Practice of Psychopharmacotherapy. 3rd ed. Philadelphia, Pa: Lippincott-Williams & Wilkins; 2001.

        8. Daniel DG, Zimbroff DL, et al. for the Ziprasidone Study Group Ziprasidone 80 mg/day and 160 mg/day in the acute exacerbation of schizophrenia and schizoaffective disorder: a 6-week placebo-controlled trial. Neuropsychopharmacol. 1999;20(5):491-505.

        9. Keck PE, Jr, Buffenstein A, Ferguson J, et al. Ziprasidone 40 and 120 mg/day in the acute exacerbation of schizophrenia and schizoaffective disorder: a 4-week placebo-controlled trial. Psychopharmacol. 1998;140:173-184.

        10. Janicak PG, Keck PE, Jr, Davis JM, et al. A double-blind, randomized, prospective evaluation of the efficacy and safety of risperidone versus haloperidol in the treatment of schizoaffective disorder. J Clin Psychopharmacol. 2001;21:360-368.

        11. Keck PE, Welge JA, McElroy SL, et al. Placebo effect in randomized, controlled studies of acute bipolar mania and depression. Biol Psychiatry. 2000;47(8):756-761.

        12. Rifkin A, Doddi S, Karajgi B, et al. Dosage of haloperidol for mania. Br J Psychiatry. 1994;165:113-116.

        13. Tohen M, Sanger TM, McElroy SL, et al. Olanzipine versus placebo in the treatment of acute mania. Olanzapine HGEH Study Group. Am J Psychiatry. 1999;156:702-709.

        14. Tohen M, Jacobs TG, Grundy SL, et al. Efficacy of olanzapine in acute bipolar mania: a double-blind, placebo-controlled study. The Olanzipine HGGW Study Group. Arch Gen Psychiatry. 2000;57:841-849.

        15. Sachs G, Ghaemi N, Grossman F, Bowden C. Risperidone plus mood stabilizer vs. placebo plus mood stabilizer for acute mania of bipolar disorder: a double-blind comparison of efficacy and safety. International Congress on Bipolar Disorders. Pittsburgh, Pa. June 14-16, 2001.

        16. Sajatovic M, Briscan DW, Perez DE, et al. Quetiapine alone and added to a mood stabilizer for serious mood disorders. J Clin Psychiatry. 2001;62:728-732.

        17. Giller E, Mandel FS, Keck P. Ziprasidone in the acute treatment of mania: a double-blind, placebo-controlled, randomized trial. Schizophr Res. 2001;49(suppl 1-2):229.-

        18. Meehan K, Zhang F, David S, Tohen N, Janicak PG, et al. A double-blind, randomized comparison of the efficacy and safety of intramuscular (IM) olanzapine versus IM lorazepam and IM placebo in acutely agitated patients diagnosed with mania associated with bipolar disorder. J Clin Psychopharmacol 2001;21:389-397.

        Author and Disclosure Information

         

        Philip G. Janicak, MD
        Professor of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, III

        Issue
        Current Psychiatry - 01(01)
        Publications
        Topics
        Page Number
        11-17
        Sections
        Author and Disclosure Information

         

        Philip G. Janicak, MD
        Professor of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, III

        Author and Disclosure Information

         

        Philip G. Janicak, MD
        Professor of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, III

        Major mood disorders are challenging to diagnose and often difficult to treat. They entail unipolar depression; bipolar disorder, which includes manic, depressed, or mixed episodes; and schizoaffective disorder, which includes both depressed and bipolar subtypes. Antidepressants and mood stabilizers are the primary pharmacological treatments. They may be insufficient, however, for patients with more severe episodes, often characterized by psychosis and treatment resistance.

        In these patients, antipsychotics have played an important but controversial part in management, primarily as oral or parenteral adjuncts. Literature and clinical experience now support another, unique role for the current generation of novel agents.

        Compared to earlier antipsychotics, these agents produce substantially fewer neurological adverse effects, including acute extrapyramidal and tardive syndromes, and can augment antidepressants and mood stabilizers. In addition, they may:

         

        • Possess a better antipsychotic profile, with enhanced therapeutic effects on positive, negative, cognitive, and mood symptoms
        • Have a role in the acute and long-term management of these disorders when anticipated parenteral formulations become available (e.g., acute intramuscular olanzapine and ziprasidone—and long-acting intramuscular risperidone)
        • Possess inherent thymoleptic properties (see “Unresolved issues with antipsychotics,” below).

         

        UNRESOLVED ISSUES WITH ANTIPSYCHOTICS

         

        1. Defining what constitutes a mood stabilizer.1 Proposed definitions suggest that the drug must entail the following:
        2. Clarifying the mechanisms underlying the apparent mood-regulating effects of novel agents
        3. Ascertaining both acute and maintenance efficacy
        4. Clarifying the propensity of some agents to switch depressed patients into mania
        5. Increasing the number of well-designed studies with sufficient sample sizes, including comparison trials assessing the relative efficacy of different novel agents
        6. Reducing the tendency to publish only positive reports when new drugs are first available
        7. Introducing parenteral formulations of novel agents
        8. Resolving concerns about weight gain, new-onset diabetes, QT c prolongation, and sedation
        9. Rectifying the current level of substantially greater costs

        Management of unipolar depression

        Neuroleptics Delusions and hallucinations indicate a more severe form of depressive disorder, with poor short- and long-term outcomes in comparison to those without psychosis. To illustrate, Table 1 lists a summary of response rates in psychotic and nonpsychotic depressed patients given a tricyclic antidepressant (TCA). The data indicate that patients suffering from psychotic depression typically do not benefit from antidepressant monotherapy and usually require a combination of antidepressant and antipsychotic or, alternatively, electroconvulsive therapy (ECT).

        There is, however, some limited clinical and neuroimaging evidence that amoxapine can be used as an effective monotherapy in this group. Amoxapine is an antidepressant whose primary active metabolite, 8-hydroxy amoxapine, may have antipsychotic properties.1 With the possible exception of amoxapine, combined antipsychotic-antidepressant treatment is the rule.

        Table 1

        Psychotic and nonpsychotic depressed patients’ response to monotherapy with a tricyclic antidepressant

         

         PsychoticNonpsychotic 
         Responders (%) (n=127)Nonresponders (%) (n=236)Responders (%) (n=464)Nonresponders (%) (n=227)Difference
        13 studies35%65%67%33%32%
        Adapted from Chan CH, Janicak PG, Davis JM, et al. Response of psychotic and nonpsychotic depressed patients to tricyclic antidepressants. J Clin Psychiatry. 1987;48:197-200.

        Historically, studies have also evaluated neuroleptic monotherapy for depressed patients. While some reported superiority over a placebo, none found conventional antipsychotics superior to imipramine. Indeed, patients with schizophrenia who are treated with a neuroleptic often develop symptoms that are difficult to distinguish from depression (e.g., secondary negative symptoms). These often improve when the neuroleptic is discontinued or the patients are switched to a novel antipsychotic such as risperidone, olanzapine, or ziprasidone, all of which have putative antidepressant effects.

        When employing an antipsychotic in depressed patients, the dosage and duration of treatment are two critical considerations. To minimize neuromotor adverse effects, use low doses of a neuroleptic (e.g., haloperidol, 1 to 5 mg/d) in conjunction with the primary antidepressant therapy. The neuroleptic should then be tapered gradually after psychotic symptoms have been controlled, usually during the acute phase of treatment. Ideally, patients would then take antidepressant monotherapy through the continuation phase and, if necessary, the maintenance phase of treatment. If psychosis recurs, re-introduce the antipsychotic intermittently.

        Novel antipsychotics In contrast to neuroleptics, novel antipsychotics have been reported to improve depression in various psychotic and mood disorders.

        For example, ziprasidone has serotonin and noradrenergic reuptake blocking effects comparable to such classic TCAs as imipramine and amitriptyline, as well as high binding affinity at the 5-HT1A, 5-HT1D, and 5-HT2C receptors. This neuroreceptor profile indicates possible antidepressant effects.

        While randomized, controlled trials with mood-disordered patients are few, there have been promising preliminary reports of augmentation of antidepressants with risperidone and olanzapine in both psychotic and nonpsychotic depressed patients.

        Ostroff and Nelson2 reported the results of an open-label study of eight SSRI-nonresponsive patients (mean treatment 7.3 weeks). These patients had no psychotic features and had a dramatic reduction in depressive symptoms, as well as some improvement in sexual dysfunction, with the addition of 0.5 mg to 1.0 mg risperidone. The clinicians suggested that risperidone’s 5-HT2A antagonism might explain its augmentation of the partial SSRI response.

         

         

        Olanzapine alone (n=3) or combined with an antidepressant (n=12) has also been reported to improve both depression and psychosis.3 In a double-blind, amitriptyline-controlled trial, Svestka and Synek4 found that olanzapine demonstrated antidepressant efficacy in 33 unipolar and seven bipolar depressed patients. Thirteen of these patients also had psychotic symptoms.

        Shelton et al5 reported the results of a two-center, 8-week, double-blind comparison of olanzapine alone, fluoxetine alone, or their combination in 28 patients suffering from treatment-resistant, non-bipolar disorder without psychosis. They found that the combination was superior to either drug alone based on improvement in the Hamilton Depression Rating Scale (HDRS) total score. From their preliminary data, it also appears that the doses required were relatively low, reducing the risk of side effects.

        Their findings, however, need to be replicated in more controlled studies with combinations, addressing possible adverse effects, the potential for clinically relevant drug interactions, decreased compliance rates, and increased cost of treatment. Earlier reports raised concern about the potential of these agents to increase switching to hypomania or mania. But in more recent reports, this has not emerged as a significant problem.7

        Finally, several case reports and case series indicate that agents such as clozapine and risperidone may augment ECT in particularly severe, treatment-resistant depressive episodes.7

        Management of bipolar and schizoaffective depressed episodes

        Neuroleptics Antipsychotics are frequently used to manage more severe, usually psychotic episodes of bipolar and schizoaffective depression. Reports indicate that affectively ill patients receiving neuroleptics may be more prone to develop neuromotor adverse effects than are those suffering from schizophrenia. Thus, their use for such patients must be well justified, limited in dosage and duration, and carefully monitored for the emergence of acute and tardive neurological events.

        Novel antipsychotics Novel antipsychotics have demonstrated fewer propensities than have neuroleptics in worsening depression or negative symptoms in schizophrenic patients, and have possible antidepressant effects. In support of this hypothesis, and reminiscent of data from earlier risperidone and olanzapine trials, ziprasidone was observed to improve the Montgomery Asberg Rating Scale (MADRS) and Brief Psychotic Rating Scale (BPRS) depressive cluster scores in three clinical trials with schizophrenic and schizoaffective patients.8,9

        Vieta et al reported the efficacy and safety of risperidone add-on therapy for treating various episodes of bipolar (n=358) and schizoaffective (n=183) disorders.6 In this multicenter, open study, 33 patients (6.1%) suffered a depressed episode and received a mean risperidone dose of 1.6 (± 2.3) mg/d added to their ongoing but ineffective drug regimen. Mean HDRS declined significantly over the 6-month course. Further, switch rates were low and in the expected range for spontaneous fluctuations seen in these disorders.

        The results of a 6-week, double-blind, controlled trial of risperidone versus haloperidol in 62 patients with schizoaffective disorder, bipolar or depressed subtype, were published.10 Risperidone (average dose of 5.5 mg/d) was comparable to haloperidol (average dose of 10.8 mg/d) in reducing the mean in the Positive and Negative Syndrome Scale and Clinician-Administered Rating Scale for Mania change scores.

        In those patients with baseline HDRS scores ≥ 20, risperidone produced a significantly greater reduction in mean change scores than did haloperidol. In addition, patients had no mood switches with risperidone or haloperidol; there was a significantly higher incidence of patients who had extra-pyramidal symptoms with haloperidol than among those taking risperidone; and six patients in the group taking haloperidol dropped out after experiencing adverse effects. None of the patients taking risperidone dropped out.

        Table 2

        Lithium versus antipsychotics for acute mania

         

         LithiumAntipsychotics 
         Responders (%) (n=64)Nonresponders (%) (n=10)Responders (%) (n=38)Nonresponders (%) (n=33)Difference
        5 studies89%11%54%46%35%
        Adapted from Janicak PG, Newman RH, Davis JM. Advances in the treatment of mania and related disorders: a reappraisal. Psychiatric Ann. 1992;22(2):94.

        Management of bipolar manic or mixed episodes

        Up to 80% of all bipolar patients receive an antipsychotic drug during the acute and/or maintenance phase of their illness, even though loading doses of valproate and benzodiazepines may also be used during an exacerbation and pose much less risk, especially in terms of adverse neurological effects.

        Neuroleptics Shortly after their introduction, neuroleptics were found to reduce mortality secondary to dehydration and exhaustion in many highly agitated patients during an acute manic episode such as lethal catatonia.7

        While earlier controlled studies found these agents to be effective in the treatment of acute mania, they are clearly less efficacious than lithium for core manic symptoms.11Table 2 demonstrates a meta-analysis of five well-controlled, double-blind studies documenting the statistical superiority of lithium over neuroleptics. These agents, however, offer the advantage of a more rapid onset of action, particularly when given in the acute parenteral formulation, and are superior to lithium in the initial control of agitation. Further, long-acting depot formulations of neuroleptics may be the only viable strategy for chronic, recurrent, noncompliant patients.

         

         

        As with psychotic depression, dosing and duration of neuroleptic treatment are important concerns. In this context, Rifkin et al demonstrated that 10 mg of haloperidol per day had comparable efficacy but fewer adverse effects than did 30 or 80 mg per day in a group of acutely manic patients.12 Despite such data, high chlorpromazine-equivalent doses are often administered acutely and maintained for sustained periods. This can be a significant problem given the apparent great sensitivity of bipolar patients to the neurological sequelae of these antipsychotic agents.

        Novel antipsychotics Early case series reports indicated that clozapine may benefit treatment-refractory bipolar patients. Given the inherent drawbacks of clozapine (e.g., agranulocytosis and seizure induction), attention now focuses on other novel agents with more benign adverse effect profiles than clopazine. Controlled trials with olanzapine and risperidone serve to reinforce the usefulness of these as well as other novel agents.

        Tohen et al published the results of a 3-week, double-blind, placebo-controlled trial of olanzapine in 139 patients experiencing an acute bipolar manic or mixed episode.13 Olanzapine produced a statistically greater mean improvement than did the placebo on the Young Mania Rating Scale (YMRS) change scores. Further, 49% of the olanzapine-treated group (n=70) met the a priori criteria for response versus only 24% of the placebo-treated group (n=69). A second study using a higher starting dose of olanzapine, less rescue medication, and longer treatment duration than the first study resulted in a similar outcome.14

        Sachs et al reported on the results of a 3-week, double-blind, placebo-controlled trial involving 156 patients with bipolar manic or mixed subtype who received a mood stabilizer (lithium or valproate) plus a placebo, risperidone (1 to 6 mg/d), or haloperidol (2 to 12 mg/d).15 The clinicians concluded that risperidone plus a mood stabilizer was statistically superior to a placebo plus a mood stabilizer, and produced more rapid reduction in manic symptoms, regardless of whether psychosis was present.

        Sajatovic et al16 published the results of a prospective, open trial with quetiapine (mean dose = 203 ± 124 mg/d) as add-on therapy in 20 patients (10 bipolar, 10 schizoaffective; 19 male, 1 female) insufficiently responsive to their mood stabilizer or antipsychotic. Pre-post assessments indicated significant improvement in the BPRS, Mania Rating Scale (MRS), and HDRS scores. While the combination was generally well tolerated, there was a mean weight gain of 4.9 kg (10.8 lb). This raises the specter of complications associated with substantial weight gain produced by several of the novel antipsychotics.

        A recent report indicates that ziprasidone may also be an effective antimanic agent. In a randomized, double-blind, placebo-controlled, multicenter trial involving 210 bipolar (manic or mixed episodes) patients, ziprasidone (80 to 160 mg/d; n=140) was compared to a placebo (n=70) for 3 weeks.17 By day 2 and all subsequent time points, ziprasidone was superior in terms of mean change scores from the baseline MRS; produced a more rapid and significantly greater improvement in overall psychopathology in both positive and negative symptoms; and did not produce significant adverse effects (including relevant ECG parameter changes) when compared with the placebo. Similar trials are being conducted for risperidone, aripiprazole, and iloperidone.

        Finally, Meehan et al18 reported on the results of an acute parenteral formulation of olanzapine used to manage agitation in an acute manic or mixed episode. This was a 24-hour, double-blind, placebo-controlled trial comparing intramuscular olanzapine to intramuscular lorazepam. The following results were indicated:

         

        • Olanzapine (doses of 5 to 10 mg) produced a significantly greater reduction in excitation than did the placebo or lorazepam at 30 minutes post-injection.
        • Twice as many patients receiving lorazepam or a placebo versus olanzapine required more than one injection.
        • Except for olanzapine-induced tachycardia in one patient, there were no significant changes in vital signs, ECG parameters, or laboratory assays among the three groups.
        • Somnolence (13%) and dizziness (9%) were the most frequent side effects in the olanzapine group.

        Treatment strategies for depression and mania

        Considering the existing research data, clinical experience, and the risk/benefit ratio, treatment strategies that emphasize the role of antipsychotics in managing severe mood disorders are presented in the algorithms in Figures 1 and 2.

        Figure 1 emphasizes the role of antipsychotics in the pharmacological management of patients with major depression. For unipolar depression with psychotic symptoms, options include an antidepressant plus an antipsychotic; amoxapine monotherapy; and possibly monotherapy with a novel agent such as ziprasidone. For bipolar depression with psychosis or schizoaffective disorder with depression, combining a mood stabilizer such as lithium plus an antipsychotic may be sufficient, but often an antidepressant must also be added. If the response is insufficient, consider switching to a novel antipsychotic (e.g., olanzapine or risperidone) plus a mood stabilizer (± antidepressant). In more serious exacerbations (e.g., high suicidality), ECT may be most appropriate. Secondary choices include clozapine with or without an antidepressant or novel antipsychotic such as risperidone combined with ECT.

         

         

        Figure 2 describes the use of antipsychotics in patients with mania. If response to a primary mood stabilizer such as lithium, valproate, or their combination in the context of a bipolar or schizoaffective disorder is insufficient—or if patients have severe manic or psychotic symptoms—an antipsychotic may be added to the primary mood stabilizer.

        Alternatively, when mood stabilizers are not tolerated or a clinical situation such as pregnancy precludes their use, a novel agent such as olanzapine or risperidone may be given as monotherapy. While the safety of these agents in pregnancy is not clearly established, clinical experience thus far indicates they may be safer than agents such as valproate or carbamazepine. These agents would be the first choice given their diminished propensity for extrapyramidal symptoms; absence of clozapine-related adverse effects such as agranulocytosis and seizures; and growing evidence of possible mood stabilizing effects.

         

        Figure 1 ANTIPSYCHOTICS IN THE TREATMENT OF MAJOR DEPRESSION


        RTEmagicC_9ae32bde6f.jpg.jpg

        Figure 2 ANTIPSYCHOTICS IN THE TREATMENT OF MANIA


        RTEmagicC_860ac76e3c.jpg.jpg

        For patients who remain nonresponsive, clozapine should be considered either as monotherapy or combined with valproate and/or lithium. Combining this agent with carbamazepine is not recommended because of the possibility of an increased risk of hematotoxicity.

        Electroconvulsive therapy may be used safely and effectively in patients who are severely ill (e.g., those with manic delirium); pose an immediate danger because of their potential for violence; are in medical crisis; or have medical contraindications to pharmacotherapy. There is preliminary evidence that ECT can be safely administered with novel antipsychotics such as clozapine, risperidone, or olanzapine to produce additional benefit in patients insufficiently responsive to either therapy alone.

        Related resources

         

        Drug brand names

         

        • Amitriptyline • Elavil
        • Amoxapine • Asendin
        • Aripiprazole • (in development)
        • Carbamazepine • Tegretol, Epitol
        • Clozapine • Clozaril
        • Haloperidol • Haldol
        • Iloperidone • (in development)
        • Imipramine • Tofranil
        • Lorazepam • Ativan
        • Olanzapine • Zyprexa
        • Quetiapine • Seroquel
        • Risperidone • Risperdal
        • Valproate sodium • Depacon
        • Ziprasidone • Geodon

        Disclosure

        The author reports that he receives research/grant support from, serves as a consultant for, and on the speaker’s bureau of Janssen Pharmaceutica. He also receives research/grant support from Genentech Inc. and Bristol-Myers Squibb Co.; serves as a consultant for Pfizer Inc., Sepracor, and Novartis Pharmaceuticals Corp.; and is on the speaker’s bureau of Abbott Laboratories, Eli Lilly and Co., Pfizer Inc., Forest Pharmaceuticals, Bristol-Myers Squibb Co., and Wyeth-Ayerst Pharmaceuticals.

        Major mood disorders are challenging to diagnose and often difficult to treat. They entail unipolar depression; bipolar disorder, which includes manic, depressed, or mixed episodes; and schizoaffective disorder, which includes both depressed and bipolar subtypes. Antidepressants and mood stabilizers are the primary pharmacological treatments. They may be insufficient, however, for patients with more severe episodes, often characterized by psychosis and treatment resistance.

        In these patients, antipsychotics have played an important but controversial part in management, primarily as oral or parenteral adjuncts. Literature and clinical experience now support another, unique role for the current generation of novel agents.

        Compared to earlier antipsychotics, these agents produce substantially fewer neurological adverse effects, including acute extrapyramidal and tardive syndromes, and can augment antidepressants and mood stabilizers. In addition, they may:

         

        • Possess a better antipsychotic profile, with enhanced therapeutic effects on positive, negative, cognitive, and mood symptoms
        • Have a role in the acute and long-term management of these disorders when anticipated parenteral formulations become available (e.g., acute intramuscular olanzapine and ziprasidone—and long-acting intramuscular risperidone)
        • Possess inherent thymoleptic properties (see “Unresolved issues with antipsychotics,” below).

         

        UNRESOLVED ISSUES WITH ANTIPSYCHOTICS

         

        1. Defining what constitutes a mood stabilizer.1 Proposed definitions suggest that the drug must entail the following:
        2. Clarifying the mechanisms underlying the apparent mood-regulating effects of novel agents
        3. Ascertaining both acute and maintenance efficacy
        4. Clarifying the propensity of some agents to switch depressed patients into mania
        5. Increasing the number of well-designed studies with sufficient sample sizes, including comparison trials assessing the relative efficacy of different novel agents
        6. Reducing the tendency to publish only positive reports when new drugs are first available
        7. Introducing parenteral formulations of novel agents
        8. Resolving concerns about weight gain, new-onset diabetes, QT c prolongation, and sedation
        9. Rectifying the current level of substantially greater costs

        Management of unipolar depression

        Neuroleptics Delusions and hallucinations indicate a more severe form of depressive disorder, with poor short- and long-term outcomes in comparison to those without psychosis. To illustrate, Table 1 lists a summary of response rates in psychotic and nonpsychotic depressed patients given a tricyclic antidepressant (TCA). The data indicate that patients suffering from psychotic depression typically do not benefit from antidepressant monotherapy and usually require a combination of antidepressant and antipsychotic or, alternatively, electroconvulsive therapy (ECT).

        There is, however, some limited clinical and neuroimaging evidence that amoxapine can be used as an effective monotherapy in this group. Amoxapine is an antidepressant whose primary active metabolite, 8-hydroxy amoxapine, may have antipsychotic properties.1 With the possible exception of amoxapine, combined antipsychotic-antidepressant treatment is the rule.

        Table 1

        Psychotic and nonpsychotic depressed patients’ response to monotherapy with a tricyclic antidepressant

         

         PsychoticNonpsychotic 
         Responders (%) (n=127)Nonresponders (%) (n=236)Responders (%) (n=464)Nonresponders (%) (n=227)Difference
        13 studies35%65%67%33%32%
        Adapted from Chan CH, Janicak PG, Davis JM, et al. Response of psychotic and nonpsychotic depressed patients to tricyclic antidepressants. J Clin Psychiatry. 1987;48:197-200.

        Historically, studies have also evaluated neuroleptic monotherapy for depressed patients. While some reported superiority over a placebo, none found conventional antipsychotics superior to imipramine. Indeed, patients with schizophrenia who are treated with a neuroleptic often develop symptoms that are difficult to distinguish from depression (e.g., secondary negative symptoms). These often improve when the neuroleptic is discontinued or the patients are switched to a novel antipsychotic such as risperidone, olanzapine, or ziprasidone, all of which have putative antidepressant effects.

        When employing an antipsychotic in depressed patients, the dosage and duration of treatment are two critical considerations. To minimize neuromotor adverse effects, use low doses of a neuroleptic (e.g., haloperidol, 1 to 5 mg/d) in conjunction with the primary antidepressant therapy. The neuroleptic should then be tapered gradually after psychotic symptoms have been controlled, usually during the acute phase of treatment. Ideally, patients would then take antidepressant monotherapy through the continuation phase and, if necessary, the maintenance phase of treatment. If psychosis recurs, re-introduce the antipsychotic intermittently.

        Novel antipsychotics In contrast to neuroleptics, novel antipsychotics have been reported to improve depression in various psychotic and mood disorders.

        For example, ziprasidone has serotonin and noradrenergic reuptake blocking effects comparable to such classic TCAs as imipramine and amitriptyline, as well as high binding affinity at the 5-HT1A, 5-HT1D, and 5-HT2C receptors. This neuroreceptor profile indicates possible antidepressant effects.

        While randomized, controlled trials with mood-disordered patients are few, there have been promising preliminary reports of augmentation of antidepressants with risperidone and olanzapine in both psychotic and nonpsychotic depressed patients.

        Ostroff and Nelson2 reported the results of an open-label study of eight SSRI-nonresponsive patients (mean treatment 7.3 weeks). These patients had no psychotic features and had a dramatic reduction in depressive symptoms, as well as some improvement in sexual dysfunction, with the addition of 0.5 mg to 1.0 mg risperidone. The clinicians suggested that risperidone’s 5-HT2A antagonism might explain its augmentation of the partial SSRI response.

         

         

        Olanzapine alone (n=3) or combined with an antidepressant (n=12) has also been reported to improve both depression and psychosis.3 In a double-blind, amitriptyline-controlled trial, Svestka and Synek4 found that olanzapine demonstrated antidepressant efficacy in 33 unipolar and seven bipolar depressed patients. Thirteen of these patients also had psychotic symptoms.

        Shelton et al5 reported the results of a two-center, 8-week, double-blind comparison of olanzapine alone, fluoxetine alone, or their combination in 28 patients suffering from treatment-resistant, non-bipolar disorder without psychosis. They found that the combination was superior to either drug alone based on improvement in the Hamilton Depression Rating Scale (HDRS) total score. From their preliminary data, it also appears that the doses required were relatively low, reducing the risk of side effects.

        Their findings, however, need to be replicated in more controlled studies with combinations, addressing possible adverse effects, the potential for clinically relevant drug interactions, decreased compliance rates, and increased cost of treatment. Earlier reports raised concern about the potential of these agents to increase switching to hypomania or mania. But in more recent reports, this has not emerged as a significant problem.7

        Finally, several case reports and case series indicate that agents such as clozapine and risperidone may augment ECT in particularly severe, treatment-resistant depressive episodes.7

        Management of bipolar and schizoaffective depressed episodes

        Neuroleptics Antipsychotics are frequently used to manage more severe, usually psychotic episodes of bipolar and schizoaffective depression. Reports indicate that affectively ill patients receiving neuroleptics may be more prone to develop neuromotor adverse effects than are those suffering from schizophrenia. Thus, their use for such patients must be well justified, limited in dosage and duration, and carefully monitored for the emergence of acute and tardive neurological events.

        Novel antipsychotics Novel antipsychotics have demonstrated fewer propensities than have neuroleptics in worsening depression or negative symptoms in schizophrenic patients, and have possible antidepressant effects. In support of this hypothesis, and reminiscent of data from earlier risperidone and olanzapine trials, ziprasidone was observed to improve the Montgomery Asberg Rating Scale (MADRS) and Brief Psychotic Rating Scale (BPRS) depressive cluster scores in three clinical trials with schizophrenic and schizoaffective patients.8,9

        Vieta et al reported the efficacy and safety of risperidone add-on therapy for treating various episodes of bipolar (n=358) and schizoaffective (n=183) disorders.6 In this multicenter, open study, 33 patients (6.1%) suffered a depressed episode and received a mean risperidone dose of 1.6 (± 2.3) mg/d added to their ongoing but ineffective drug regimen. Mean HDRS declined significantly over the 6-month course. Further, switch rates were low and in the expected range for spontaneous fluctuations seen in these disorders.

        The results of a 6-week, double-blind, controlled trial of risperidone versus haloperidol in 62 patients with schizoaffective disorder, bipolar or depressed subtype, were published.10 Risperidone (average dose of 5.5 mg/d) was comparable to haloperidol (average dose of 10.8 mg/d) in reducing the mean in the Positive and Negative Syndrome Scale and Clinician-Administered Rating Scale for Mania change scores.

        In those patients with baseline HDRS scores ≥ 20, risperidone produced a significantly greater reduction in mean change scores than did haloperidol. In addition, patients had no mood switches with risperidone or haloperidol; there was a significantly higher incidence of patients who had extra-pyramidal symptoms with haloperidol than among those taking risperidone; and six patients in the group taking haloperidol dropped out after experiencing adverse effects. None of the patients taking risperidone dropped out.

        Table 2

        Lithium versus antipsychotics for acute mania

         

         LithiumAntipsychotics 
         Responders (%) (n=64)Nonresponders (%) (n=10)Responders (%) (n=38)Nonresponders (%) (n=33)Difference
        5 studies89%11%54%46%35%
        Adapted from Janicak PG, Newman RH, Davis JM. Advances in the treatment of mania and related disorders: a reappraisal. Psychiatric Ann. 1992;22(2):94.

        Management of bipolar manic or mixed episodes

        Up to 80% of all bipolar patients receive an antipsychotic drug during the acute and/or maintenance phase of their illness, even though loading doses of valproate and benzodiazepines may also be used during an exacerbation and pose much less risk, especially in terms of adverse neurological effects.

        Neuroleptics Shortly after their introduction, neuroleptics were found to reduce mortality secondary to dehydration and exhaustion in many highly agitated patients during an acute manic episode such as lethal catatonia.7

        While earlier controlled studies found these agents to be effective in the treatment of acute mania, they are clearly less efficacious than lithium for core manic symptoms.11Table 2 demonstrates a meta-analysis of five well-controlled, double-blind studies documenting the statistical superiority of lithium over neuroleptics. These agents, however, offer the advantage of a more rapid onset of action, particularly when given in the acute parenteral formulation, and are superior to lithium in the initial control of agitation. Further, long-acting depot formulations of neuroleptics may be the only viable strategy for chronic, recurrent, noncompliant patients.

         

         

        As with psychotic depression, dosing and duration of neuroleptic treatment are important concerns. In this context, Rifkin et al demonstrated that 10 mg of haloperidol per day had comparable efficacy but fewer adverse effects than did 30 or 80 mg per day in a group of acutely manic patients.12 Despite such data, high chlorpromazine-equivalent doses are often administered acutely and maintained for sustained periods. This can be a significant problem given the apparent great sensitivity of bipolar patients to the neurological sequelae of these antipsychotic agents.

        Novel antipsychotics Early case series reports indicated that clozapine may benefit treatment-refractory bipolar patients. Given the inherent drawbacks of clozapine (e.g., agranulocytosis and seizure induction), attention now focuses on other novel agents with more benign adverse effect profiles than clopazine. Controlled trials with olanzapine and risperidone serve to reinforce the usefulness of these as well as other novel agents.

        Tohen et al published the results of a 3-week, double-blind, placebo-controlled trial of olanzapine in 139 patients experiencing an acute bipolar manic or mixed episode.13 Olanzapine produced a statistically greater mean improvement than did the placebo on the Young Mania Rating Scale (YMRS) change scores. Further, 49% of the olanzapine-treated group (n=70) met the a priori criteria for response versus only 24% of the placebo-treated group (n=69). A second study using a higher starting dose of olanzapine, less rescue medication, and longer treatment duration than the first study resulted in a similar outcome.14

        Sachs et al reported on the results of a 3-week, double-blind, placebo-controlled trial involving 156 patients with bipolar manic or mixed subtype who received a mood stabilizer (lithium or valproate) plus a placebo, risperidone (1 to 6 mg/d), or haloperidol (2 to 12 mg/d).15 The clinicians concluded that risperidone plus a mood stabilizer was statistically superior to a placebo plus a mood stabilizer, and produced more rapid reduction in manic symptoms, regardless of whether psychosis was present.

        Sajatovic et al16 published the results of a prospective, open trial with quetiapine (mean dose = 203 ± 124 mg/d) as add-on therapy in 20 patients (10 bipolar, 10 schizoaffective; 19 male, 1 female) insufficiently responsive to their mood stabilizer or antipsychotic. Pre-post assessments indicated significant improvement in the BPRS, Mania Rating Scale (MRS), and HDRS scores. While the combination was generally well tolerated, there was a mean weight gain of 4.9 kg (10.8 lb). This raises the specter of complications associated with substantial weight gain produced by several of the novel antipsychotics.

        A recent report indicates that ziprasidone may also be an effective antimanic agent. In a randomized, double-blind, placebo-controlled, multicenter trial involving 210 bipolar (manic or mixed episodes) patients, ziprasidone (80 to 160 mg/d; n=140) was compared to a placebo (n=70) for 3 weeks.17 By day 2 and all subsequent time points, ziprasidone was superior in terms of mean change scores from the baseline MRS; produced a more rapid and significantly greater improvement in overall psychopathology in both positive and negative symptoms; and did not produce significant adverse effects (including relevant ECG parameter changes) when compared with the placebo. Similar trials are being conducted for risperidone, aripiprazole, and iloperidone.

        Finally, Meehan et al18 reported on the results of an acute parenteral formulation of olanzapine used to manage agitation in an acute manic or mixed episode. This was a 24-hour, double-blind, placebo-controlled trial comparing intramuscular olanzapine to intramuscular lorazepam. The following results were indicated:

         

        • Olanzapine (doses of 5 to 10 mg) produced a significantly greater reduction in excitation than did the placebo or lorazepam at 30 minutes post-injection.
        • Twice as many patients receiving lorazepam or a placebo versus olanzapine required more than one injection.
        • Except for olanzapine-induced tachycardia in one patient, there were no significant changes in vital signs, ECG parameters, or laboratory assays among the three groups.
        • Somnolence (13%) and dizziness (9%) were the most frequent side effects in the olanzapine group.

        Treatment strategies for depression and mania

        Considering the existing research data, clinical experience, and the risk/benefit ratio, treatment strategies that emphasize the role of antipsychotics in managing severe mood disorders are presented in the algorithms in Figures 1 and 2.

        Figure 1 emphasizes the role of antipsychotics in the pharmacological management of patients with major depression. For unipolar depression with psychotic symptoms, options include an antidepressant plus an antipsychotic; amoxapine monotherapy; and possibly monotherapy with a novel agent such as ziprasidone. For bipolar depression with psychosis or schizoaffective disorder with depression, combining a mood stabilizer such as lithium plus an antipsychotic may be sufficient, but often an antidepressant must also be added. If the response is insufficient, consider switching to a novel antipsychotic (e.g., olanzapine or risperidone) plus a mood stabilizer (± antidepressant). In more serious exacerbations (e.g., high suicidality), ECT may be most appropriate. Secondary choices include clozapine with or without an antidepressant or novel antipsychotic such as risperidone combined with ECT.

         

         

        Figure 2 describes the use of antipsychotics in patients with mania. If response to a primary mood stabilizer such as lithium, valproate, or their combination in the context of a bipolar or schizoaffective disorder is insufficient—or if patients have severe manic or psychotic symptoms—an antipsychotic may be added to the primary mood stabilizer.

        Alternatively, when mood stabilizers are not tolerated or a clinical situation such as pregnancy precludes their use, a novel agent such as olanzapine or risperidone may be given as monotherapy. While the safety of these agents in pregnancy is not clearly established, clinical experience thus far indicates they may be safer than agents such as valproate or carbamazepine. These agents would be the first choice given their diminished propensity for extrapyramidal symptoms; absence of clozapine-related adverse effects such as agranulocytosis and seizures; and growing evidence of possible mood stabilizing effects.

         

        Figure 1 ANTIPSYCHOTICS IN THE TREATMENT OF MAJOR DEPRESSION


        RTEmagicC_9ae32bde6f.jpg.jpg

        Figure 2 ANTIPSYCHOTICS IN THE TREATMENT OF MANIA


        RTEmagicC_860ac76e3c.jpg.jpg

        For patients who remain nonresponsive, clozapine should be considered either as monotherapy or combined with valproate and/or lithium. Combining this agent with carbamazepine is not recommended because of the possibility of an increased risk of hematotoxicity.

        Electroconvulsive therapy may be used safely and effectively in patients who are severely ill (e.g., those with manic delirium); pose an immediate danger because of their potential for violence; are in medical crisis; or have medical contraindications to pharmacotherapy. There is preliminary evidence that ECT can be safely administered with novel antipsychotics such as clozapine, risperidone, or olanzapine to produce additional benefit in patients insufficiently responsive to either therapy alone.

        Related resources

         

        Drug brand names

         

        • Amitriptyline • Elavil
        • Amoxapine • Asendin
        • Aripiprazole • (in development)
        • Carbamazepine • Tegretol, Epitol
        • Clozapine • Clozaril
        • Haloperidol • Haldol
        • Iloperidone • (in development)
        • Imipramine • Tofranil
        • Lorazepam • Ativan
        • Olanzapine • Zyprexa
        • Quetiapine • Seroquel
        • Risperidone • Risperdal
        • Valproate sodium • Depacon
        • Ziprasidone • Geodon

        Disclosure

        The author reports that he receives research/grant support from, serves as a consultant for, and on the speaker’s bureau of Janssen Pharmaceutica. He also receives research/grant support from Genentech Inc. and Bristol-Myers Squibb Co.; serves as a consultant for Pfizer Inc., Sepracor, and Novartis Pharmaceuticals Corp.; and is on the speaker’s bureau of Abbott Laboratories, Eli Lilly and Co., Pfizer Inc., Forest Pharmaceuticals, Bristol-Myers Squibb Co., and Wyeth-Ayerst Pharmaceuticals.

        References

         

        1. Kapur S, Cho R, Jones C, et al. Is amoxapine an atypical antipsychotic? Positronemission tomography investigation of its dopamine2 and serotonin2 occupancy. Biol Psychiatry. 1999;45:1217-1220.

        2. Ostroff RB, Nelson JC. Risperidone augmentation of selective serotonin reuptake inhibitors in major depression. J Clin Psychiatry. 1999;60:256-259.

        3. Rothschild AJ, Bates KS, Boehringer KL, Syed A. Olanzapine response in psychotic depression. J Clin Psychiatry. 1999;60:116-118.

        4. Svestka J, Synek O. Does olanzapine have antidepressant effect? A double-blind amitriptyline-controlled study [abstract]. Int J Neuropsychopharmacol. 2000;3(suppl 1):S251.-

        5. Shelton RC, Tollefson GD, Tohen M, et al. A novel augmentation strategy for treating resistant major depression. Am J Psychiatry. 2001;158:131-134.

        6. Vieta E, Goikolea JM, Corbella B, et al. Risperidone safety and efficacy in the treatment of bipolar and schizoaffective disorders: results from a 5-month, multicenter, open study. J Clin Psychiatry. 2001;62(10):818.-

        7. Janicak PG, Davis JM, et al. Principles and Practice of Psychopharmacotherapy. 3rd ed. Philadelphia, Pa: Lippincott-Williams & Wilkins; 2001.

        8. Daniel DG, Zimbroff DL, et al. for the Ziprasidone Study Group Ziprasidone 80 mg/day and 160 mg/day in the acute exacerbation of schizophrenia and schizoaffective disorder: a 6-week placebo-controlled trial. Neuropsychopharmacol. 1999;20(5):491-505.

        9. Keck PE, Jr, Buffenstein A, Ferguson J, et al. Ziprasidone 40 and 120 mg/day in the acute exacerbation of schizophrenia and schizoaffective disorder: a 4-week placebo-controlled trial. Psychopharmacol. 1998;140:173-184.

        10. Janicak PG, Keck PE, Jr, Davis JM, et al. A double-blind, randomized, prospective evaluation of the efficacy and safety of risperidone versus haloperidol in the treatment of schizoaffective disorder. J Clin Psychopharmacol. 2001;21:360-368.

        11. Keck PE, Welge JA, McElroy SL, et al. Placebo effect in randomized, controlled studies of acute bipolar mania and depression. Biol Psychiatry. 2000;47(8):756-761.

        12. Rifkin A, Doddi S, Karajgi B, et al. Dosage of haloperidol for mania. Br J Psychiatry. 1994;165:113-116.

        13. Tohen M, Sanger TM, McElroy SL, et al. Olanzipine versus placebo in the treatment of acute mania. Olanzapine HGEH Study Group. Am J Psychiatry. 1999;156:702-709.

        14. Tohen M, Jacobs TG, Grundy SL, et al. Efficacy of olanzapine in acute bipolar mania: a double-blind, placebo-controlled study. The Olanzipine HGGW Study Group. Arch Gen Psychiatry. 2000;57:841-849.

        15. Sachs G, Ghaemi N, Grossman F, Bowden C. Risperidone plus mood stabilizer vs. placebo plus mood stabilizer for acute mania of bipolar disorder: a double-blind comparison of efficacy and safety. International Congress on Bipolar Disorders. Pittsburgh, Pa. June 14-16, 2001.

        16. Sajatovic M, Briscan DW, Perez DE, et al. Quetiapine alone and added to a mood stabilizer for serious mood disorders. J Clin Psychiatry. 2001;62:728-732.

        17. Giller E, Mandel FS, Keck P. Ziprasidone in the acute treatment of mania: a double-blind, placebo-controlled, randomized trial. Schizophr Res. 2001;49(suppl 1-2):229.-

        18. Meehan K, Zhang F, David S, Tohen N, Janicak PG, et al. A double-blind, randomized comparison of the efficacy and safety of intramuscular (IM) olanzapine versus IM lorazepam and IM placebo in acutely agitated patients diagnosed with mania associated with bipolar disorder. J Clin Psychopharmacol 2001;21:389-397.

        References

         

        1. Kapur S, Cho R, Jones C, et al. Is amoxapine an atypical antipsychotic? Positronemission tomography investigation of its dopamine2 and serotonin2 occupancy. Biol Psychiatry. 1999;45:1217-1220.

        2. Ostroff RB, Nelson JC. Risperidone augmentation of selective serotonin reuptake inhibitors in major depression. J Clin Psychiatry. 1999;60:256-259.

        3. Rothschild AJ, Bates KS, Boehringer KL, Syed A. Olanzapine response in psychotic depression. J Clin Psychiatry. 1999;60:116-118.

        4. Svestka J, Synek O. Does olanzapine have antidepressant effect? A double-blind amitriptyline-controlled study [abstract]. Int J Neuropsychopharmacol. 2000;3(suppl 1):S251.-

        5. Shelton RC, Tollefson GD, Tohen M, et al. A novel augmentation strategy for treating resistant major depression. Am J Psychiatry. 2001;158:131-134.

        6. Vieta E, Goikolea JM, Corbella B, et al. Risperidone safety and efficacy in the treatment of bipolar and schizoaffective disorders: results from a 5-month, multicenter, open study. J Clin Psychiatry. 2001;62(10):818.-

        7. Janicak PG, Davis JM, et al. Principles and Practice of Psychopharmacotherapy. 3rd ed. Philadelphia, Pa: Lippincott-Williams & Wilkins; 2001.

        8. Daniel DG, Zimbroff DL, et al. for the Ziprasidone Study Group Ziprasidone 80 mg/day and 160 mg/day in the acute exacerbation of schizophrenia and schizoaffective disorder: a 6-week placebo-controlled trial. Neuropsychopharmacol. 1999;20(5):491-505.

        9. Keck PE, Jr, Buffenstein A, Ferguson J, et al. Ziprasidone 40 and 120 mg/day in the acute exacerbation of schizophrenia and schizoaffective disorder: a 4-week placebo-controlled trial. Psychopharmacol. 1998;140:173-184.

        10. Janicak PG, Keck PE, Jr, Davis JM, et al. A double-blind, randomized, prospective evaluation of the efficacy and safety of risperidone versus haloperidol in the treatment of schizoaffective disorder. J Clin Psychopharmacol. 2001;21:360-368.

        11. Keck PE, Welge JA, McElroy SL, et al. Placebo effect in randomized, controlled studies of acute bipolar mania and depression. Biol Psychiatry. 2000;47(8):756-761.

        12. Rifkin A, Doddi S, Karajgi B, et al. Dosage of haloperidol for mania. Br J Psychiatry. 1994;165:113-116.

        13. Tohen M, Sanger TM, McElroy SL, et al. Olanzipine versus placebo in the treatment of acute mania. Olanzapine HGEH Study Group. Am J Psychiatry. 1999;156:702-709.

        14. Tohen M, Jacobs TG, Grundy SL, et al. Efficacy of olanzapine in acute bipolar mania: a double-blind, placebo-controlled study. The Olanzipine HGGW Study Group. Arch Gen Psychiatry. 2000;57:841-849.

        15. Sachs G, Ghaemi N, Grossman F, Bowden C. Risperidone plus mood stabilizer vs. placebo plus mood stabilizer for acute mania of bipolar disorder: a double-blind comparison of efficacy and safety. International Congress on Bipolar Disorders. Pittsburgh, Pa. June 14-16, 2001.

        16. Sajatovic M, Briscan DW, Perez DE, et al. Quetiapine alone and added to a mood stabilizer for serious mood disorders. J Clin Psychiatry. 2001;62:728-732.

        17. Giller E, Mandel FS, Keck P. Ziprasidone in the acute treatment of mania: a double-blind, placebo-controlled, randomized trial. Schizophr Res. 2001;49(suppl 1-2):229.-

        18. Meehan K, Zhang F, David S, Tohen N, Janicak PG, et al. A double-blind, randomized comparison of the efficacy and safety of intramuscular (IM) olanzapine versus IM lorazepam and IM placebo in acutely agitated patients diagnosed with mania associated with bipolar disorder. J Clin Psychopharmacol 2001;21:389-397.

        Issue
        Current Psychiatry - 01(01)
        Issue
        Current Psychiatry - 01(01)
        Page Number
        11-17
        Page Number
        11-17
        Publications
        Publications
        Topics
        Article Type
        Display Headline
        Antipsychotics and mood disorders: A complicated alliance
        Display Headline
        Antipsychotics and mood disorders: A complicated alliance
        Sections
        PURLs Copyright

        Disallow All Ads
        Alternative CME
        Use ProPublica
        Hide sidebar & use full width
        render the right sidebar.