Fatigue associated with depression

Article Type
Changed
Thu, 09/27/2018 - 15:53
Display Headline
Fatigue associated with depression

Author and Disclosure Information

Dr. Macaluso is Assistant Professor, Director of Residency Training, and Director of Clinical Trails Research, Department of Psychiatry and Behavioral Sciences, University of Kansas School of Medicine–Wichita,
Wichita, Kansas.

Issue
Current Psychiatry - 14(6)
Publications
Topics
Legacy Keywords
depressive disorders, depression, fatigue, fatigue and depression, fatigue caused by depression, depression caused by fatigue
Sections
Author and Disclosure Information

Dr. Macaluso is Assistant Professor, Director of Residency Training, and Director of Clinical Trails Research, Department of Psychiatry and Behavioral Sciences, University of Kansas School of Medicine–Wichita,
Wichita, Kansas.

Author and Disclosure Information

Dr. Macaluso is Assistant Professor, Director of Residency Training, and Director of Clinical Trails Research, Department of Psychiatry and Behavioral Sciences, University of Kansas School of Medicine–Wichita,
Wichita, Kansas.

Issue
Current Psychiatry - 14(6)
Issue
Current Psychiatry - 14(6)
Publications
Publications
Topics
Article Type
Display Headline
Fatigue associated with depression
Display Headline
Fatigue associated with depression
Legacy Keywords
depressive disorders, depression, fatigue, fatigue and depression, fatigue caused by depression, depression caused by fatigue
Legacy Keywords
depressive disorders, depression, fatigue, fatigue and depression, fatigue caused by depression, depression caused by fatigue
Sections
Disallow All Ads
Alternative CME
Use ProPublica

Fatigue after depression responds to therapy. What are the next steps?

Article Type
Changed
Tue, 12/11/2018 - 15:02
Display Headline
Fatigue after depression responds to therapy. What are the next steps?

Fatigue and depression can be viewed as a “vicious cycle”: Fatigue can be a symptom of major depression, and fatigue can be a risk factor for depression.1 For example, fatigue associated with a general medical condition or traumatic brain injury can be a risk factor for developing major depressive disorder (MDD).1-3 It isn’t surprising that fatigue has been studied as a predictor of relapse after previous response to treatment in patients with MDD.

Despite the observed association between fatigue and depression, their underlying relationship often is unclear. The literature does not differentiate among fatigue associated with depression, fatigue as a treatment-emergent adverse effect, and fatigue as a residual symptom of depression that is partially responsive to treatment.4,5 To complicate the situation, many medications used to treat MDD can cause fatigue.

Patients often describe fatigue as (1) feeling tired, exhausted, or drained and (2) lacking energy and motivation. Fatigue can be related to impaired wakefulness but is believed to be a different entity than sleepiness.6 Residual fatigue can affect social, cognitive, emotional, and physical health.

We reviewed the literature about fatigue as a symptom of MDD by conducting a search of Medline, PubMed, and Google Scholar, using keywords depression, fatigue, residual symptoms, and treatment. We chose the papers cited in this article based on our consensus and because these publications represent expert opinion or the highest quality evi­dence available.


Residual fatigue has an effect on prognosis

Fatigue is a common symptom of MDD that persists in 20% to 30% of patients whose symptoms of depression otherwise remit.4,7-9 Several studies have linked residual fatigue with the overall prognosis of MDD.5 Data from a prospective study demonstrate that depressed patients have a higher risk of relapse when they continue to report symp­toms of fatigue after their symptoms of depression have otherwise entered partial remission.10 Another study demonstrated that the severity of residual symptoms of depression is a strong predictor of another major depressive episode.11

In a large-scale study, the prevalence of residual fatigue after adequate treat­ment of MDD in both partial responders and remitters was 84.6%.12 The same study showed that one-third of patients who had been treated for MDD had persistent and clinically significant fatigue, which could suggest a relationship between fatigue and selective serotonin reuptake inhibitors (SSRIs) and other antidepressants.

Another study demonstrated that 64.6% of patients who responded to antidepressant treatment and who had baseline fatigue con­tinued to exhibit symptoms of fatigue after an adequate trial of an antidepressant.13


Neurobiological considerations
Studies have shown that the neuronal circuits that malfunction in fatigue are different from those that malfunction in depression.14 Although the neurobiol­ogy of fatigue has not been determined, decreased neuronal activity in the prefron­tal circuits has been associated with symp­toms of fatigue.15

In addition, evidence from the litera­ture shows a decrease in hormone secre­tion16 and cognitive abilities in patients exhibiting symptoms of fatigue.17 These findings have led some experts to hypoth­esize that symptoms of fatigue associated with depression could be the result of (1) immune dysregulation18 and (2) an inability of available antidepressants to tar­get the underlying biology of the disorder.2

Despite the hypothesis that fatigue asso­ciated with depression might be biologically related to immune dysregulation, some authors continue to point to an imbalance in neurotransmitters—norepinephrine, his­tamine, dopamine, acetylcholine—as being  associated with fatigue.14 For example, a study demonstrated that drugs targeting noradrenergic reuptake inhibition were more effective at preventing a relapse of fatigue compared with serotonergic drugs.19 Another study showed improvement in energy with an increase in the plasma level of desipramine, which affects noradrener­gic neurotransmission.20

Inflammatory cytokines also have been explored in the search for an understand­ing of the etiology of fatigue and depres­sion.21 Physical and mental stress promote the release of cytokines, which activate the immune system by inducing an inflam­matory response; this response has been etiologically linked to depressive disor­ders.22 Furthermore, studies have demon­strated an elevated level of inflammatory cytokines in patients who have MDD— suggesting that MDD is associated with a chronic low level of inflammation that crosses the blood−brain barrier.23


Clinical considerations: A role for rating scales?

Despite the significance of residual fatigue on the quality of life of patients who have MDD, most common rating scales, such as the Hamilton Depression Rating Scale24 and the Montgomery-Åsberg Depression Rating Scale,25 have limited sensitivity for measuring fatigue.26 The Fatigue Associated with Depression (FAsD)27 questionnaire, designed according to FDA guidelines,28 is used to assess fatigue associated with depression. The final version of the FAsD includes 13 items: a 6-item experience sub­scale and a 7-item impact subscale.

Is the FAsD helpful? The experience sub­scale of the FAsD assesses how often the patient experiences different aspects of fatigue (tiredness, exhaustion, lack of energy, physical weakness, and a feeling that everything requires too much effort). The impact subscale of the FAsD assesses the effect of fatigue on daily life.

 

 

The overall FAsD score is calculated by taking the mean of each subscale; a change of 0.67 on the experience subscale and 0.57 on the impact subscale are considered clini­cally meaningful.27 The measurement prop­erties of the questionnaire showed internal consistency, reliability, and validity in test­ing. Researchers note, however, that FAsD does not include items to assess the impact of fatigue on cognition. This means that the FAsD might not distinguish between physi­cal and mental aspects of fatigue.


Treatment

It isn’t surprising that residual depres­sion can increase health care utilization and economic burden, including such indirect costs as lost productivity and wages.29 Despite these impacts, there is a paucity of studies evaluating the relation­ship between residual symptoms, such as fatigue, and work productivity. It has been established that improving a depressed patient’s level of energy correlates with improved performance at work.

Treating fatigue as a residual symp­tom of MDD can be complicated because symptoms of fatigue might be:
   • a discrete symptom of MDD
   • a prodromal symptom of another disorder
   • an adverse effect of an antidepressant.2,30

It is a major clinical problem, there­fore, that antidepressants can alleviate and cause symptoms of fatigue.31 Treatment strategy should focus on identifying anti­depressants that are less likely to cause fatigue (ie, noradrenergic or dopaminergic drugs, or both). Adjunctive treatments to target residual fatigue also can be used.32

There are limited published data on the effective treatment of residual fatigue in patients with MDD. Given the absence of sufficient evidence, agents that promote noradrenergic and dopaminergic neuro­transmission have been the treatment of choice when targeting fatigue in depressed patients.2,14,21,33

The Table34-37 lists poten­tial treatment options often used to treat fatigue associated with depression.


SSRIs.
Treatment with SSRIs has been asso­ciated with a low probability of achiev­ing remission when targeting fatigue as a symptom of MDD.21

One study reported that, after 8 weeks of treatment with an SSRI, treatment-emergent adverse events, such as worsening fatigue and weakness, were observed—along with an overall lack of efficacy in targeting all symptoms of depression.38

Another study demonstrated positive effects when a noradrenergic agent was added to an SSRI in partial responders who continued to complain of residual fatigue.33

However, studies that compared the effects of SSRIs with those of antidepres­sants that have pronoradrenergic effects showed that the 2 mechanisms of action were not significantly different from each other in their ability to resolve residual symptoms of fatigue.21 A limiting factor might be that these studies were retrospec­tive and did not analyze the efficacy of a noradrenergic agent as an adjunct for alle­viating symptoms of fatigue.39

Bupropion. This commonly used medica­tion for fatigue is believed to cause a sig­nificantly lower level of fatigue compared with SSRIs.40 The potential utility of bupro­pion in this area could be a reflection of its mechanism of action—ie, the drug targets both noradrenergic and dopaminergic neurotransmission.41

A study comparing bupropion with SSRIs in targeting somatic symptoms of depression reported a small but statisti­cally significant difference in favor of the bupropion-treated group. However, this finding was confounded by the small effect size and difficulty quantifying somatic symptoms.40

Stimulants and modafinil. Psycho-stimulants have been shown to be effica­cious for depression and fatigue, both as monotherapy and adjunctively.39,42

Modafinil has demonstrated efficacy in open-label trials for improving residual fatigue, but failed to separate from pla­cebo in controlled trials.43 At least 1 other failed study has been published examining modafinil as a treatment for fatigue associ­ated with depression.43

Adjunctive therapy with CNS stimu­lants, such as amphetamine/dextroam­phetamine and methylphenidate, has been used to treat fatigue, with positive results.16 Modafinil and stimulants also could be tried as an augmentation strategy to other antidepressants; such use is off-label and should be attempted only after careful consideration.16

Exercise might be a nonpharmacothera­peutic modality that targets the underly­ing physiology associated with fatigue. Exercise releases endorphins, which can affect overall brain chemistry and which have been theorized to diminish symp­toms of fatigue and depression.44 Consider exercise in addition to treatment with an antidepressant in selected patients.45


To sum up

In general, the literature does not recom­mend one medication as superior to any other for treating fatigue that is a residual symptom of depression. Such hesitation suggests that more empirical studies are needed to determine what is the best and proper management of treating fatigue associated with depression.


Bottom LinE
Fatigue can be a symptom of major depressive disorder (MDD) or a risk factor for depression. Fatigue has been studied as a predictor of relapse after previous response to treatment in patients with MDD. Residual fatigue can affect social, cognitive, emotional, and physical health and can result in increased utilization of health care services. A number of treatment options are available; none has been shown to be superior to the others.

 

 


Related Resources

• Leone SS. A disabling combination: fatigue and depression. Br J Psychiatry. 2010;197(2):86-87.
• Targum SD, Fava M. Fatigue as a residual symptom of de­pression. Innov Clin Neurosci. 2011;8(10):40-43.
• Illiades C. How to fight depression fatigue. Everyday Health. http://www.everydayhealth.com/health-report/major-depression-living-well/fight-depression-fatigue.aspx.
• Kerr M. Depression and fatigue: a vicious cycle. Healthline. http://www.healthline.com/health/depression/fatigue.


Drug Brand Names

Amphetamine/dextroamphetamine • Adderall              
Bupropion • Wellbutrin                                                
Desipramine • Norpramin                                           
Methylphenidate • Ritalin
Modafinil • Provigil
Sertraline • Zoloft
Venlafaxine • Effexor

Disclosures
Dr. Sohail reports no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Macaluso has conducted clinical trials research as principal investigator for the following pharmaceutical manufacturers in the past 12 months: AbbVie, Inc.; Alkermes; AssureRx Health, Inc.; Eisai Co., Ltd.; FORUM Pharmaceuticals, Inc.; Janssen Pharmaceuticals, Inc.; and Naurex Inc. All clinical trial and study contracts were with, and payments were made to, University of Kansas Medical Center Research Institute, Kansas City, Kansas, a research institute affiliated with University of Kansas School of Medicine−Wichita.

References


1. Schönberger M, Herrberg M, Ponsford J. Fatigue as a cause, not a consequence of depression and daytime sleepiness: a cross-lagged analysis. J Head Trauma Rehabil. 2014;29(5):427-431.
2. Demyttenaere K, De Fruyt J, Stahl, SM. The many faces of fatigue in major depressive disorder. Int J Neuropsychopharmacol. 2005;8(1):93-105.
3. Skapinakis P, Lewis G, Mavreas V. Temporal relations between unexplained fatigue and depression: longitudinal data from an international study in primary care. Psychosom Med. 2004;66(3):330-335.
4. Nierenberg AA, Husain MM, Trivedi MH, et al. Residual symptoms after remission of major depressive disorder with citalopram and risk of relapse: a STAR*D report. Psychol Med. 2010;40(1):41-50.
5. Kennedy N, Paykel ES. Residual symptoms at remission from depression: impact on long-term outcome. J Affect Disord. 2004;80(2-3):135-144.
6. Shen J, Barbera J, Shapiro CM. Distinguishing sleepiness and fatigue: focus on definition and measurement. Sleep Med Rev. 2006;10:63-76.
7. Nierenberg AA, Keefe BR, Leslie VC, et al. Residual symptoms in depressed patients who respond acutely to fluoxetine. J Clin Psychiatry. 1999;60(4):221-225.
8. Tylee A, Gastpar M, Lépine JP, et al. DEPRES II (Depression Research in European Society II): a patient survey of the symptoms, disability and current management of depression in the community. DEPRES Steering Committee. Int Clin Psychopharmacol. 1999;14(3):139-151.
9. Marcus SM, Young EA, Kerber KB, et al. Gender differences in depression: findings from the STAR*D study. J Affect Disord. 2005;87(2-3):141-150.
10. Paykel ES, Ramana, R, Cooper Z, et al. Residual symptoms after partial remission: an important outcome in depression. Psychol Med. 1995;25(6):1171-1180.
11. Bockting CL, Spinhoven P, Koeter MW, et al; Depression Evaluation Longitudinal Therapy Assessment Study Group. Prediction of recurrence in recurrent depression and the influence of consecutive episodes on vulnerability for depression: a 2-year prospective study. J Clin Psychiatry. 2006;67(5):747-755.
12. Greco T, Eckert G, Kroenke K. The outcome of physical symptoms with treatment of depression. J Gen Intern Med. 2004;19(8):813-818.
13. McClintock SM, Husain MM, Wisniewski SR, et al. Residual symptoms in depressed outpatients who respond by 50% but do not remit to antidepressant medication. J Clin Psychopharmacol. 2011;31(2):180-186.
14. Stahl SM, Zhang L, Damatarca C, et al. Brain circuits determine destiny in depression: a novel approach to the psychopharmacology of wakefulness, fatigue, and executive dysfunction in major depressive disorder. J Clin Psychiatry. 2003;64(suppl 14):6-17.
15. MacHale SM, Law´rie SM, Cavanagh JT, et al. Cerebral perfusion in chronic fatigue syndrome and depression. Br J Psychiatry. 2000;176:550-556.
16. Paykel ES. Achieving gains beyond response. Acta Psychiatrica Scandinavica Suppl. 2002;(415):12-17.
17. van den Heuvel OA, Groenewegen HJ, Barkhof F, et al. Frontostriatal system in planning complexity: a parametric functional magnetic resonance version of Tower of London task. Neuroimage. 2003;18(2):367-374.
18. Jaremka LM, Fagundes CP, Glaser R, et al. Loneliness predicts pain, depression, and fatigue: understanding the role of immune dysregulation. Psychoneuroendocrinology. 2013;38(8):1310-1317.
19. Delgado PL, Charney DS, Price LH, et al. Serotonin function and the mechanism of antidepressant action. Reversal of antidepressant-induced remission by rapid depletion of plasma tryptophan. Arch Gen Psychiatry. 1990;47(5):411-418.
20. Nelson JC, Mazure C, Quinlan DM, et al. Drug-responsive symptoms in melancholia. Arch Gen Psychiatry. 1984;41(7):663-668.
21. Fava M, Ball S, Nelson, JC, et al. Clinical relevance of fatigue as a residual symptom in major depressive disorder. Depress Anxiety. 2014;31(3):250-257.
22. Anisman H, Merali Z, Poulter MO, et al. Cytokines as a precipitant of depressive illness: animal and human studies. Curr Pharm Des. 2005;11(8):963-972.
23. Simon NM, McNamara K, Chow CW, et al. A detailed examination of cytokine abnormalities in major depressive disorder. Eur Neuropsychopharmacol. 2008;18(3):230-233.
24. Hamilton M. A rating scale for depression. J Neurol Neurosurg Psychiatry. 1960;23:56-62.
25. Montgomery SA, Asberg M. A new depression scale designed to be sensitive to change. Br J Psychiatry. 1979;134:382-389.
26. Matza LS, Phillips GA, Revicki DA, et al. Development and validation of a patient-report measure of fatigue associated with depression. J Affect Disord. 2011;134(1-3):294-303.
27. Matza LS, Wyrwich KW, Phillips GA, et al. The Fatigue Associated with Depression Questionnaire (FAsD): responsiveness and responder definition. Qual Life Res. 2013;22(2):351-360.
28. Guidance for industry. Patient-reported outcome measures: use in medical product development to support labeling claims. Food and Drug Administration. http://www.fda. gov/downloads/Drugs/Guidances/UCM193282.pdf. Published December 2009. Accessed May 7, 2015.
29. Knoth RL, Bolge SC, Kim E, et al. Effect of inadequate response to treatment in patients with depression. Am J Manag Care. 2010;16(8):e188-e196.
30. Fava M. Symptoms of fatigue and cognitive/executive dysfunction in major depressive disorder before and after antidepressant treatment. J Clin Psychiatry. 2003;64(suppl 14):30-34.
31. Chang T, Fava M. The future of psychopharmacology of depression. J Clin Psychiatry. 2010;71(8):971-975.
32. Baldwin DS, Papakostas GI. Symptoms of fatigue and sleepiness in major depressive disorder. J Clin Psychiatry. 2006;67(suppl 6):9-15.
33. Ball SG, Dellva MA, D’Souza D, et al. A double-blind, placebo-controlled study of augmentation with LY2216684 for major depressive disorder patients who are partial responders to selective serotonin reuptake inhibitors [abstract P 05]. Int J Psych Clin Pract. 2010;14(suppl 1):19.
34. Stahl SM. Using secondary binding properties to select a not so elective serotonin reuptake inhibitor. J Clin Psychiatry. 1998;59(12):642-643.
35. Stahl SM. Essential psychopharmacology: neuroscientific basis and practical applications. 2nd ed. New York, NY: Cambridge University Press; 2000.
36. Bymaster FP, Katner JS, Nelson DL, et al. Atomoxetine increases extracellular levels of norepinephrine and dopamine in prefrontal cortex of rat: a potential mechanism for efficacy in attention deficit/hyperactivity disorder. Neuropsychopharmacology. 2002;27(5):699-711.
37. Scammell TE, Estabrooke IV, McCarthy MT, et al. Hypothalamic arousal regions are activated during modafinil-induced wakefulness. J Neurosci. 2000;20(22):8620-8628.
38. Daly EJ, Trivedi MH, Fava M, et al. The relationship between adverse events during selective serotonin reuptake inhibitor treatment for major depressive disorder and nonremission in the suicide assessment methodology study. J Clin Psychopharmacol. 2011;31(1):31-38.
39. Nelson JC. A review of the efficacy of serotonergic and noradrenergic reuptake inhibitors for treatment of major depression. Biol Psychiatry. 1999;46(9):1301-1308.
40. Papakostas GI, Nutt DJ, Hallett LA, et al. Resolution of sleepiness and fatigue in major depressive disorder: a comparison of bupropion and the selective serotonin reuptake inhibitors. Biol Psychiatry. 2006;60(12):1350-1355.
41. Fava M, Rush AJ, Thase ME, et al. 15 years of clinical experience with bupropion HCl: from bupropion to bupropion SR to bupropion XL. Prim Care Companion J Clin Psychiatry. 2005;7(3):106-113.
42. Candy M, Jones CB, Williams R, et al. Psychostimulants for depression. Cochrane Database Syst Rev. 2008;(2):CD006722. doi: 10.1002/14651858.CD006722.pub2.
43. Lam JY, Freeman MK, Cates ME. Modafinil augmentation for residual symptoms of fatigue in patients with a partial response to antidepressants. Ann Pharmacother. 2007;41(6):1005-1012.
44. Salmon P. Effects of physical exercise on anxiety, depression, and sensitivity to stress: a unifying theory. Clinical Psychol Rev. 2001;21(1):33-61.
45. Trivedi MH, Greer TL, Grannemann BD, et al. Exercise as an augmentation strategy for treatment of major depression. J Psychiatr Pract. 2006;12(4):205-213.

Article PDF
Author and Disclosure Information

Zohaib Sohail, MD
PGY-1 Resident in Psychiatry

Matthew Macaluso, DO
Assistant Professor
Director of Residency Training
Director of Clinical Trials Research

Department of Psychiatry and Behavioral Sciences
University of Kansas School of Medicine−Wichita
Wichita, Kansas

Audio / Podcast
Issue
Current Psychiatry - 14(6)
Publications
Topics
Page Number
16-17, 28-32
Legacy Keywords
fatigue associated with depression, depression, depressive disorders, fatigue and depression, depression caused by fatigue, fatigue caused by depression,
Sections
Audio / Podcast
Audio / Podcast
Author and Disclosure Information

Zohaib Sohail, MD
PGY-1 Resident in Psychiatry

Matthew Macaluso, DO
Assistant Professor
Director of Residency Training
Director of Clinical Trials Research

Department of Psychiatry and Behavioral Sciences
University of Kansas School of Medicine−Wichita
Wichita, Kansas

Author and Disclosure Information

Zohaib Sohail, MD
PGY-1 Resident in Psychiatry

Matthew Macaluso, DO
Assistant Professor
Director of Residency Training
Director of Clinical Trials Research

Department of Psychiatry and Behavioral Sciences
University of Kansas School of Medicine−Wichita
Wichita, Kansas

Article PDF
Article PDF

Fatigue and depression can be viewed as a “vicious cycle”: Fatigue can be a symptom of major depression, and fatigue can be a risk factor for depression.1 For example, fatigue associated with a general medical condition or traumatic brain injury can be a risk factor for developing major depressive disorder (MDD).1-3 It isn’t surprising that fatigue has been studied as a predictor of relapse after previous response to treatment in patients with MDD.

Despite the observed association between fatigue and depression, their underlying relationship often is unclear. The literature does not differentiate among fatigue associated with depression, fatigue as a treatment-emergent adverse effect, and fatigue as a residual symptom of depression that is partially responsive to treatment.4,5 To complicate the situation, many medications used to treat MDD can cause fatigue.

Patients often describe fatigue as (1) feeling tired, exhausted, or drained and (2) lacking energy and motivation. Fatigue can be related to impaired wakefulness but is believed to be a different entity than sleepiness.6 Residual fatigue can affect social, cognitive, emotional, and physical health.

We reviewed the literature about fatigue as a symptom of MDD by conducting a search of Medline, PubMed, and Google Scholar, using keywords depression, fatigue, residual symptoms, and treatment. We chose the papers cited in this article based on our consensus and because these publications represent expert opinion or the highest quality evi­dence available.


Residual fatigue has an effect on prognosis

Fatigue is a common symptom of MDD that persists in 20% to 30% of patients whose symptoms of depression otherwise remit.4,7-9 Several studies have linked residual fatigue with the overall prognosis of MDD.5 Data from a prospective study demonstrate that depressed patients have a higher risk of relapse when they continue to report symp­toms of fatigue after their symptoms of depression have otherwise entered partial remission.10 Another study demonstrated that the severity of residual symptoms of depression is a strong predictor of another major depressive episode.11

In a large-scale study, the prevalence of residual fatigue after adequate treat­ment of MDD in both partial responders and remitters was 84.6%.12 The same study showed that one-third of patients who had been treated for MDD had persistent and clinically significant fatigue, which could suggest a relationship between fatigue and selective serotonin reuptake inhibitors (SSRIs) and other antidepressants.

Another study demonstrated that 64.6% of patients who responded to antidepressant treatment and who had baseline fatigue con­tinued to exhibit symptoms of fatigue after an adequate trial of an antidepressant.13


Neurobiological considerations
Studies have shown that the neuronal circuits that malfunction in fatigue are different from those that malfunction in depression.14 Although the neurobiol­ogy of fatigue has not been determined, decreased neuronal activity in the prefron­tal circuits has been associated with symp­toms of fatigue.15

In addition, evidence from the litera­ture shows a decrease in hormone secre­tion16 and cognitive abilities in patients exhibiting symptoms of fatigue.17 These findings have led some experts to hypoth­esize that symptoms of fatigue associated with depression could be the result of (1) immune dysregulation18 and (2) an inability of available antidepressants to tar­get the underlying biology of the disorder.2

Despite the hypothesis that fatigue asso­ciated with depression might be biologically related to immune dysregulation, some authors continue to point to an imbalance in neurotransmitters—norepinephrine, his­tamine, dopamine, acetylcholine—as being  associated with fatigue.14 For example, a study demonstrated that drugs targeting noradrenergic reuptake inhibition were more effective at preventing a relapse of fatigue compared with serotonergic drugs.19 Another study showed improvement in energy with an increase in the plasma level of desipramine, which affects noradrener­gic neurotransmission.20

Inflammatory cytokines also have been explored in the search for an understand­ing of the etiology of fatigue and depres­sion.21 Physical and mental stress promote the release of cytokines, which activate the immune system by inducing an inflam­matory response; this response has been etiologically linked to depressive disor­ders.22 Furthermore, studies have demon­strated an elevated level of inflammatory cytokines in patients who have MDD— suggesting that MDD is associated with a chronic low level of inflammation that crosses the blood−brain barrier.23


Clinical considerations: A role for rating scales?

Despite the significance of residual fatigue on the quality of life of patients who have MDD, most common rating scales, such as the Hamilton Depression Rating Scale24 and the Montgomery-Åsberg Depression Rating Scale,25 have limited sensitivity for measuring fatigue.26 The Fatigue Associated with Depression (FAsD)27 questionnaire, designed according to FDA guidelines,28 is used to assess fatigue associated with depression. The final version of the FAsD includes 13 items: a 6-item experience sub­scale and a 7-item impact subscale.

Is the FAsD helpful? The experience sub­scale of the FAsD assesses how often the patient experiences different aspects of fatigue (tiredness, exhaustion, lack of energy, physical weakness, and a feeling that everything requires too much effort). The impact subscale of the FAsD assesses the effect of fatigue on daily life.

 

 

The overall FAsD score is calculated by taking the mean of each subscale; a change of 0.67 on the experience subscale and 0.57 on the impact subscale are considered clini­cally meaningful.27 The measurement prop­erties of the questionnaire showed internal consistency, reliability, and validity in test­ing. Researchers note, however, that FAsD does not include items to assess the impact of fatigue on cognition. This means that the FAsD might not distinguish between physi­cal and mental aspects of fatigue.


Treatment

It isn’t surprising that residual depres­sion can increase health care utilization and economic burden, including such indirect costs as lost productivity and wages.29 Despite these impacts, there is a paucity of studies evaluating the relation­ship between residual symptoms, such as fatigue, and work productivity. It has been established that improving a depressed patient’s level of energy correlates with improved performance at work.

Treating fatigue as a residual symp­tom of MDD can be complicated because symptoms of fatigue might be:
   • a discrete symptom of MDD
   • a prodromal symptom of another disorder
   • an adverse effect of an antidepressant.2,30

It is a major clinical problem, there­fore, that antidepressants can alleviate and cause symptoms of fatigue.31 Treatment strategy should focus on identifying anti­depressants that are less likely to cause fatigue (ie, noradrenergic or dopaminergic drugs, or both). Adjunctive treatments to target residual fatigue also can be used.32

There are limited published data on the effective treatment of residual fatigue in patients with MDD. Given the absence of sufficient evidence, agents that promote noradrenergic and dopaminergic neuro­transmission have been the treatment of choice when targeting fatigue in depressed patients.2,14,21,33

The Table34-37 lists poten­tial treatment options often used to treat fatigue associated with depression.


SSRIs.
Treatment with SSRIs has been asso­ciated with a low probability of achiev­ing remission when targeting fatigue as a symptom of MDD.21

One study reported that, after 8 weeks of treatment with an SSRI, treatment-emergent adverse events, such as worsening fatigue and weakness, were observed—along with an overall lack of efficacy in targeting all symptoms of depression.38

Another study demonstrated positive effects when a noradrenergic agent was added to an SSRI in partial responders who continued to complain of residual fatigue.33

However, studies that compared the effects of SSRIs with those of antidepres­sants that have pronoradrenergic effects showed that the 2 mechanisms of action were not significantly different from each other in their ability to resolve residual symptoms of fatigue.21 A limiting factor might be that these studies were retrospec­tive and did not analyze the efficacy of a noradrenergic agent as an adjunct for alle­viating symptoms of fatigue.39

Bupropion. This commonly used medica­tion for fatigue is believed to cause a sig­nificantly lower level of fatigue compared with SSRIs.40 The potential utility of bupro­pion in this area could be a reflection of its mechanism of action—ie, the drug targets both noradrenergic and dopaminergic neurotransmission.41

A study comparing bupropion with SSRIs in targeting somatic symptoms of depression reported a small but statisti­cally significant difference in favor of the bupropion-treated group. However, this finding was confounded by the small effect size and difficulty quantifying somatic symptoms.40

Stimulants and modafinil. Psycho-stimulants have been shown to be effica­cious for depression and fatigue, both as monotherapy and adjunctively.39,42

Modafinil has demonstrated efficacy in open-label trials for improving residual fatigue, but failed to separate from pla­cebo in controlled trials.43 At least 1 other failed study has been published examining modafinil as a treatment for fatigue associ­ated with depression.43

Adjunctive therapy with CNS stimu­lants, such as amphetamine/dextroam­phetamine and methylphenidate, has been used to treat fatigue, with positive results.16 Modafinil and stimulants also could be tried as an augmentation strategy to other antidepressants; such use is off-label and should be attempted only after careful consideration.16

Exercise might be a nonpharmacothera­peutic modality that targets the underly­ing physiology associated with fatigue. Exercise releases endorphins, which can affect overall brain chemistry and which have been theorized to diminish symp­toms of fatigue and depression.44 Consider exercise in addition to treatment with an antidepressant in selected patients.45


To sum up

In general, the literature does not recom­mend one medication as superior to any other for treating fatigue that is a residual symptom of depression. Such hesitation suggests that more empirical studies are needed to determine what is the best and proper management of treating fatigue associated with depression.


Bottom LinE
Fatigue can be a symptom of major depressive disorder (MDD) or a risk factor for depression. Fatigue has been studied as a predictor of relapse after previous response to treatment in patients with MDD. Residual fatigue can affect social, cognitive, emotional, and physical health and can result in increased utilization of health care services. A number of treatment options are available; none has been shown to be superior to the others.

 

 


Related Resources

• Leone SS. A disabling combination: fatigue and depression. Br J Psychiatry. 2010;197(2):86-87.
• Targum SD, Fava M. Fatigue as a residual symptom of de­pression. Innov Clin Neurosci. 2011;8(10):40-43.
• Illiades C. How to fight depression fatigue. Everyday Health. http://www.everydayhealth.com/health-report/major-depression-living-well/fight-depression-fatigue.aspx.
• Kerr M. Depression and fatigue: a vicious cycle. Healthline. http://www.healthline.com/health/depression/fatigue.


Drug Brand Names

Amphetamine/dextroamphetamine • Adderall              
Bupropion • Wellbutrin                                                
Desipramine • Norpramin                                           
Methylphenidate • Ritalin
Modafinil • Provigil
Sertraline • Zoloft
Venlafaxine • Effexor

Disclosures
Dr. Sohail reports no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Macaluso has conducted clinical trials research as principal investigator for the following pharmaceutical manufacturers in the past 12 months: AbbVie, Inc.; Alkermes; AssureRx Health, Inc.; Eisai Co., Ltd.; FORUM Pharmaceuticals, Inc.; Janssen Pharmaceuticals, Inc.; and Naurex Inc. All clinical trial and study contracts were with, and payments were made to, University of Kansas Medical Center Research Institute, Kansas City, Kansas, a research institute affiliated with University of Kansas School of Medicine−Wichita.

Fatigue and depression can be viewed as a “vicious cycle”: Fatigue can be a symptom of major depression, and fatigue can be a risk factor for depression.1 For example, fatigue associated with a general medical condition or traumatic brain injury can be a risk factor for developing major depressive disorder (MDD).1-3 It isn’t surprising that fatigue has been studied as a predictor of relapse after previous response to treatment in patients with MDD.

Despite the observed association between fatigue and depression, their underlying relationship often is unclear. The literature does not differentiate among fatigue associated with depression, fatigue as a treatment-emergent adverse effect, and fatigue as a residual symptom of depression that is partially responsive to treatment.4,5 To complicate the situation, many medications used to treat MDD can cause fatigue.

Patients often describe fatigue as (1) feeling tired, exhausted, or drained and (2) lacking energy and motivation. Fatigue can be related to impaired wakefulness but is believed to be a different entity than sleepiness.6 Residual fatigue can affect social, cognitive, emotional, and physical health.

We reviewed the literature about fatigue as a symptom of MDD by conducting a search of Medline, PubMed, and Google Scholar, using keywords depression, fatigue, residual symptoms, and treatment. We chose the papers cited in this article based on our consensus and because these publications represent expert opinion or the highest quality evi­dence available.


Residual fatigue has an effect on prognosis

Fatigue is a common symptom of MDD that persists in 20% to 30% of patients whose symptoms of depression otherwise remit.4,7-9 Several studies have linked residual fatigue with the overall prognosis of MDD.5 Data from a prospective study demonstrate that depressed patients have a higher risk of relapse when they continue to report symp­toms of fatigue after their symptoms of depression have otherwise entered partial remission.10 Another study demonstrated that the severity of residual symptoms of depression is a strong predictor of another major depressive episode.11

In a large-scale study, the prevalence of residual fatigue after adequate treat­ment of MDD in both partial responders and remitters was 84.6%.12 The same study showed that one-third of patients who had been treated for MDD had persistent and clinically significant fatigue, which could suggest a relationship between fatigue and selective serotonin reuptake inhibitors (SSRIs) and other antidepressants.

Another study demonstrated that 64.6% of patients who responded to antidepressant treatment and who had baseline fatigue con­tinued to exhibit symptoms of fatigue after an adequate trial of an antidepressant.13


Neurobiological considerations
Studies have shown that the neuronal circuits that malfunction in fatigue are different from those that malfunction in depression.14 Although the neurobiol­ogy of fatigue has not been determined, decreased neuronal activity in the prefron­tal circuits has been associated with symp­toms of fatigue.15

In addition, evidence from the litera­ture shows a decrease in hormone secre­tion16 and cognitive abilities in patients exhibiting symptoms of fatigue.17 These findings have led some experts to hypoth­esize that symptoms of fatigue associated with depression could be the result of (1) immune dysregulation18 and (2) an inability of available antidepressants to tar­get the underlying biology of the disorder.2

Despite the hypothesis that fatigue asso­ciated with depression might be biologically related to immune dysregulation, some authors continue to point to an imbalance in neurotransmitters—norepinephrine, his­tamine, dopamine, acetylcholine—as being  associated with fatigue.14 For example, a study demonstrated that drugs targeting noradrenergic reuptake inhibition were more effective at preventing a relapse of fatigue compared with serotonergic drugs.19 Another study showed improvement in energy with an increase in the plasma level of desipramine, which affects noradrener­gic neurotransmission.20

Inflammatory cytokines also have been explored in the search for an understand­ing of the etiology of fatigue and depres­sion.21 Physical and mental stress promote the release of cytokines, which activate the immune system by inducing an inflam­matory response; this response has been etiologically linked to depressive disor­ders.22 Furthermore, studies have demon­strated an elevated level of inflammatory cytokines in patients who have MDD— suggesting that MDD is associated with a chronic low level of inflammation that crosses the blood−brain barrier.23


Clinical considerations: A role for rating scales?

Despite the significance of residual fatigue on the quality of life of patients who have MDD, most common rating scales, such as the Hamilton Depression Rating Scale24 and the Montgomery-Åsberg Depression Rating Scale,25 have limited sensitivity for measuring fatigue.26 The Fatigue Associated with Depression (FAsD)27 questionnaire, designed according to FDA guidelines,28 is used to assess fatigue associated with depression. The final version of the FAsD includes 13 items: a 6-item experience sub­scale and a 7-item impact subscale.

Is the FAsD helpful? The experience sub­scale of the FAsD assesses how often the patient experiences different aspects of fatigue (tiredness, exhaustion, lack of energy, physical weakness, and a feeling that everything requires too much effort). The impact subscale of the FAsD assesses the effect of fatigue on daily life.

 

 

The overall FAsD score is calculated by taking the mean of each subscale; a change of 0.67 on the experience subscale and 0.57 on the impact subscale are considered clini­cally meaningful.27 The measurement prop­erties of the questionnaire showed internal consistency, reliability, and validity in test­ing. Researchers note, however, that FAsD does not include items to assess the impact of fatigue on cognition. This means that the FAsD might not distinguish between physi­cal and mental aspects of fatigue.


Treatment

It isn’t surprising that residual depres­sion can increase health care utilization and economic burden, including such indirect costs as lost productivity and wages.29 Despite these impacts, there is a paucity of studies evaluating the relation­ship between residual symptoms, such as fatigue, and work productivity. It has been established that improving a depressed patient’s level of energy correlates with improved performance at work.

Treating fatigue as a residual symp­tom of MDD can be complicated because symptoms of fatigue might be:
   • a discrete symptom of MDD
   • a prodromal symptom of another disorder
   • an adverse effect of an antidepressant.2,30

It is a major clinical problem, there­fore, that antidepressants can alleviate and cause symptoms of fatigue.31 Treatment strategy should focus on identifying anti­depressants that are less likely to cause fatigue (ie, noradrenergic or dopaminergic drugs, or both). Adjunctive treatments to target residual fatigue also can be used.32

There are limited published data on the effective treatment of residual fatigue in patients with MDD. Given the absence of sufficient evidence, agents that promote noradrenergic and dopaminergic neuro­transmission have been the treatment of choice when targeting fatigue in depressed patients.2,14,21,33

The Table34-37 lists poten­tial treatment options often used to treat fatigue associated with depression.


SSRIs.
Treatment with SSRIs has been asso­ciated with a low probability of achiev­ing remission when targeting fatigue as a symptom of MDD.21

One study reported that, after 8 weeks of treatment with an SSRI, treatment-emergent adverse events, such as worsening fatigue and weakness, were observed—along with an overall lack of efficacy in targeting all symptoms of depression.38

Another study demonstrated positive effects when a noradrenergic agent was added to an SSRI in partial responders who continued to complain of residual fatigue.33

However, studies that compared the effects of SSRIs with those of antidepres­sants that have pronoradrenergic effects showed that the 2 mechanisms of action were not significantly different from each other in their ability to resolve residual symptoms of fatigue.21 A limiting factor might be that these studies were retrospec­tive and did not analyze the efficacy of a noradrenergic agent as an adjunct for alle­viating symptoms of fatigue.39

Bupropion. This commonly used medica­tion for fatigue is believed to cause a sig­nificantly lower level of fatigue compared with SSRIs.40 The potential utility of bupro­pion in this area could be a reflection of its mechanism of action—ie, the drug targets both noradrenergic and dopaminergic neurotransmission.41

A study comparing bupropion with SSRIs in targeting somatic symptoms of depression reported a small but statisti­cally significant difference in favor of the bupropion-treated group. However, this finding was confounded by the small effect size and difficulty quantifying somatic symptoms.40

Stimulants and modafinil. Psycho-stimulants have been shown to be effica­cious for depression and fatigue, both as monotherapy and adjunctively.39,42

Modafinil has demonstrated efficacy in open-label trials for improving residual fatigue, but failed to separate from pla­cebo in controlled trials.43 At least 1 other failed study has been published examining modafinil as a treatment for fatigue associ­ated with depression.43

Adjunctive therapy with CNS stimu­lants, such as amphetamine/dextroam­phetamine and methylphenidate, has been used to treat fatigue, with positive results.16 Modafinil and stimulants also could be tried as an augmentation strategy to other antidepressants; such use is off-label and should be attempted only after careful consideration.16

Exercise might be a nonpharmacothera­peutic modality that targets the underly­ing physiology associated with fatigue. Exercise releases endorphins, which can affect overall brain chemistry and which have been theorized to diminish symp­toms of fatigue and depression.44 Consider exercise in addition to treatment with an antidepressant in selected patients.45


To sum up

In general, the literature does not recom­mend one medication as superior to any other for treating fatigue that is a residual symptom of depression. Such hesitation suggests that more empirical studies are needed to determine what is the best and proper management of treating fatigue associated with depression.


Bottom LinE
Fatigue can be a symptom of major depressive disorder (MDD) or a risk factor for depression. Fatigue has been studied as a predictor of relapse after previous response to treatment in patients with MDD. Residual fatigue can affect social, cognitive, emotional, and physical health and can result in increased utilization of health care services. A number of treatment options are available; none has been shown to be superior to the others.

 

 


Related Resources

• Leone SS. A disabling combination: fatigue and depression. Br J Psychiatry. 2010;197(2):86-87.
• Targum SD, Fava M. Fatigue as a residual symptom of de­pression. Innov Clin Neurosci. 2011;8(10):40-43.
• Illiades C. How to fight depression fatigue. Everyday Health. http://www.everydayhealth.com/health-report/major-depression-living-well/fight-depression-fatigue.aspx.
• Kerr M. Depression and fatigue: a vicious cycle. Healthline. http://www.healthline.com/health/depression/fatigue.


Drug Brand Names

Amphetamine/dextroamphetamine • Adderall              
Bupropion • Wellbutrin                                                
Desipramine • Norpramin                                           
Methylphenidate • Ritalin
Modafinil • Provigil
Sertraline • Zoloft
Venlafaxine • Effexor

Disclosures
Dr. Sohail reports no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Macaluso has conducted clinical trials research as principal investigator for the following pharmaceutical manufacturers in the past 12 months: AbbVie, Inc.; Alkermes; AssureRx Health, Inc.; Eisai Co., Ltd.; FORUM Pharmaceuticals, Inc.; Janssen Pharmaceuticals, Inc.; and Naurex Inc. All clinical trial and study contracts were with, and payments were made to, University of Kansas Medical Center Research Institute, Kansas City, Kansas, a research institute affiliated with University of Kansas School of Medicine−Wichita.

References


1. Schönberger M, Herrberg M, Ponsford J. Fatigue as a cause, not a consequence of depression and daytime sleepiness: a cross-lagged analysis. J Head Trauma Rehabil. 2014;29(5):427-431.
2. Demyttenaere K, De Fruyt J, Stahl, SM. The many faces of fatigue in major depressive disorder. Int J Neuropsychopharmacol. 2005;8(1):93-105.
3. Skapinakis P, Lewis G, Mavreas V. Temporal relations between unexplained fatigue and depression: longitudinal data from an international study in primary care. Psychosom Med. 2004;66(3):330-335.
4. Nierenberg AA, Husain MM, Trivedi MH, et al. Residual symptoms after remission of major depressive disorder with citalopram and risk of relapse: a STAR*D report. Psychol Med. 2010;40(1):41-50.
5. Kennedy N, Paykel ES. Residual symptoms at remission from depression: impact on long-term outcome. J Affect Disord. 2004;80(2-3):135-144.
6. Shen J, Barbera J, Shapiro CM. Distinguishing sleepiness and fatigue: focus on definition and measurement. Sleep Med Rev. 2006;10:63-76.
7. Nierenberg AA, Keefe BR, Leslie VC, et al. Residual symptoms in depressed patients who respond acutely to fluoxetine. J Clin Psychiatry. 1999;60(4):221-225.
8. Tylee A, Gastpar M, Lépine JP, et al. DEPRES II (Depression Research in European Society II): a patient survey of the symptoms, disability and current management of depression in the community. DEPRES Steering Committee. Int Clin Psychopharmacol. 1999;14(3):139-151.
9. Marcus SM, Young EA, Kerber KB, et al. Gender differences in depression: findings from the STAR*D study. J Affect Disord. 2005;87(2-3):141-150.
10. Paykel ES, Ramana, R, Cooper Z, et al. Residual symptoms after partial remission: an important outcome in depression. Psychol Med. 1995;25(6):1171-1180.
11. Bockting CL, Spinhoven P, Koeter MW, et al; Depression Evaluation Longitudinal Therapy Assessment Study Group. Prediction of recurrence in recurrent depression and the influence of consecutive episodes on vulnerability for depression: a 2-year prospective study. J Clin Psychiatry. 2006;67(5):747-755.
12. Greco T, Eckert G, Kroenke K. The outcome of physical symptoms with treatment of depression. J Gen Intern Med. 2004;19(8):813-818.
13. McClintock SM, Husain MM, Wisniewski SR, et al. Residual symptoms in depressed outpatients who respond by 50% but do not remit to antidepressant medication. J Clin Psychopharmacol. 2011;31(2):180-186.
14. Stahl SM, Zhang L, Damatarca C, et al. Brain circuits determine destiny in depression: a novel approach to the psychopharmacology of wakefulness, fatigue, and executive dysfunction in major depressive disorder. J Clin Psychiatry. 2003;64(suppl 14):6-17.
15. MacHale SM, Law´rie SM, Cavanagh JT, et al. Cerebral perfusion in chronic fatigue syndrome and depression. Br J Psychiatry. 2000;176:550-556.
16. Paykel ES. Achieving gains beyond response. Acta Psychiatrica Scandinavica Suppl. 2002;(415):12-17.
17. van den Heuvel OA, Groenewegen HJ, Barkhof F, et al. Frontostriatal system in planning complexity: a parametric functional magnetic resonance version of Tower of London task. Neuroimage. 2003;18(2):367-374.
18. Jaremka LM, Fagundes CP, Glaser R, et al. Loneliness predicts pain, depression, and fatigue: understanding the role of immune dysregulation. Psychoneuroendocrinology. 2013;38(8):1310-1317.
19. Delgado PL, Charney DS, Price LH, et al. Serotonin function and the mechanism of antidepressant action. Reversal of antidepressant-induced remission by rapid depletion of plasma tryptophan. Arch Gen Psychiatry. 1990;47(5):411-418.
20. Nelson JC, Mazure C, Quinlan DM, et al. Drug-responsive symptoms in melancholia. Arch Gen Psychiatry. 1984;41(7):663-668.
21. Fava M, Ball S, Nelson, JC, et al. Clinical relevance of fatigue as a residual symptom in major depressive disorder. Depress Anxiety. 2014;31(3):250-257.
22. Anisman H, Merali Z, Poulter MO, et al. Cytokines as a precipitant of depressive illness: animal and human studies. Curr Pharm Des. 2005;11(8):963-972.
23. Simon NM, McNamara K, Chow CW, et al. A detailed examination of cytokine abnormalities in major depressive disorder. Eur Neuropsychopharmacol. 2008;18(3):230-233.
24. Hamilton M. A rating scale for depression. J Neurol Neurosurg Psychiatry. 1960;23:56-62.
25. Montgomery SA, Asberg M. A new depression scale designed to be sensitive to change. Br J Psychiatry. 1979;134:382-389.
26. Matza LS, Phillips GA, Revicki DA, et al. Development and validation of a patient-report measure of fatigue associated with depression. J Affect Disord. 2011;134(1-3):294-303.
27. Matza LS, Wyrwich KW, Phillips GA, et al. The Fatigue Associated with Depression Questionnaire (FAsD): responsiveness and responder definition. Qual Life Res. 2013;22(2):351-360.
28. Guidance for industry. Patient-reported outcome measures: use in medical product development to support labeling claims. Food and Drug Administration. http://www.fda. gov/downloads/Drugs/Guidances/UCM193282.pdf. Published December 2009. Accessed May 7, 2015.
29. Knoth RL, Bolge SC, Kim E, et al. Effect of inadequate response to treatment in patients with depression. Am J Manag Care. 2010;16(8):e188-e196.
30. Fava M. Symptoms of fatigue and cognitive/executive dysfunction in major depressive disorder before and after antidepressant treatment. J Clin Psychiatry. 2003;64(suppl 14):30-34.
31. Chang T, Fava M. The future of psychopharmacology of depression. J Clin Psychiatry. 2010;71(8):971-975.
32. Baldwin DS, Papakostas GI. Symptoms of fatigue and sleepiness in major depressive disorder. J Clin Psychiatry. 2006;67(suppl 6):9-15.
33. Ball SG, Dellva MA, D’Souza D, et al. A double-blind, placebo-controlled study of augmentation with LY2216684 for major depressive disorder patients who are partial responders to selective serotonin reuptake inhibitors [abstract P 05]. Int J Psych Clin Pract. 2010;14(suppl 1):19.
34. Stahl SM. Using secondary binding properties to select a not so elective serotonin reuptake inhibitor. J Clin Psychiatry. 1998;59(12):642-643.
35. Stahl SM. Essential psychopharmacology: neuroscientific basis and practical applications. 2nd ed. New York, NY: Cambridge University Press; 2000.
36. Bymaster FP, Katner JS, Nelson DL, et al. Atomoxetine increases extracellular levels of norepinephrine and dopamine in prefrontal cortex of rat: a potential mechanism for efficacy in attention deficit/hyperactivity disorder. Neuropsychopharmacology. 2002;27(5):699-711.
37. Scammell TE, Estabrooke IV, McCarthy MT, et al. Hypothalamic arousal regions are activated during modafinil-induced wakefulness. J Neurosci. 2000;20(22):8620-8628.
38. Daly EJ, Trivedi MH, Fava M, et al. The relationship between adverse events during selective serotonin reuptake inhibitor treatment for major depressive disorder and nonremission in the suicide assessment methodology study. J Clin Psychopharmacol. 2011;31(1):31-38.
39. Nelson JC. A review of the efficacy of serotonergic and noradrenergic reuptake inhibitors for treatment of major depression. Biol Psychiatry. 1999;46(9):1301-1308.
40. Papakostas GI, Nutt DJ, Hallett LA, et al. Resolution of sleepiness and fatigue in major depressive disorder: a comparison of bupropion and the selective serotonin reuptake inhibitors. Biol Psychiatry. 2006;60(12):1350-1355.
41. Fava M, Rush AJ, Thase ME, et al. 15 years of clinical experience with bupropion HCl: from bupropion to bupropion SR to bupropion XL. Prim Care Companion J Clin Psychiatry. 2005;7(3):106-113.
42. Candy M, Jones CB, Williams R, et al. Psychostimulants for depression. Cochrane Database Syst Rev. 2008;(2):CD006722. doi: 10.1002/14651858.CD006722.pub2.
43. Lam JY, Freeman MK, Cates ME. Modafinil augmentation for residual symptoms of fatigue in patients with a partial response to antidepressants. Ann Pharmacother. 2007;41(6):1005-1012.
44. Salmon P. Effects of physical exercise on anxiety, depression, and sensitivity to stress: a unifying theory. Clinical Psychol Rev. 2001;21(1):33-61.
45. Trivedi MH, Greer TL, Grannemann BD, et al. Exercise as an augmentation strategy for treatment of major depression. J Psychiatr Pract. 2006;12(4):205-213.

References


1. Schönberger M, Herrberg M, Ponsford J. Fatigue as a cause, not a consequence of depression and daytime sleepiness: a cross-lagged analysis. J Head Trauma Rehabil. 2014;29(5):427-431.
2. Demyttenaere K, De Fruyt J, Stahl, SM. The many faces of fatigue in major depressive disorder. Int J Neuropsychopharmacol. 2005;8(1):93-105.
3. Skapinakis P, Lewis G, Mavreas V. Temporal relations between unexplained fatigue and depression: longitudinal data from an international study in primary care. Psychosom Med. 2004;66(3):330-335.
4. Nierenberg AA, Husain MM, Trivedi MH, et al. Residual symptoms after remission of major depressive disorder with citalopram and risk of relapse: a STAR*D report. Psychol Med. 2010;40(1):41-50.
5. Kennedy N, Paykel ES. Residual symptoms at remission from depression: impact on long-term outcome. J Affect Disord. 2004;80(2-3):135-144.
6. Shen J, Barbera J, Shapiro CM. Distinguishing sleepiness and fatigue: focus on definition and measurement. Sleep Med Rev. 2006;10:63-76.
7. Nierenberg AA, Keefe BR, Leslie VC, et al. Residual symptoms in depressed patients who respond acutely to fluoxetine. J Clin Psychiatry. 1999;60(4):221-225.
8. Tylee A, Gastpar M, Lépine JP, et al. DEPRES II (Depression Research in European Society II): a patient survey of the symptoms, disability and current management of depression in the community. DEPRES Steering Committee. Int Clin Psychopharmacol. 1999;14(3):139-151.
9. Marcus SM, Young EA, Kerber KB, et al. Gender differences in depression: findings from the STAR*D study. J Affect Disord. 2005;87(2-3):141-150.
10. Paykel ES, Ramana, R, Cooper Z, et al. Residual symptoms after partial remission: an important outcome in depression. Psychol Med. 1995;25(6):1171-1180.
11. Bockting CL, Spinhoven P, Koeter MW, et al; Depression Evaluation Longitudinal Therapy Assessment Study Group. Prediction of recurrence in recurrent depression and the influence of consecutive episodes on vulnerability for depression: a 2-year prospective study. J Clin Psychiatry. 2006;67(5):747-755.
12. Greco T, Eckert G, Kroenke K. The outcome of physical symptoms with treatment of depression. J Gen Intern Med. 2004;19(8):813-818.
13. McClintock SM, Husain MM, Wisniewski SR, et al. Residual symptoms in depressed outpatients who respond by 50% but do not remit to antidepressant medication. J Clin Psychopharmacol. 2011;31(2):180-186.
14. Stahl SM, Zhang L, Damatarca C, et al. Brain circuits determine destiny in depression: a novel approach to the psychopharmacology of wakefulness, fatigue, and executive dysfunction in major depressive disorder. J Clin Psychiatry. 2003;64(suppl 14):6-17.
15. MacHale SM, Law´rie SM, Cavanagh JT, et al. Cerebral perfusion in chronic fatigue syndrome and depression. Br J Psychiatry. 2000;176:550-556.
16. Paykel ES. Achieving gains beyond response. Acta Psychiatrica Scandinavica Suppl. 2002;(415):12-17.
17. van den Heuvel OA, Groenewegen HJ, Barkhof F, et al. Frontostriatal system in planning complexity: a parametric functional magnetic resonance version of Tower of London task. Neuroimage. 2003;18(2):367-374.
18. Jaremka LM, Fagundes CP, Glaser R, et al. Loneliness predicts pain, depression, and fatigue: understanding the role of immune dysregulation. Psychoneuroendocrinology. 2013;38(8):1310-1317.
19. Delgado PL, Charney DS, Price LH, et al. Serotonin function and the mechanism of antidepressant action. Reversal of antidepressant-induced remission by rapid depletion of plasma tryptophan. Arch Gen Psychiatry. 1990;47(5):411-418.
20. Nelson JC, Mazure C, Quinlan DM, et al. Drug-responsive symptoms in melancholia. Arch Gen Psychiatry. 1984;41(7):663-668.
21. Fava M, Ball S, Nelson, JC, et al. Clinical relevance of fatigue as a residual symptom in major depressive disorder. Depress Anxiety. 2014;31(3):250-257.
22. Anisman H, Merali Z, Poulter MO, et al. Cytokines as a precipitant of depressive illness: animal and human studies. Curr Pharm Des. 2005;11(8):963-972.
23. Simon NM, McNamara K, Chow CW, et al. A detailed examination of cytokine abnormalities in major depressive disorder. Eur Neuropsychopharmacol. 2008;18(3):230-233.
24. Hamilton M. A rating scale for depression. J Neurol Neurosurg Psychiatry. 1960;23:56-62.
25. Montgomery SA, Asberg M. A new depression scale designed to be sensitive to change. Br J Psychiatry. 1979;134:382-389.
26. Matza LS, Phillips GA, Revicki DA, et al. Development and validation of a patient-report measure of fatigue associated with depression. J Affect Disord. 2011;134(1-3):294-303.
27. Matza LS, Wyrwich KW, Phillips GA, et al. The Fatigue Associated with Depression Questionnaire (FAsD): responsiveness and responder definition. Qual Life Res. 2013;22(2):351-360.
28. Guidance for industry. Patient-reported outcome measures: use in medical product development to support labeling claims. Food and Drug Administration. http://www.fda. gov/downloads/Drugs/Guidances/UCM193282.pdf. Published December 2009. Accessed May 7, 2015.
29. Knoth RL, Bolge SC, Kim E, et al. Effect of inadequate response to treatment in patients with depression. Am J Manag Care. 2010;16(8):e188-e196.
30. Fava M. Symptoms of fatigue and cognitive/executive dysfunction in major depressive disorder before and after antidepressant treatment. J Clin Psychiatry. 2003;64(suppl 14):30-34.
31. Chang T, Fava M. The future of psychopharmacology of depression. J Clin Psychiatry. 2010;71(8):971-975.
32. Baldwin DS, Papakostas GI. Symptoms of fatigue and sleepiness in major depressive disorder. J Clin Psychiatry. 2006;67(suppl 6):9-15.
33. Ball SG, Dellva MA, D’Souza D, et al. A double-blind, placebo-controlled study of augmentation with LY2216684 for major depressive disorder patients who are partial responders to selective serotonin reuptake inhibitors [abstract P 05]. Int J Psych Clin Pract. 2010;14(suppl 1):19.
34. Stahl SM. Using secondary binding properties to select a not so elective serotonin reuptake inhibitor. J Clin Psychiatry. 1998;59(12):642-643.
35. Stahl SM. Essential psychopharmacology: neuroscientific basis and practical applications. 2nd ed. New York, NY: Cambridge University Press; 2000.
36. Bymaster FP, Katner JS, Nelson DL, et al. Atomoxetine increases extracellular levels of norepinephrine and dopamine in prefrontal cortex of rat: a potential mechanism for efficacy in attention deficit/hyperactivity disorder. Neuropsychopharmacology. 2002;27(5):699-711.
37. Scammell TE, Estabrooke IV, McCarthy MT, et al. Hypothalamic arousal regions are activated during modafinil-induced wakefulness. J Neurosci. 2000;20(22):8620-8628.
38. Daly EJ, Trivedi MH, Fava M, et al. The relationship between adverse events during selective serotonin reuptake inhibitor treatment for major depressive disorder and nonremission in the suicide assessment methodology study. J Clin Psychopharmacol. 2011;31(1):31-38.
39. Nelson JC. A review of the efficacy of serotonergic and noradrenergic reuptake inhibitors for treatment of major depression. Biol Psychiatry. 1999;46(9):1301-1308.
40. Papakostas GI, Nutt DJ, Hallett LA, et al. Resolution of sleepiness and fatigue in major depressive disorder: a comparison of bupropion and the selective serotonin reuptake inhibitors. Biol Psychiatry. 2006;60(12):1350-1355.
41. Fava M, Rush AJ, Thase ME, et al. 15 years of clinical experience with bupropion HCl: from bupropion to bupropion SR to bupropion XL. Prim Care Companion J Clin Psychiatry. 2005;7(3):106-113.
42. Candy M, Jones CB, Williams R, et al. Psychostimulants for depression. Cochrane Database Syst Rev. 2008;(2):CD006722. doi: 10.1002/14651858.CD006722.pub2.
43. Lam JY, Freeman MK, Cates ME. Modafinil augmentation for residual symptoms of fatigue in patients with a partial response to antidepressants. Ann Pharmacother. 2007;41(6):1005-1012.
44. Salmon P. Effects of physical exercise on anxiety, depression, and sensitivity to stress: a unifying theory. Clinical Psychol Rev. 2001;21(1):33-61.
45. Trivedi MH, Greer TL, Grannemann BD, et al. Exercise as an augmentation strategy for treatment of major depression. J Psychiatr Pract. 2006;12(4):205-213.

Issue
Current Psychiatry - 14(6)
Issue
Current Psychiatry - 14(6)
Page Number
16-17, 28-32
Page Number
16-17, 28-32
Publications
Publications
Topics
Article Type
Display Headline
Fatigue after depression responds to therapy. What are the next steps?
Display Headline
Fatigue after depression responds to therapy. What are the next steps?
Legacy Keywords
fatigue associated with depression, depression, depressive disorders, fatigue and depression, depression caused by fatigue, fatigue caused by depression,
Legacy Keywords
fatigue associated with depression, depression, depressive disorders, fatigue and depression, depression caused by fatigue, fatigue caused by depression,
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Levomilnacipran for the treatment of major depressive disorder

Article Type
Changed
Tue, 12/11/2018 - 15:36
Display Headline
Levomilnacipran for the treatment of major depressive disorder

In July 2013, the FDA approved levomilnacipran for the treatment of major depressive disorder (MDD) in adults.1 It is available in a once-daily, extended-release formulation (Table 1).1 The drug is the fifth serotonin-norepinephrine reuptake inhibitor (SNRI) to be sold in the United States and the fourth to receive FDA approval for treating MDD.

Levomilnacipran is believed to be the more active enantiomer of milnacipran, which has been available in Europe for years and was approved by the FDA in 2009 for treating fibromyalgia. Efficacy of levomilnacipran for treating patients with MDD was established in three 8-week randomized controlled trials (RCTs).1

Clinical implications

Levomilnacipran is indicated for treating MDD in adults and is unique compared with other SNRIs because it is relatively more selective for norepinephrine reuptake inhibition (NRI) compared with serotonin reuptake inhibition (SRI).1 In vitro studies demonstrate that the drug has >10-fold greater selectivity for norepinephrine reuptake inhibition than it does for serotonin reuptake inhibition, compared with duloxetine or venlafaxine.2

This difference in selectivity could lend itself to treating symptoms of MDD that might be related to norepinephrine deficiency; these include decreased concentration, lassitude, mental and physical slowing, and decreased self-care.3,4 Some authors claim that individual patients could experience improvement in their social and occupational functioning in addition to improvement in the core symptoms of depression.5

Levomilnacipran is the more active enantiomer of milnacipran, an SNRI that is approved for treating fibromyalgia in the United States and approved for treating depression in many other countries.6 In general, enantiomeric formulations are believed to have advantages over racemic formulations because they are less complex and have a more selective pharmacodynamic profile, better therapeutic index, lower liability for drug-drug interactions (DDIs), and a less complicated relationship between plasma concentration and pharmacodynamic effect.6 In addition, regulatory guidelines in the United States recommend development of enantiomers over racemates where appropriate.7

How it works

Levomilnacipran’s exact mechanism of action is unknown. Similar to other SNRIs, it binds with high affinity to the serotonin (5-HT) and norepinephrine (NE) transporters and potently inhibits 5-HT and NE reuptake. Levomilnacipran lacks significant affinity for any other receptors, ion channels, or transporters tested in vitro.2 It differs from other SNRIs such as venlafaxine and duloxetine in having higher selectivity for norepinephrine vs serotonin reuptake inhibition. In vitro studies demonstrated a 2-fold preference for NE over 5-HT reuptake inhibition.2

Pharmacokinetics

Levomilnacipran reaches maximum plasma concentration within 6 to 8 hours of oral administration and has a half-life of approximately 12 hours, which makes it suitable for once-daily dosing. The concentration of levomilnacipran at steady state is proportional to the dosage of the drug when administered within the range of 25 to 300 mg once daily.1

The drug’s mean apparent total clearance is 21 to 29 liters/hour and its bioavailability is not significantly affected when taken with food. The drug is widely distributed in the body and is converted primarily to 2 metabolites: desethy levomilnacipran and p-hydroxy-levomilnacipran. Both metabolites are inactive and undergo further conjugation with glucuronide. The drug is eliminated primarily via renal excretion.1

The major enzyme that catalyzes metabolism of levomilnacipram is cytochrome P 450 (CYP) 3A4, which makes it susceptible to DDIs with drugs that inhibit or induce this enzyme. For example, a person taking levomilnacipran with a potent CYP3A4 inhibitor, such as ketoconazole, may require a dosage adjustment. No dosage adjustment is needed when given with a CYP3A4 inducer or substrate. Drinking alcohol with levomilnacipran may cause more rapid release of the drug into the blood stream.1

Efficacy

Levomilnacipran decreased core symptoms of MDD and showed a statistically significant separation from placebo in 2 phase III RCTs (Table 2).3,5 The first study was a 10-week flexible dose (75 or 100 mg) trial in 563 outpatients age 18 to 70 who met DSM-IV-TR criteria of MDD for >1 month and had a 17-item Hamilton Depression Rating Scale (HDRS-17) score >22 and a Sheehan Disability Scale (SDS) score >10.3 The primary efficacy measure was change in Montgomery-Åsberg Depression Rating Scale (MADRS) score from baseline to week 10. Secondary efficacy measures included the HDRS-17, SDS, and Clinical Global Impressions-improvement scale. Efficacy analyses included 276 subjects treated with levomilnacipran and 277 treated with placebo.3

Levomilnacipran was significantly superior to placebo on the MADRS and HDRS-17 from baseline to week 10. Response and remission rates were
significantly greater for the levomilnacipran group compared with pla
cebo. Response exceeded the 10% average advantage for drug vs placebo and 46% of levomilnacipran-treated patients achieved remission.3

The number needed to treat (NNT), based on the MADRS scores for the levomilnacipran group compared with the placebo group, was 6 for response and 5 for remission.3 By comparison, most studies of venlafaxine demonstrate a NNT of 8.3

 

 

Levomilnacipran generally was reported to be safe and well tolerated. The most common adverse events leading to discontinuation in the levomilnacipran group were nausea, vomiting, change in systolic and diastolic blood pressure, and increase in heart rate. The favorable tolerability profile of levomilnacipran may relate to the 2-fold greater potency for NE reuptake inhibition compared with 5-HT reuptake inhibition.3

The second study was an 11-week, fixed-dose trial of levomilnacipran using 40, 80, or 120 mg. A total of 724 outpatients age 18 to 65 who met DSM-IV-TR criteria for MDD and who had an ongoing episode of depression lasting >8 weeks were randomly assigned to receive placebo (n = 179) or levomilnacipran at 40 mg (n = 181), 80 mg (n = 181), or 120 mg (n = 183) once daily for 8 weeks of double-blind treatment followed by a 2-week, double-blind taper of the drug.5 The primary efficacy parameter was change from baseline on the MADRS and the secondary efficacy parameter was change from baseline in SDS total score. HDRS-17, CGI-I, and CGI-S were included as secondary outcome measures.5

Significant difference in MADRS total score were seen in the levomilnacipran group compared with the placebo group (least mean squared difference: 40 mg/d,  −3.23; 80 mg/d, −3.99; and 120 mg/d,  −4.86). Higher dosages produced a numerically greater change and significant separation from placebo occurred sooner in the 80-mg and 120-mg groups compared with the 40-mg group.5

Significant differences vs placebo were consistently observed across secondary outcome measures for the higher dosages of levomilnacipran, and improvement in SDS total score was noted in all levomilnacipran groups compared with the placebo group. When dosed at 120 mg/d, levomilnacipran produced significant improvement vs placebo on all SDS subscales and was as well tolerated as the 80 mg dosage.5

No new safety concerns were observed in the study. A dose-response relationship in tolerability was not demonstrated and the number of patients reporting adverse events and who discontinued participation because of adverse events was higher in the 80-mg group than in the 40-mg and 120-mg groups.5

Tolerability

Overall, levomilnacipran was well tolerated in clinical trials, during which 2,673 subjects were exposed to the drug—translating to 942 patient-years of exposure. These patients ranged in age from 18 to 78; 825 of these subjects were enrolled in long-term studies for 1 year. Dosing of levomilnacipran during these studies ranged from 40 to 120 mg once daily, without regard to food.1

Nine percent of patients who received levomilnacipran in short-term studies discontinued because of adverse events, compared with 3% of patients who received placebo. The most common adverse event reported was nausea; other common adverse events reported included constipation, hyperhidrosis, elevated heart rate, erectile dysfunction, tachycardia, palpitations, and vomiting. Of these events, only erectile dysfunction and urinary hesitation were dose-related.1 Among levo­milnacipran-treated female patients, <2% reported adverse events related to sexual dysfunction.

All SNRIs have well established associations with elevation in blood pressure and heart rate. Levomilnacipran resulted in a mean increase of 3 mm HG in systolic and 3.2 mm Hg in diastolic blood pressure in short-term, placebo-controlled trials.1

Orthostatic hypotension was observed in 11.6% of patients in the levomilnacipran groups, compared with 9.7% in placebo groups in all short-term studies. Orthostatic reductions of blood pressure occurred in 5.8%, 6.1%, and 9.8% of levomilnacipran-treated patients with dosages of 40, 80, and 120 mg/d respectively, indicating a dose-dependent adverse event. A mean increase in heart rate of 7.2 beats per minute (bpm) also was seen in short-term studies in the levomilnacipran-treated group compared with 0.3 bpm in the placebo-treated group.1 Clinicians should monitor blood pressure and heart rate routinely because of potential increases seen in some subjects in these studies, which excluded those who had significant cardiovascular disease.

Unique clinical issues

Both in-vitro and in-vivo studies found that levomilnacipran exhibited more potency for NE reuptake inhibition than for 5-HT reuptake inhibition at the lowest effective dosage (10 mg/kg). However as the dosage was increased (20 mg/kg and 40 mg/kg), it was equally potent at NE and 5-HT reuptake inhibition. This is in contrast to venlafaxine, which demonstrates a similar, but opposite, effect in terms of potentiation at the 5-HT and NE reuptake pumps.2

The greater noradrenergic effect of levomilnacipran could lend itself to treating certain subgroups of patients whose symptoms are believed to be related to deficiencies in NE (eg, lassitude).4 This concept is theoretical, and was not explicitly studied in clinical trials and the drug is not labeled in this way.

Contraindications

Contraindications to levomilnacipran are similar to those seen with SSRIs and SNRIs, including concomitant use of a monoamine oxidase inhibitor (MAOI) and the use of the levomilnacipran within 14 days of stopping an MAOI. Contraindications unique to levomilnacipran include hypersensitivity to levomilnacipran, milnacipran, or any component specific to the formulation; and uncontrolled narrow-angle glaucoma.1

 

 

Dosing

The recommended dosage range of levomilnacipran is 40 to 120 mg once
daily with or without food. The capsules should be swallowed whole and should not be opened or crushed. As with most psychotropics, levomilnacipran should be taken at approximately the same time each day.1

The manufacturer recommends an initial dose of levomilnacipran of 20 mg once daily for 2 days, increased to 40 mg once daily. Based on efficacy and tolerably, levomilnacipran can be increased in increments of 40 mg every 2 days.

Dosage adjustment is recommended for patients with moderate or severe renal impairment; and the maintenance dosage should not exceed 80 mg and 40 mg respectively in these populations. As with many antidepressants, gradual dosage reduction is recommended to avoid discontinuation symptoms.

Bottom Line

Levomilnacipran is FDA-approved for treating major depressive disorder in adults. In 2 randomized controlled trials, the drug showed a significant separation from placebo. Levomilnacipran generally was reported to be safe and well tolerated; common adverse events were nausea, vomiting, changes in blood pressure, and an increase in heart rate.

Related Resources

 

  • Citrome L. Levomilnacipran for major depressive disorder: a systematic review of the efficacy and safety profile for this newly approved antidepressant - what is the number needed to treat, number needed to harm and likelihood to be helped or harmed? [published online September 8, 2013]. Int J Clin Pract. doi: 10.1111/ijcp.12298.
  • Mago R, Forero G, Greenberg WM, et al. Safety and tolerability of levomilnacipran ER in major depressive disorder: results from an open-label, 48-week extension study. Clin Drug Investig. 2013;33(10):761-771.


Drug Brand Names
Duloxetine • Cymbalta        Milnacipran • Savella
Ketoconazole • Nizoral        Venlafaxine • Effexor    Levomilnacipran • Fetzima
 

Disclosures

Dr. Macaluso has been the principal investigator for clinical trials for AbbVie, Eisai, Envivo, Janssen, Naurex, and Pfizer. All clinical trial and study contracts and payments were made through the Kansas University Medical Center Research Institute. Drs. Kazanchi and Malhotra report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

 

1. Fetzima [package insert]. St. Louis, MO: Forest Laboratories; 2013.

2. Auclair AL, Martel JC, Assié MB, et al. Levomilnacipran (F2695), a norepinephrine-preferring SNRI: profile in vitro and in models of depression and anxiety. Neuropharmacology. 2013;70:338-347.

3. Montgomery SA, Mansuy L, Ruth A, et al. Efficacy and Safety of levomilnacipran sustained release in moderate to severe major depressive disorder: a randomized, double-blind, placebo-controlled, proof-of-concept study. J Clin Psychiatry. 2013;74(4):363-369.

4. Kasper S, Meshkat D, Kutzelnigg A. Improvement of the noradrenergic symptom cluster following treatment with milnacipran. Neuropsychiatric Dis Treat. 2011; 7(suppl 1):21-27.

5. Asnis GM, Bose A, Gommoll CP, et al. Efficacy and safety of levomilnacipran sustained release 40 mg, 80 mg, or 120 mg in major depressive disorder: a phase 3, randomized, double-blind, placebo-controlled study. J Clin Psychiatry. 2013;74(3):242-248.

6. Hutt AJ, Vanetová J. The chiral switch: the development of single enantiomer drugs from racemates. Acta Facultatis Pharmaceuticae Universitatis Comenianae. 2003; 50(7):23.

7. U.S. Food and Drug Administration. Development of new stereoisomeric drugs. Published May 1, 1992. http://www.fda.gov/drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm122883.htm#.UKHEWm4ZyYE.email. Accessed October 8, 2013.

Article PDF
Author and Disclosure Information

 

Matthew Macaluso, DO
Assistant Professor
Director of Research
Associate Director of Residency Training


Hala Kazanchi, MD

Senior Resident

Vikram Malhotra, MD
Senior Resident

University of Kansas School of Medicine-Wichita
Wichita, Kansas

Issue
Current Psychiatry - 12(12)
Publications
Topics
Page Number
50-52, 54-55
Sections
Author and Disclosure Information

 

Matthew Macaluso, DO
Assistant Professor
Director of Research
Associate Director of Residency Training


Hala Kazanchi, MD

Senior Resident

Vikram Malhotra, MD
Senior Resident

University of Kansas School of Medicine-Wichita
Wichita, Kansas

Author and Disclosure Information

 

Matthew Macaluso, DO
Assistant Professor
Director of Research
Associate Director of Residency Training


Hala Kazanchi, MD

Senior Resident

Vikram Malhotra, MD
Senior Resident

University of Kansas School of Medicine-Wichita
Wichita, Kansas

Article PDF
Article PDF

In July 2013, the FDA approved levomilnacipran for the treatment of major depressive disorder (MDD) in adults.1 It is available in a once-daily, extended-release formulation (Table 1).1 The drug is the fifth serotonin-norepinephrine reuptake inhibitor (SNRI) to be sold in the United States and the fourth to receive FDA approval for treating MDD.

Levomilnacipran is believed to be the more active enantiomer of milnacipran, which has been available in Europe for years and was approved by the FDA in 2009 for treating fibromyalgia. Efficacy of levomilnacipran for treating patients with MDD was established in three 8-week randomized controlled trials (RCTs).1

Clinical implications

Levomilnacipran is indicated for treating MDD in adults and is unique compared with other SNRIs because it is relatively more selective for norepinephrine reuptake inhibition (NRI) compared with serotonin reuptake inhibition (SRI).1 In vitro studies demonstrate that the drug has >10-fold greater selectivity for norepinephrine reuptake inhibition than it does for serotonin reuptake inhibition, compared with duloxetine or venlafaxine.2

This difference in selectivity could lend itself to treating symptoms of MDD that might be related to norepinephrine deficiency; these include decreased concentration, lassitude, mental and physical slowing, and decreased self-care.3,4 Some authors claim that individual patients could experience improvement in their social and occupational functioning in addition to improvement in the core symptoms of depression.5

Levomilnacipran is the more active enantiomer of milnacipran, an SNRI that is approved for treating fibromyalgia in the United States and approved for treating depression in many other countries.6 In general, enantiomeric formulations are believed to have advantages over racemic formulations because they are less complex and have a more selective pharmacodynamic profile, better therapeutic index, lower liability for drug-drug interactions (DDIs), and a less complicated relationship between plasma concentration and pharmacodynamic effect.6 In addition, regulatory guidelines in the United States recommend development of enantiomers over racemates where appropriate.7

How it works

Levomilnacipran’s exact mechanism of action is unknown. Similar to other SNRIs, it binds with high affinity to the serotonin (5-HT) and norepinephrine (NE) transporters and potently inhibits 5-HT and NE reuptake. Levomilnacipran lacks significant affinity for any other receptors, ion channels, or transporters tested in vitro.2 It differs from other SNRIs such as venlafaxine and duloxetine in having higher selectivity for norepinephrine vs serotonin reuptake inhibition. In vitro studies demonstrated a 2-fold preference for NE over 5-HT reuptake inhibition.2

Pharmacokinetics

Levomilnacipran reaches maximum plasma concentration within 6 to 8 hours of oral administration and has a half-life of approximately 12 hours, which makes it suitable for once-daily dosing. The concentration of levomilnacipran at steady state is proportional to the dosage of the drug when administered within the range of 25 to 300 mg once daily.1

The drug’s mean apparent total clearance is 21 to 29 liters/hour and its bioavailability is not significantly affected when taken with food. The drug is widely distributed in the body and is converted primarily to 2 metabolites: desethy levomilnacipran and p-hydroxy-levomilnacipran. Both metabolites are inactive and undergo further conjugation with glucuronide. The drug is eliminated primarily via renal excretion.1

The major enzyme that catalyzes metabolism of levomilnacipram is cytochrome P 450 (CYP) 3A4, which makes it susceptible to DDIs with drugs that inhibit or induce this enzyme. For example, a person taking levomilnacipran with a potent CYP3A4 inhibitor, such as ketoconazole, may require a dosage adjustment. No dosage adjustment is needed when given with a CYP3A4 inducer or substrate. Drinking alcohol with levomilnacipran may cause more rapid release of the drug into the blood stream.1

Efficacy

Levomilnacipran decreased core symptoms of MDD and showed a statistically significant separation from placebo in 2 phase III RCTs (Table 2).3,5 The first study was a 10-week flexible dose (75 or 100 mg) trial in 563 outpatients age 18 to 70 who met DSM-IV-TR criteria of MDD for >1 month and had a 17-item Hamilton Depression Rating Scale (HDRS-17) score >22 and a Sheehan Disability Scale (SDS) score >10.3 The primary efficacy measure was change in Montgomery-Åsberg Depression Rating Scale (MADRS) score from baseline to week 10. Secondary efficacy measures included the HDRS-17, SDS, and Clinical Global Impressions-improvement scale. Efficacy analyses included 276 subjects treated with levomilnacipran and 277 treated with placebo.3

Levomilnacipran was significantly superior to placebo on the MADRS and HDRS-17 from baseline to week 10. Response and remission rates were
significantly greater for the levomilnacipran group compared with pla
cebo. Response exceeded the 10% average advantage for drug vs placebo and 46% of levomilnacipran-treated patients achieved remission.3

The number needed to treat (NNT), based on the MADRS scores for the levomilnacipran group compared with the placebo group, was 6 for response and 5 for remission.3 By comparison, most studies of venlafaxine demonstrate a NNT of 8.3

 

 

Levomilnacipran generally was reported to be safe and well tolerated. The most common adverse events leading to discontinuation in the levomilnacipran group were nausea, vomiting, change in systolic and diastolic blood pressure, and increase in heart rate. The favorable tolerability profile of levomilnacipran may relate to the 2-fold greater potency for NE reuptake inhibition compared with 5-HT reuptake inhibition.3

The second study was an 11-week, fixed-dose trial of levomilnacipran using 40, 80, or 120 mg. A total of 724 outpatients age 18 to 65 who met DSM-IV-TR criteria for MDD and who had an ongoing episode of depression lasting >8 weeks were randomly assigned to receive placebo (n = 179) or levomilnacipran at 40 mg (n = 181), 80 mg (n = 181), or 120 mg (n = 183) once daily for 8 weeks of double-blind treatment followed by a 2-week, double-blind taper of the drug.5 The primary efficacy parameter was change from baseline on the MADRS and the secondary efficacy parameter was change from baseline in SDS total score. HDRS-17, CGI-I, and CGI-S were included as secondary outcome measures.5

Significant difference in MADRS total score were seen in the levomilnacipran group compared with the placebo group (least mean squared difference: 40 mg/d,  −3.23; 80 mg/d, −3.99; and 120 mg/d,  −4.86). Higher dosages produced a numerically greater change and significant separation from placebo occurred sooner in the 80-mg and 120-mg groups compared with the 40-mg group.5

Significant differences vs placebo were consistently observed across secondary outcome measures for the higher dosages of levomilnacipran, and improvement in SDS total score was noted in all levomilnacipran groups compared with the placebo group. When dosed at 120 mg/d, levomilnacipran produced significant improvement vs placebo on all SDS subscales and was as well tolerated as the 80 mg dosage.5

No new safety concerns were observed in the study. A dose-response relationship in tolerability was not demonstrated and the number of patients reporting adverse events and who discontinued participation because of adverse events was higher in the 80-mg group than in the 40-mg and 120-mg groups.5

Tolerability

Overall, levomilnacipran was well tolerated in clinical trials, during which 2,673 subjects were exposed to the drug—translating to 942 patient-years of exposure. These patients ranged in age from 18 to 78; 825 of these subjects were enrolled in long-term studies for 1 year. Dosing of levomilnacipran during these studies ranged from 40 to 120 mg once daily, without regard to food.1

Nine percent of patients who received levomilnacipran in short-term studies discontinued because of adverse events, compared with 3% of patients who received placebo. The most common adverse event reported was nausea; other common adverse events reported included constipation, hyperhidrosis, elevated heart rate, erectile dysfunction, tachycardia, palpitations, and vomiting. Of these events, only erectile dysfunction and urinary hesitation were dose-related.1 Among levo­milnacipran-treated female patients, <2% reported adverse events related to sexual dysfunction.

All SNRIs have well established associations with elevation in blood pressure and heart rate. Levomilnacipran resulted in a mean increase of 3 mm HG in systolic and 3.2 mm Hg in diastolic blood pressure in short-term, placebo-controlled trials.1

Orthostatic hypotension was observed in 11.6% of patients in the levomilnacipran groups, compared with 9.7% in placebo groups in all short-term studies. Orthostatic reductions of blood pressure occurred in 5.8%, 6.1%, and 9.8% of levomilnacipran-treated patients with dosages of 40, 80, and 120 mg/d respectively, indicating a dose-dependent adverse event. A mean increase in heart rate of 7.2 beats per minute (bpm) also was seen in short-term studies in the levomilnacipran-treated group compared with 0.3 bpm in the placebo-treated group.1 Clinicians should monitor blood pressure and heart rate routinely because of potential increases seen in some subjects in these studies, which excluded those who had significant cardiovascular disease.

Unique clinical issues

Both in-vitro and in-vivo studies found that levomilnacipran exhibited more potency for NE reuptake inhibition than for 5-HT reuptake inhibition at the lowest effective dosage (10 mg/kg). However as the dosage was increased (20 mg/kg and 40 mg/kg), it was equally potent at NE and 5-HT reuptake inhibition. This is in contrast to venlafaxine, which demonstrates a similar, but opposite, effect in terms of potentiation at the 5-HT and NE reuptake pumps.2

The greater noradrenergic effect of levomilnacipran could lend itself to treating certain subgroups of patients whose symptoms are believed to be related to deficiencies in NE (eg, lassitude).4 This concept is theoretical, and was not explicitly studied in clinical trials and the drug is not labeled in this way.

Contraindications

Contraindications to levomilnacipran are similar to those seen with SSRIs and SNRIs, including concomitant use of a monoamine oxidase inhibitor (MAOI) and the use of the levomilnacipran within 14 days of stopping an MAOI. Contraindications unique to levomilnacipran include hypersensitivity to levomilnacipran, milnacipran, or any component specific to the formulation; and uncontrolled narrow-angle glaucoma.1

 

 

Dosing

The recommended dosage range of levomilnacipran is 40 to 120 mg once
daily with or without food. The capsules should be swallowed whole and should not be opened or crushed. As with most psychotropics, levomilnacipran should be taken at approximately the same time each day.1

The manufacturer recommends an initial dose of levomilnacipran of 20 mg once daily for 2 days, increased to 40 mg once daily. Based on efficacy and tolerably, levomilnacipran can be increased in increments of 40 mg every 2 days.

Dosage adjustment is recommended for patients with moderate or severe renal impairment; and the maintenance dosage should not exceed 80 mg and 40 mg respectively in these populations. As with many antidepressants, gradual dosage reduction is recommended to avoid discontinuation symptoms.

Bottom Line

Levomilnacipran is FDA-approved for treating major depressive disorder in adults. In 2 randomized controlled trials, the drug showed a significant separation from placebo. Levomilnacipran generally was reported to be safe and well tolerated; common adverse events were nausea, vomiting, changes in blood pressure, and an increase in heart rate.

Related Resources

 

  • Citrome L. Levomilnacipran for major depressive disorder: a systematic review of the efficacy and safety profile for this newly approved antidepressant - what is the number needed to treat, number needed to harm and likelihood to be helped or harmed? [published online September 8, 2013]. Int J Clin Pract. doi: 10.1111/ijcp.12298.
  • Mago R, Forero G, Greenberg WM, et al. Safety and tolerability of levomilnacipran ER in major depressive disorder: results from an open-label, 48-week extension study. Clin Drug Investig. 2013;33(10):761-771.


Drug Brand Names
Duloxetine • Cymbalta        Milnacipran • Savella
Ketoconazole • Nizoral        Venlafaxine • Effexor    Levomilnacipran • Fetzima
 

Disclosures

Dr. Macaluso has been the principal investigator for clinical trials for AbbVie, Eisai, Envivo, Janssen, Naurex, and Pfizer. All clinical trial and study contracts and payments were made through the Kansas University Medical Center Research Institute. Drs. Kazanchi and Malhotra report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

In July 2013, the FDA approved levomilnacipran for the treatment of major depressive disorder (MDD) in adults.1 It is available in a once-daily, extended-release formulation (Table 1).1 The drug is the fifth serotonin-norepinephrine reuptake inhibitor (SNRI) to be sold in the United States and the fourth to receive FDA approval for treating MDD.

Levomilnacipran is believed to be the more active enantiomer of milnacipran, which has been available in Europe for years and was approved by the FDA in 2009 for treating fibromyalgia. Efficacy of levomilnacipran for treating patients with MDD was established in three 8-week randomized controlled trials (RCTs).1

Clinical implications

Levomilnacipran is indicated for treating MDD in adults and is unique compared with other SNRIs because it is relatively more selective for norepinephrine reuptake inhibition (NRI) compared with serotonin reuptake inhibition (SRI).1 In vitro studies demonstrate that the drug has >10-fold greater selectivity for norepinephrine reuptake inhibition than it does for serotonin reuptake inhibition, compared with duloxetine or venlafaxine.2

This difference in selectivity could lend itself to treating symptoms of MDD that might be related to norepinephrine deficiency; these include decreased concentration, lassitude, mental and physical slowing, and decreased self-care.3,4 Some authors claim that individual patients could experience improvement in their social and occupational functioning in addition to improvement in the core symptoms of depression.5

Levomilnacipran is the more active enantiomer of milnacipran, an SNRI that is approved for treating fibromyalgia in the United States and approved for treating depression in many other countries.6 In general, enantiomeric formulations are believed to have advantages over racemic formulations because they are less complex and have a more selective pharmacodynamic profile, better therapeutic index, lower liability for drug-drug interactions (DDIs), and a less complicated relationship between plasma concentration and pharmacodynamic effect.6 In addition, regulatory guidelines in the United States recommend development of enantiomers over racemates where appropriate.7

How it works

Levomilnacipran’s exact mechanism of action is unknown. Similar to other SNRIs, it binds with high affinity to the serotonin (5-HT) and norepinephrine (NE) transporters and potently inhibits 5-HT and NE reuptake. Levomilnacipran lacks significant affinity for any other receptors, ion channels, or transporters tested in vitro.2 It differs from other SNRIs such as venlafaxine and duloxetine in having higher selectivity for norepinephrine vs serotonin reuptake inhibition. In vitro studies demonstrated a 2-fold preference for NE over 5-HT reuptake inhibition.2

Pharmacokinetics

Levomilnacipran reaches maximum plasma concentration within 6 to 8 hours of oral administration and has a half-life of approximately 12 hours, which makes it suitable for once-daily dosing. The concentration of levomilnacipran at steady state is proportional to the dosage of the drug when administered within the range of 25 to 300 mg once daily.1

The drug’s mean apparent total clearance is 21 to 29 liters/hour and its bioavailability is not significantly affected when taken with food. The drug is widely distributed in the body and is converted primarily to 2 metabolites: desethy levomilnacipran and p-hydroxy-levomilnacipran. Both metabolites are inactive and undergo further conjugation with glucuronide. The drug is eliminated primarily via renal excretion.1

The major enzyme that catalyzes metabolism of levomilnacipram is cytochrome P 450 (CYP) 3A4, which makes it susceptible to DDIs with drugs that inhibit or induce this enzyme. For example, a person taking levomilnacipran with a potent CYP3A4 inhibitor, such as ketoconazole, may require a dosage adjustment. No dosage adjustment is needed when given with a CYP3A4 inducer or substrate. Drinking alcohol with levomilnacipran may cause more rapid release of the drug into the blood stream.1

Efficacy

Levomilnacipran decreased core symptoms of MDD and showed a statistically significant separation from placebo in 2 phase III RCTs (Table 2).3,5 The first study was a 10-week flexible dose (75 or 100 mg) trial in 563 outpatients age 18 to 70 who met DSM-IV-TR criteria of MDD for >1 month and had a 17-item Hamilton Depression Rating Scale (HDRS-17) score >22 and a Sheehan Disability Scale (SDS) score >10.3 The primary efficacy measure was change in Montgomery-Åsberg Depression Rating Scale (MADRS) score from baseline to week 10. Secondary efficacy measures included the HDRS-17, SDS, and Clinical Global Impressions-improvement scale. Efficacy analyses included 276 subjects treated with levomilnacipran and 277 treated with placebo.3

Levomilnacipran was significantly superior to placebo on the MADRS and HDRS-17 from baseline to week 10. Response and remission rates were
significantly greater for the levomilnacipran group compared with pla
cebo. Response exceeded the 10% average advantage for drug vs placebo and 46% of levomilnacipran-treated patients achieved remission.3

The number needed to treat (NNT), based on the MADRS scores for the levomilnacipran group compared with the placebo group, was 6 for response and 5 for remission.3 By comparison, most studies of venlafaxine demonstrate a NNT of 8.3

 

 

Levomilnacipran generally was reported to be safe and well tolerated. The most common adverse events leading to discontinuation in the levomilnacipran group were nausea, vomiting, change in systolic and diastolic blood pressure, and increase in heart rate. The favorable tolerability profile of levomilnacipran may relate to the 2-fold greater potency for NE reuptake inhibition compared with 5-HT reuptake inhibition.3

The second study was an 11-week, fixed-dose trial of levomilnacipran using 40, 80, or 120 mg. A total of 724 outpatients age 18 to 65 who met DSM-IV-TR criteria for MDD and who had an ongoing episode of depression lasting >8 weeks were randomly assigned to receive placebo (n = 179) or levomilnacipran at 40 mg (n = 181), 80 mg (n = 181), or 120 mg (n = 183) once daily for 8 weeks of double-blind treatment followed by a 2-week, double-blind taper of the drug.5 The primary efficacy parameter was change from baseline on the MADRS and the secondary efficacy parameter was change from baseline in SDS total score. HDRS-17, CGI-I, and CGI-S were included as secondary outcome measures.5

Significant difference in MADRS total score were seen in the levomilnacipran group compared with the placebo group (least mean squared difference: 40 mg/d,  −3.23; 80 mg/d, −3.99; and 120 mg/d,  −4.86). Higher dosages produced a numerically greater change and significant separation from placebo occurred sooner in the 80-mg and 120-mg groups compared with the 40-mg group.5

Significant differences vs placebo were consistently observed across secondary outcome measures for the higher dosages of levomilnacipran, and improvement in SDS total score was noted in all levomilnacipran groups compared with the placebo group. When dosed at 120 mg/d, levomilnacipran produced significant improvement vs placebo on all SDS subscales and was as well tolerated as the 80 mg dosage.5

No new safety concerns were observed in the study. A dose-response relationship in tolerability was not demonstrated and the number of patients reporting adverse events and who discontinued participation because of adverse events was higher in the 80-mg group than in the 40-mg and 120-mg groups.5

Tolerability

Overall, levomilnacipran was well tolerated in clinical trials, during which 2,673 subjects were exposed to the drug—translating to 942 patient-years of exposure. These patients ranged in age from 18 to 78; 825 of these subjects were enrolled in long-term studies for 1 year. Dosing of levomilnacipran during these studies ranged from 40 to 120 mg once daily, without regard to food.1

Nine percent of patients who received levomilnacipran in short-term studies discontinued because of adverse events, compared with 3% of patients who received placebo. The most common adverse event reported was nausea; other common adverse events reported included constipation, hyperhidrosis, elevated heart rate, erectile dysfunction, tachycardia, palpitations, and vomiting. Of these events, only erectile dysfunction and urinary hesitation were dose-related.1 Among levo­milnacipran-treated female patients, <2% reported adverse events related to sexual dysfunction.

All SNRIs have well established associations with elevation in blood pressure and heart rate. Levomilnacipran resulted in a mean increase of 3 mm HG in systolic and 3.2 mm Hg in diastolic blood pressure in short-term, placebo-controlled trials.1

Orthostatic hypotension was observed in 11.6% of patients in the levomilnacipran groups, compared with 9.7% in placebo groups in all short-term studies. Orthostatic reductions of blood pressure occurred in 5.8%, 6.1%, and 9.8% of levomilnacipran-treated patients with dosages of 40, 80, and 120 mg/d respectively, indicating a dose-dependent adverse event. A mean increase in heart rate of 7.2 beats per minute (bpm) also was seen in short-term studies in the levomilnacipran-treated group compared with 0.3 bpm in the placebo-treated group.1 Clinicians should monitor blood pressure and heart rate routinely because of potential increases seen in some subjects in these studies, which excluded those who had significant cardiovascular disease.

Unique clinical issues

Both in-vitro and in-vivo studies found that levomilnacipran exhibited more potency for NE reuptake inhibition than for 5-HT reuptake inhibition at the lowest effective dosage (10 mg/kg). However as the dosage was increased (20 mg/kg and 40 mg/kg), it was equally potent at NE and 5-HT reuptake inhibition. This is in contrast to venlafaxine, which demonstrates a similar, but opposite, effect in terms of potentiation at the 5-HT and NE reuptake pumps.2

The greater noradrenergic effect of levomilnacipran could lend itself to treating certain subgroups of patients whose symptoms are believed to be related to deficiencies in NE (eg, lassitude).4 This concept is theoretical, and was not explicitly studied in clinical trials and the drug is not labeled in this way.

Contraindications

Contraindications to levomilnacipran are similar to those seen with SSRIs and SNRIs, including concomitant use of a monoamine oxidase inhibitor (MAOI) and the use of the levomilnacipran within 14 days of stopping an MAOI. Contraindications unique to levomilnacipran include hypersensitivity to levomilnacipran, milnacipran, or any component specific to the formulation; and uncontrolled narrow-angle glaucoma.1

 

 

Dosing

The recommended dosage range of levomilnacipran is 40 to 120 mg once
daily with or without food. The capsules should be swallowed whole and should not be opened or crushed. As with most psychotropics, levomilnacipran should be taken at approximately the same time each day.1

The manufacturer recommends an initial dose of levomilnacipran of 20 mg once daily for 2 days, increased to 40 mg once daily. Based on efficacy and tolerably, levomilnacipran can be increased in increments of 40 mg every 2 days.

Dosage adjustment is recommended for patients with moderate or severe renal impairment; and the maintenance dosage should not exceed 80 mg and 40 mg respectively in these populations. As with many antidepressants, gradual dosage reduction is recommended to avoid discontinuation symptoms.

Bottom Line

Levomilnacipran is FDA-approved for treating major depressive disorder in adults. In 2 randomized controlled trials, the drug showed a significant separation from placebo. Levomilnacipran generally was reported to be safe and well tolerated; common adverse events were nausea, vomiting, changes in blood pressure, and an increase in heart rate.

Related Resources

 

  • Citrome L. Levomilnacipran for major depressive disorder: a systematic review of the efficacy and safety profile for this newly approved antidepressant - what is the number needed to treat, number needed to harm and likelihood to be helped or harmed? [published online September 8, 2013]. Int J Clin Pract. doi: 10.1111/ijcp.12298.
  • Mago R, Forero G, Greenberg WM, et al. Safety and tolerability of levomilnacipran ER in major depressive disorder: results from an open-label, 48-week extension study. Clin Drug Investig. 2013;33(10):761-771.


Drug Brand Names
Duloxetine • Cymbalta        Milnacipran • Savella
Ketoconazole • Nizoral        Venlafaxine • Effexor    Levomilnacipran • Fetzima
 

Disclosures

Dr. Macaluso has been the principal investigator for clinical trials for AbbVie, Eisai, Envivo, Janssen, Naurex, and Pfizer. All clinical trial and study contracts and payments were made through the Kansas University Medical Center Research Institute. Drs. Kazanchi and Malhotra report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

 

1. Fetzima [package insert]. St. Louis, MO: Forest Laboratories; 2013.

2. Auclair AL, Martel JC, Assié MB, et al. Levomilnacipran (F2695), a norepinephrine-preferring SNRI: profile in vitro and in models of depression and anxiety. Neuropharmacology. 2013;70:338-347.

3. Montgomery SA, Mansuy L, Ruth A, et al. Efficacy and Safety of levomilnacipran sustained release in moderate to severe major depressive disorder: a randomized, double-blind, placebo-controlled, proof-of-concept study. J Clin Psychiatry. 2013;74(4):363-369.

4. Kasper S, Meshkat D, Kutzelnigg A. Improvement of the noradrenergic symptom cluster following treatment with milnacipran. Neuropsychiatric Dis Treat. 2011; 7(suppl 1):21-27.

5. Asnis GM, Bose A, Gommoll CP, et al. Efficacy and safety of levomilnacipran sustained release 40 mg, 80 mg, or 120 mg in major depressive disorder: a phase 3, randomized, double-blind, placebo-controlled study. J Clin Psychiatry. 2013;74(3):242-248.

6. Hutt AJ, Vanetová J. The chiral switch: the development of single enantiomer drugs from racemates. Acta Facultatis Pharmaceuticae Universitatis Comenianae. 2003; 50(7):23.

7. U.S. Food and Drug Administration. Development of new stereoisomeric drugs. Published May 1, 1992. http://www.fda.gov/drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm122883.htm#.UKHEWm4ZyYE.email. Accessed October 8, 2013.

References

 

1. Fetzima [package insert]. St. Louis, MO: Forest Laboratories; 2013.

2. Auclair AL, Martel JC, Assié MB, et al. Levomilnacipran (F2695), a norepinephrine-preferring SNRI: profile in vitro and in models of depression and anxiety. Neuropharmacology. 2013;70:338-347.

3. Montgomery SA, Mansuy L, Ruth A, et al. Efficacy and Safety of levomilnacipran sustained release in moderate to severe major depressive disorder: a randomized, double-blind, placebo-controlled, proof-of-concept study. J Clin Psychiatry. 2013;74(4):363-369.

4. Kasper S, Meshkat D, Kutzelnigg A. Improvement of the noradrenergic symptom cluster following treatment with milnacipran. Neuropsychiatric Dis Treat. 2011; 7(suppl 1):21-27.

5. Asnis GM, Bose A, Gommoll CP, et al. Efficacy and safety of levomilnacipran sustained release 40 mg, 80 mg, or 120 mg in major depressive disorder: a phase 3, randomized, double-blind, placebo-controlled study. J Clin Psychiatry. 2013;74(3):242-248.

6. Hutt AJ, Vanetová J. The chiral switch: the development of single enantiomer drugs from racemates. Acta Facultatis Pharmaceuticae Universitatis Comenianae. 2003; 50(7):23.

7. U.S. Food and Drug Administration. Development of new stereoisomeric drugs. Published May 1, 1992. http://www.fda.gov/drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm122883.htm#.UKHEWm4ZyYE.email. Accessed October 8, 2013.

Issue
Current Psychiatry - 12(12)
Issue
Current Psychiatry - 12(12)
Page Number
50-52, 54-55
Page Number
50-52, 54-55
Publications
Publications
Topics
Article Type
Display Headline
Levomilnacipran for the treatment of major depressive disorder
Display Headline
Levomilnacipran for the treatment of major depressive disorder
Sections
Disallow All Ads
Alternative CME
Article PDF Media

Overcoming medication nonadherence in schizophrenia: Strategies that can reduce harm

Article Type
Changed
Tue, 12/11/2018 - 15:03
Display Headline
Overcoming medication nonadherence in schizophrenia: Strategies that can reduce harm

Medication nonadherence is a common problem when treating patients with schizophrenia that can worsen prognosis and lead to sub-optimal treatment outcomes. In this article, we discuss common reasons for nonadherence and describe evidence-based treatments intended to increase adherence and improve outcomes (Box).1-6

Common reasons for nonadherence

The primary predictor of future nonadherence is a history of nonadherence. It is important to understand patients’ reasons for nonadherence so that practical and evidence-based solutions can be implemented into the treatment plans of individual patients.

The 2009 Expert Consensus Guidelines on Adherence Problems in Patients with Serious and Persistent Mental Illness divided variables related to nonadherence into 3 categories:

  • those that lie within the patient (intrinsic)
  • those that are related to the patient’s relationship with healthcare providers, family, or caregivers (extrinsic)
  • those that are related to the healthcare delivery system (extrinsic).7

Among intrinsic variables, studies have shown a correlation between nonadherence and education level, lower socioeconomic status, homelessness, and male sex.7 (The Expert Consensus Guidelines considered homelessness to be an intrinsic factor because it was used as a demographic variable in the studies.)

Cognitive and negative symptoms associated with schizophrenia are an intrinsic risk factor for nonadherence because patients might not remember when or how to take medication.7 In a study by Freudenreich and co-workers8 of 81 outpatients who had a diagnosis of schizophrenia, the presence of negative symptoms predicted a negative attitude toward psychotropic medications. Poor insight might be the result of cognitive dysfunction associated with schizophrenia, and often is due to a lack of awareness of the importance of taking medications.

Limited insight into the need for treatment can be problematic early in the course of the illness when it may be directly related to positive symptoms. Perkins and colleagues9 demonstrated that patients recovering from a first psychotic episode who had limited insight into their illness and lacked desire to seek treatment were less adherent with medication. In another study, 5% of psychiatrists surveyed thought that many of their patients with schizophrenia were nonadherent because those patients did not believe that medications were effective or useful.10

Comorbid substance abuse disorders can contribute to medication nonadherence. In an analysis of 6,731 patients with schizophrenia, Novick and co-workers reported that alcohol dependence and substance abuse in the previous month predicted medication nonadherence.11 Hunt and colleagues demonstrated that, among 99 nonadherent patients with schizophrenia, time to first readmission was shorter for patients with comorbid substance abuse disorders compared with patients who had a diagnosis of schizophrenia only. Over the 4-year study period, the 28 patients who had a dual diagnosis (schizophrenia and substance abuse) accounted for 57% of all hospital readmissions.12

Several variables that affect medication adherence are related to the patient’s relationship with healthcare providers, family, caregivers, and the service delivery system.7 These include:

  • the perceived stigma of being given a diagnosis of a serious mental illness
  • adverse effects related to medications
  • poor social and family support
  • difficulty gaining access to mental health services.7,10

Societal stigma associated with seeking treatment from a mental health professional may contribute to nonadherence in some patients. In 1 study,13 36% of people surveyed would not want to work closely with a person who has a serious mental illness.

Adverse effects contribute significantly to nonadherence

Limited treatment options (which may be expensive) can make it difficult to manage the adverse effects of antipsychotics. In a cross-sectional survey of 876 patients, investigators reported that: 1) <50% of patients were adherent with medication, and 2) 80% experienced ≥1 side effect that was reported to be “somewhat bothersome” in self-ratings (Table 1).14 Extrapyramidal symptoms (EPS) and agitation were most strongly associated with nonadherence; weight gain, akathisia, and sexual dysfunction also were associated with nonadherence.14 This study did not distinguish adverse effects associated with first-generation antipsychotics (FGAs) from those associated with second-generation antipsychotics (SGAs), even though 71.7% of patients studied were taking an SGA.

A meta-analysis by Leucht and co-workers15 compared 15 antipsychotics (the FGAs haloperidol and chlorpromazine and 13 SGAs) for efficacy and tolerability in schizophrenia. Haloperidol had the highest rate of discontinuation for any

cause; chlorpromazine was eighth, compared with all other drugs. Haloperidol and chlorpromazine were first and third, respectively, in terms of causing EPS. Haloperidol was the least likely to cause weight gain; only olanzapine, clozapine, and iloperidone caused more weight gain than chlorpromazine. Haloperidol was eighth in terms of sedation; only clozapine was reportedly more sedating than chlorpromazine.15

Antipsychotic binding affinities to dopamine 2 (D2), serotonin 2A (5-HT2A), histamine (H1), and other receptors have an impact on a medication’s side-effect profile. Because of individual patient characteristics, you might be faced with choosing a medication that has a lower risk of EPS but a higher risk of weight gain and metabolic complications—or the inverse. Understanding binding affinities, side-effect profiles, and how to minimize or utilize adverse effects (ie, giving a drug that is approved to treat schizophrenia and is associated with weight gain to a patient with schizophrenia who has lost weight) may lead to greater adherence (Table 216 and Table 317).

 

 

Adequate support is essential

The therapeutic alliance plays a key role in patients’ attitudes toward taking medication. Magura and colleagues18 found that one-third of psychiatric patients (13% of whom had a diagnosis of schizophrenia) reported that their psychiatrist did not spend enough time with them explaining side effects, and felt “rushed.”

Patients with schizophrenia often require access to social support systems provided by family members, friends, and community agencies that provide case management and attendant care services. Patients who are adherent to medication tend to have greater perceived family involvement in medication treatment, and tend to have been raised in a family that had more of a positive attitude toward medication.19

In our practice, we have observed that recent state and federal budget cuts have resulted in patients having greater difficulty gaining access to case management and attendant care services, which then leads to increased rates of medication nonadherence. Be aware that variables such as limited office hours, financial hardship, and cultural and language barriers can compromise a patient’s ability to seek and continue care.

In the following section, we lay out techniques for improving adherence in patients with schizophrenia.

Employ general and specific strategies to boost adherence

How can you raise medication adherence concerns with patients, keeping in mind that they often overestimate their adherence?

Ask. Some clinicians ask questions such as “Are you taking your medication?”, although a more effective approach might be to ask how the patient is taking his (her) medication. Asking questions such as “When do you take your medication?” and “In the past week, how many doses do you think you missed?” might be more effective ways to inquire about adherence.7

The Expert Consensus Guidelines recommend asking patients about medication adherence monthly for those who are stable, doing well, and believed to be adherent. For those who are new to a practice or who are not doing well, inquire about medication adherence at least weekly.7

In our practice, patients who are unstable but do not require inpatient hospitalization typically are seen more often in the clinic, or are referred to intensive outpatient or partial hospitalization programs. If an unstable patient is unable to come in for more frequent appointments, we arrange phone conferences between her and her provider. If a patient is not doing well and has a case manager, we often ask that case manager to visit the patient, in person, more often than he (she) would otherwise.

Take a nonjudgmental approach when raising these issues with patients. Questions such as “We all forget to take our medication sometimes; do you?” help to normalize nonadherence, and improve the therapeutic alliance, and might result in the patient being more honest with the clinician.7 Because patients may be apprehensive about discussing adverse events, clinicians must be proactive about improving the therapeutic alliance and making patients feel comfortable when discussing sensitive topics. Clinicians should try to convey the idea that, although adherence is a concern, so is quality of life. A clinicians’ willingness to take a flexible approach that is nonpunitive nor authoritarian can aid the therapeutic alliance and improve overall adherence.

Be sensitive to financial, cultural, and language variables that can affect access to care. The Expert Consensus Guidelines recommend asking patients if they can afford their medication. In our practice, we have seen patients with schizophrenia discharged from the hospital only to be readmitted 1 month later because they could not afford to fill their prescriptions.

It is important to have translation services available, in person or by phone, for patients who do not speak English. Furthermore, it is important to understand the limitations that your practice might place on access to care. Ask patients if they have ever had trouble making an appointment when they needed to be seen, or if they called the office with a question and did not receive an answer in a timely fashion; doing so allows you to assess the practice’s ability to meet patients’ needs and helps you build a therapeutic alliance.

Make objective assessments. It is important for practitioners to not base their assessment of medication adherence solely on subjective findings. Asking patients to bring in their medication bottles for pill counts and checking with the patients’ pharmacies for information about refill frequency can provide some objective data. Electronic monitoring systems use microprocessors inserted into bottle caps to record the occurrence and timing of each bottle opening. Studies show that these electronic monitoring systems are the gold standard for determining medication adherence and could be used in cases where it is unclear if the patient is taking his (her) medication.7,20 Such systems have successfully monitored medication adherence in clinical trials, but their use in clinical practice is complicated by ethical and legal considerations and cost issues.

 

 

Simplify the regimen. Using medications with once-daily dosing, for example, can help improve adherence. Pfeiffer and co-workers21 found that patients whose medication regimens were changed from once daily to more than once daily experienced a decrease in medication adherence. Conversely, a decrease in dosing frequency was significantly associated with improved adherence. More than once-daily dosing was only weakly associated with poorer adherence among patients already on a stable regimen.

Discussing positive and negative aspects of past medication trials with a patient and inquiring if she prefers a specific medication can be an effective way to build the therapeutic relationship and help with adherence.

Direct patients to psychosocial interventions. These can be broadly classified as:

  • educational approaches
  • group therapy approaches
  • family interventions
  • cognitive treatments
  • combination approaches.

Psychoeducational approaches have limited effect on improving adherence when delivered to individual patients. However, 1 study showed that psychoeducation was effective at improving adherence when extended to include the patient’s family.22

Motivational interviewing techniques, behavioral approaches, and family interventions are effective at increasing medication adherence. One study looked at the value of training a patient-identified informant to supervise and administer medication. This person, usually a family member or close support, was responsible for obtaining medication from the pharmacy, administering the medication, and recording adherence. After 1 year, 67% of patients who used an informant were adherent, compared with only 45% in the group that did not have informant support.22 Case managers, attendant care workers, home health nurses, and assertive community treatment (ACT) teams also can participate in this manner; it is important, therefore, for you to be aware of the resources available in your community and to understand your role as patient advocate.

Substance abuse is a strong risk factor for nonadherence among patients with schizophrenia,18 which makes it important to assess patients for substance use and encourage those who do abuse to seek treatment. Although 1 study showed no correlation between Alcoholics Anonymous (AA) attendance and medication adherence,12 many AA and Narcotics Anonymous groups do not discuss psychiatric medications during group meetings. Magura and colleagues encouraged the use of “dual focus” groups that involve mental health professionals and addiction treatment specialists discussing mental health and substance abuse issues at the same setting.18

Prescribe long-acting injectable antipsychotics. Typically, long-acting injectable antipsychotics (LAIs) are reserved for patients who have a history of nonadherence. In a small study (N = 97) comparing LAI risperidone and oral risperidone or oral haloperidol, patients treated with an LAI had significantly fewer all-cause discontinuations (26.0%, compared with 70.2%) at 24 months.23 The Adherence to Treatment and Therapeutic Strategies in Schizophrenic Patients study examined 1,848 patients with schizophrenia and reached similar findings regarding LAI antipsychotics.24 (Note: Aripiprazole, fluphenazine, haloperidol, olanzapine, and paliperidone also are available in an LAI formulation.)

Bottom Line

Antipsychotic nonadherence in schizophrenia is a major problem for patients, families, and society. Being able to identify patients at risk for nonadherence, understanding the reasons for their nonadherence, and seeking practical solutions to the problem are all the responsibility of the treating physician. Psychoeducation, addressing substance abuse, modifying dosing, and using long-acting injectable antipsychotics may help improve adherence.

Related Resources

  • Velligan DI, Weiden PJ, Sajatovic M, et al; Expert Consensus Panel on Adherence Problems in Serious and Persistent Mental Illness. Assessment of adherence problems in patients with serious and persistent mental illness: recommendations from the Expert Consensus Guidelines. J Clin Psychiatry. 2009;70(suppl 4):1-46.
  • National Alliance on Mental Illness. www.nami.org.
  • Assertive Community Treatment (ACT) Organization. www.actassociation.org.

Drug Brand Names

Aripiprazole • Abilify        Chlorpromazine • Thorazine      Clozapine • Clozaril       Fluphenazine • Permitil    Haloperidol • Haldol                  Iloperidone • Fanapt       Olanzapine • Zyprexa      Paliperidone • Invega               Perphenazine • Trilafon    Quetiapine • Seroquel      Risperidone • Risperdal            Ziprasidone • Geodon

Disclosures

Dr. Macaluso has been the principal investigator for clinical trials for AbbVie, Eisai, Envivo, Janssen, Naurex, and Pfizer. All clinical trial and study contracts and payments were made through the Kansas University Medical Center Research Institute. Dr. McKnight reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Sun SX, Liu GG, Christensen DB, et al. Review and analysis of hospitalization costs associated with antipsychotic nonadherence in the treatment of schizophrenia in the United States. Curr Med Res Opin. 2007;23:2305-2312.

2. Lacro JP, Dunn LB, Dolder CR, et al. Prevalence of and risk factors for medication nonadherence in patients with schizophrenia: a comprehensive review of recent literature. J Clin Psychiatry. 2002;63:892-909.

3. Fenton WS, Blyler CR, Heinssen RK. Determinants of medication compliance in schizophrenia: empirical and clinical findings. Schizophr Bull. 1997;23(4):637-651.

4. Olfson M, Mechanic D, Hansell S, et al. Predicting medication noncompliance after hospital discharge among patients with schizophrenia. Psychiatr Serv. 2000;51(2):216-222.

5. Herings RM, Erkens JA. Increased suicide attempt rate among patients interrupting use of atypical antipsychotics. Pharmacoepidemial Drug Saf. 2003;12(5):423-424.

6. Weiden PJ, Kozma C, Grogg A, et al. Partial compliance and risk of rehospitalization among California Medicaid patients with schizophrenia. Psychiatr Serv. 2004;55(8):886-891.

7. Velligan D, Weiden P, Sajatovic M, et al. Assessment of adherence problems in patients with serious and persistent mental illness. J Psychiatr Pract. 2010;16(1):34-45.

8. Freudenreich O, Cather C, Evins A, et al. Attitudes of schizophrenia outpatients toward psychiatric medications: relationship to clinical variables and insight. J Clin Psychiatry. 2004;65(10):1372-1376.

9. Perkins DO, Johnson JL, Hamer RM, et al. Predictors of antipsychotic medication adherence in patients recovering from a first psychotic episode. Schizophr Res. 2006;83(1):53-63.

10. Olivares JM, Alptekin K, Azorin JM, et al. Psychiatrists’ awareness of adherence to antipsychotic medication in patients with schizophrenia: results from a survey conducted across Europe, the Middle East, and Africa. Patient Prefer Adherence. 2013;7:121-132.

11. Novick D, Haro J, Suarez D, et al. Predictors and clinical consequences of nonadherence with antipsychotic medication in the outpatient treatment of schizophrenia. Psychiatry Res. 2010;176(2-3):109-113.

12. Hunt GE, Bergen J, Bashir M, et al. Medication compliance and comorbid substance abuse in schizophrenia: impact on community survival four years after a relapse. Schizophr Res. 2002;54(3):253-264.

13. McGinty EE, Webster DW, Barry CL. Effects of news media messages about mass shootings on attitudes toward persons with serious mental illness and public support for gun control policies. Am J Psychiatry. 2013;170(5):494-501.

14. DiBonaventura M, Gabriel S, Dupclay L, et al. A patient perspective of the impact of medication side effects on adherence: results of a cross-sectional nationwide survey of patients with schizophrenia. BMC Psychiatry. 2012;12:20.

15. Leucht S, Cipriani A, Spineli L, et al. Comparative efficacy and tolerability of 15 antipsychotic drugs in schizophrenia: a multiple-treatments meta-analysis. Lancet. 2013;1382(9896): 951-962.

16. Robinson DS. Antipsychotics: pharmacology and clinical decision making. Primary Psychiatry. 2007;14(10):23-25.

17. Robinson D, Correll CU, Kane JM, et al. Practical dosing strategies in the treatment of schizophrenia. CNS Spectr. 2010;15:4(suppl 6):1-16.

18. Magura S, Rosenblum A, Fong C. Factors associated with medication adherence among psychiatric outpatients at substance abuse risk. Open Addict J. 2011;4:58-64.

19. Baloush-Kleinman V, Levine SZ, Roe D, et al. Adherence to antipsychotic drug treatment in early-episode schizophrenia: a six-month naturalistic follow-up study. Schizophr Res. 2011;130(1-3):176-181.

20. Byerly M, Nakonezny P, Lescouflair E. Antipsychotic medication adherence in schizophrenia. Psychiatr Clin North Am. 2007;30:437-452.

21. Pfeiffer PN, Ganoczy D, Valenstein M. Dosing frequency and adherence to antipsychotic medications. Psychiatr Serv. 2008;59(10):1207-1210.

22. Farooq S, Nazar Z, Irfan M, et al. Schizophrenia medication adherence in a resource-poor setting: randomized controlled trial of supervised treatment in out-patients for schizophrenia (STOPS). Br J Psychiatry. 2011;199(6):467-472.

23. Emsley R, Oosthuizen P, Koen L, et al. Oral vs injectable antipsychotic treatment in early psychosis: post hoc comparison of two studies. Clin Ther. 2008;30(12):2378-2386.

24. Gutierrez-Casares JR, Canãs F, Rodriguez-Morales A, et al. Adherence to treatment and therapeutic strategies in schizophrenic patients: the ADHERE study. CNS Spectr. 2010;15(5):327-337.

Article PDF
Author and Disclosure Information

Matthew Macaluso, DO
Assistant Professor
Director of Research
Associate Director, Residency Training
Psychiatry and Behavioral Sciences
University of Kansas School of Medicine-Wichita


Shean McKnight, MD

Chief Resident
Psychiatry and Behavioral Sciences
University of Kansas School of Medicine-Wichita

Wichita, Kansas

Issue
Current Psychiatry - 12(11)
Publications
Topics
Page Number
14-20
Legacy Keywords
medication nonadherence, dosing strategies, psychoeducation, schizophrenia, substance abuse, antipsychotics, first-generation antipsychotics, FGAs, second-generation antipsychotics, SGAs
Sections
Author and Disclosure Information

Matthew Macaluso, DO
Assistant Professor
Director of Research
Associate Director, Residency Training
Psychiatry and Behavioral Sciences
University of Kansas School of Medicine-Wichita


Shean McKnight, MD

Chief Resident
Psychiatry and Behavioral Sciences
University of Kansas School of Medicine-Wichita

Wichita, Kansas

Author and Disclosure Information

Matthew Macaluso, DO
Assistant Professor
Director of Research
Associate Director, Residency Training
Psychiatry and Behavioral Sciences
University of Kansas School of Medicine-Wichita


Shean McKnight, MD

Chief Resident
Psychiatry and Behavioral Sciences
University of Kansas School of Medicine-Wichita

Wichita, Kansas

Article PDF
Article PDF
Related Articles

Medication nonadherence is a common problem when treating patients with schizophrenia that can worsen prognosis and lead to sub-optimal treatment outcomes. In this article, we discuss common reasons for nonadherence and describe evidence-based treatments intended to increase adherence and improve outcomes (Box).1-6

Common reasons for nonadherence

The primary predictor of future nonadherence is a history of nonadherence. It is important to understand patients’ reasons for nonadherence so that practical and evidence-based solutions can be implemented into the treatment plans of individual patients.

The 2009 Expert Consensus Guidelines on Adherence Problems in Patients with Serious and Persistent Mental Illness divided variables related to nonadherence into 3 categories:

  • those that lie within the patient (intrinsic)
  • those that are related to the patient’s relationship with healthcare providers, family, or caregivers (extrinsic)
  • those that are related to the healthcare delivery system (extrinsic).7

Among intrinsic variables, studies have shown a correlation between nonadherence and education level, lower socioeconomic status, homelessness, and male sex.7 (The Expert Consensus Guidelines considered homelessness to be an intrinsic factor because it was used as a demographic variable in the studies.)

Cognitive and negative symptoms associated with schizophrenia are an intrinsic risk factor for nonadherence because patients might not remember when or how to take medication.7 In a study by Freudenreich and co-workers8 of 81 outpatients who had a diagnosis of schizophrenia, the presence of negative symptoms predicted a negative attitude toward psychotropic medications. Poor insight might be the result of cognitive dysfunction associated with schizophrenia, and often is due to a lack of awareness of the importance of taking medications.

Limited insight into the need for treatment can be problematic early in the course of the illness when it may be directly related to positive symptoms. Perkins and colleagues9 demonstrated that patients recovering from a first psychotic episode who had limited insight into their illness and lacked desire to seek treatment were less adherent with medication. In another study, 5% of psychiatrists surveyed thought that many of their patients with schizophrenia were nonadherent because those patients did not believe that medications were effective or useful.10

Comorbid substance abuse disorders can contribute to medication nonadherence. In an analysis of 6,731 patients with schizophrenia, Novick and co-workers reported that alcohol dependence and substance abuse in the previous month predicted medication nonadherence.11 Hunt and colleagues demonstrated that, among 99 nonadherent patients with schizophrenia, time to first readmission was shorter for patients with comorbid substance abuse disorders compared with patients who had a diagnosis of schizophrenia only. Over the 4-year study period, the 28 patients who had a dual diagnosis (schizophrenia and substance abuse) accounted for 57% of all hospital readmissions.12

Several variables that affect medication adherence are related to the patient’s relationship with healthcare providers, family, caregivers, and the service delivery system.7 These include:

  • the perceived stigma of being given a diagnosis of a serious mental illness
  • adverse effects related to medications
  • poor social and family support
  • difficulty gaining access to mental health services.7,10

Societal stigma associated with seeking treatment from a mental health professional may contribute to nonadherence in some patients. In 1 study,13 36% of people surveyed would not want to work closely with a person who has a serious mental illness.

Adverse effects contribute significantly to nonadherence

Limited treatment options (which may be expensive) can make it difficult to manage the adverse effects of antipsychotics. In a cross-sectional survey of 876 patients, investigators reported that: 1) <50% of patients were adherent with medication, and 2) 80% experienced ≥1 side effect that was reported to be “somewhat bothersome” in self-ratings (Table 1).14 Extrapyramidal symptoms (EPS) and agitation were most strongly associated with nonadherence; weight gain, akathisia, and sexual dysfunction also were associated with nonadherence.14 This study did not distinguish adverse effects associated with first-generation antipsychotics (FGAs) from those associated with second-generation antipsychotics (SGAs), even though 71.7% of patients studied were taking an SGA.

A meta-analysis by Leucht and co-workers15 compared 15 antipsychotics (the FGAs haloperidol and chlorpromazine and 13 SGAs) for efficacy and tolerability in schizophrenia. Haloperidol had the highest rate of discontinuation for any

cause; chlorpromazine was eighth, compared with all other drugs. Haloperidol and chlorpromazine were first and third, respectively, in terms of causing EPS. Haloperidol was the least likely to cause weight gain; only olanzapine, clozapine, and iloperidone caused more weight gain than chlorpromazine. Haloperidol was eighth in terms of sedation; only clozapine was reportedly more sedating than chlorpromazine.15

Antipsychotic binding affinities to dopamine 2 (D2), serotonin 2A (5-HT2A), histamine (H1), and other receptors have an impact on a medication’s side-effect profile. Because of individual patient characteristics, you might be faced with choosing a medication that has a lower risk of EPS but a higher risk of weight gain and metabolic complications—or the inverse. Understanding binding affinities, side-effect profiles, and how to minimize or utilize adverse effects (ie, giving a drug that is approved to treat schizophrenia and is associated with weight gain to a patient with schizophrenia who has lost weight) may lead to greater adherence (Table 216 and Table 317).

 

 

Adequate support is essential

The therapeutic alliance plays a key role in patients’ attitudes toward taking medication. Magura and colleagues18 found that one-third of psychiatric patients (13% of whom had a diagnosis of schizophrenia) reported that their psychiatrist did not spend enough time with them explaining side effects, and felt “rushed.”

Patients with schizophrenia often require access to social support systems provided by family members, friends, and community agencies that provide case management and attendant care services. Patients who are adherent to medication tend to have greater perceived family involvement in medication treatment, and tend to have been raised in a family that had more of a positive attitude toward medication.19

In our practice, we have observed that recent state and federal budget cuts have resulted in patients having greater difficulty gaining access to case management and attendant care services, which then leads to increased rates of medication nonadherence. Be aware that variables such as limited office hours, financial hardship, and cultural and language barriers can compromise a patient’s ability to seek and continue care.

In the following section, we lay out techniques for improving adherence in patients with schizophrenia.

Employ general and specific strategies to boost adherence

How can you raise medication adherence concerns with patients, keeping in mind that they often overestimate their adherence?

Ask. Some clinicians ask questions such as “Are you taking your medication?”, although a more effective approach might be to ask how the patient is taking his (her) medication. Asking questions such as “When do you take your medication?” and “In the past week, how many doses do you think you missed?” might be more effective ways to inquire about adherence.7

The Expert Consensus Guidelines recommend asking patients about medication adherence monthly for those who are stable, doing well, and believed to be adherent. For those who are new to a practice or who are not doing well, inquire about medication adherence at least weekly.7

In our practice, patients who are unstable but do not require inpatient hospitalization typically are seen more often in the clinic, or are referred to intensive outpatient or partial hospitalization programs. If an unstable patient is unable to come in for more frequent appointments, we arrange phone conferences between her and her provider. If a patient is not doing well and has a case manager, we often ask that case manager to visit the patient, in person, more often than he (she) would otherwise.

Take a nonjudgmental approach when raising these issues with patients. Questions such as “We all forget to take our medication sometimes; do you?” help to normalize nonadherence, and improve the therapeutic alliance, and might result in the patient being more honest with the clinician.7 Because patients may be apprehensive about discussing adverse events, clinicians must be proactive about improving the therapeutic alliance and making patients feel comfortable when discussing sensitive topics. Clinicians should try to convey the idea that, although adherence is a concern, so is quality of life. A clinicians’ willingness to take a flexible approach that is nonpunitive nor authoritarian can aid the therapeutic alliance and improve overall adherence.

Be sensitive to financial, cultural, and language variables that can affect access to care. The Expert Consensus Guidelines recommend asking patients if they can afford their medication. In our practice, we have seen patients with schizophrenia discharged from the hospital only to be readmitted 1 month later because they could not afford to fill their prescriptions.

It is important to have translation services available, in person or by phone, for patients who do not speak English. Furthermore, it is important to understand the limitations that your practice might place on access to care. Ask patients if they have ever had trouble making an appointment when they needed to be seen, or if they called the office with a question and did not receive an answer in a timely fashion; doing so allows you to assess the practice’s ability to meet patients’ needs and helps you build a therapeutic alliance.

Make objective assessments. It is important for practitioners to not base their assessment of medication adherence solely on subjective findings. Asking patients to bring in their medication bottles for pill counts and checking with the patients’ pharmacies for information about refill frequency can provide some objective data. Electronic monitoring systems use microprocessors inserted into bottle caps to record the occurrence and timing of each bottle opening. Studies show that these electronic monitoring systems are the gold standard for determining medication adherence and could be used in cases where it is unclear if the patient is taking his (her) medication.7,20 Such systems have successfully monitored medication adherence in clinical trials, but their use in clinical practice is complicated by ethical and legal considerations and cost issues.

 

 

Simplify the regimen. Using medications with once-daily dosing, for example, can help improve adherence. Pfeiffer and co-workers21 found that patients whose medication regimens were changed from once daily to more than once daily experienced a decrease in medication adherence. Conversely, a decrease in dosing frequency was significantly associated with improved adherence. More than once-daily dosing was only weakly associated with poorer adherence among patients already on a stable regimen.

Discussing positive and negative aspects of past medication trials with a patient and inquiring if she prefers a specific medication can be an effective way to build the therapeutic relationship and help with adherence.

Direct patients to psychosocial interventions. These can be broadly classified as:

  • educational approaches
  • group therapy approaches
  • family interventions
  • cognitive treatments
  • combination approaches.

Psychoeducational approaches have limited effect on improving adherence when delivered to individual patients. However, 1 study showed that psychoeducation was effective at improving adherence when extended to include the patient’s family.22

Motivational interviewing techniques, behavioral approaches, and family interventions are effective at increasing medication adherence. One study looked at the value of training a patient-identified informant to supervise and administer medication. This person, usually a family member or close support, was responsible for obtaining medication from the pharmacy, administering the medication, and recording adherence. After 1 year, 67% of patients who used an informant were adherent, compared with only 45% in the group that did not have informant support.22 Case managers, attendant care workers, home health nurses, and assertive community treatment (ACT) teams also can participate in this manner; it is important, therefore, for you to be aware of the resources available in your community and to understand your role as patient advocate.

Substance abuse is a strong risk factor for nonadherence among patients with schizophrenia,18 which makes it important to assess patients for substance use and encourage those who do abuse to seek treatment. Although 1 study showed no correlation between Alcoholics Anonymous (AA) attendance and medication adherence,12 many AA and Narcotics Anonymous groups do not discuss psychiatric medications during group meetings. Magura and colleagues encouraged the use of “dual focus” groups that involve mental health professionals and addiction treatment specialists discussing mental health and substance abuse issues at the same setting.18

Prescribe long-acting injectable antipsychotics. Typically, long-acting injectable antipsychotics (LAIs) are reserved for patients who have a history of nonadherence. In a small study (N = 97) comparing LAI risperidone and oral risperidone or oral haloperidol, patients treated with an LAI had significantly fewer all-cause discontinuations (26.0%, compared with 70.2%) at 24 months.23 The Adherence to Treatment and Therapeutic Strategies in Schizophrenic Patients study examined 1,848 patients with schizophrenia and reached similar findings regarding LAI antipsychotics.24 (Note: Aripiprazole, fluphenazine, haloperidol, olanzapine, and paliperidone also are available in an LAI formulation.)

Bottom Line

Antipsychotic nonadherence in schizophrenia is a major problem for patients, families, and society. Being able to identify patients at risk for nonadherence, understanding the reasons for their nonadherence, and seeking practical solutions to the problem are all the responsibility of the treating physician. Psychoeducation, addressing substance abuse, modifying dosing, and using long-acting injectable antipsychotics may help improve adherence.

Related Resources

  • Velligan DI, Weiden PJ, Sajatovic M, et al; Expert Consensus Panel on Adherence Problems in Serious and Persistent Mental Illness. Assessment of adherence problems in patients with serious and persistent mental illness: recommendations from the Expert Consensus Guidelines. J Clin Psychiatry. 2009;70(suppl 4):1-46.
  • National Alliance on Mental Illness. www.nami.org.
  • Assertive Community Treatment (ACT) Organization. www.actassociation.org.

Drug Brand Names

Aripiprazole • Abilify        Chlorpromazine • Thorazine      Clozapine • Clozaril       Fluphenazine • Permitil    Haloperidol • Haldol                  Iloperidone • Fanapt       Olanzapine • Zyprexa      Paliperidone • Invega               Perphenazine • Trilafon    Quetiapine • Seroquel      Risperidone • Risperdal            Ziprasidone • Geodon

Disclosures

Dr. Macaluso has been the principal investigator for clinical trials for AbbVie, Eisai, Envivo, Janssen, Naurex, and Pfizer. All clinical trial and study contracts and payments were made through the Kansas University Medical Center Research Institute. Dr. McKnight reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Medication nonadherence is a common problem when treating patients with schizophrenia that can worsen prognosis and lead to sub-optimal treatment outcomes. In this article, we discuss common reasons for nonadherence and describe evidence-based treatments intended to increase adherence and improve outcomes (Box).1-6

Common reasons for nonadherence

The primary predictor of future nonadherence is a history of nonadherence. It is important to understand patients’ reasons for nonadherence so that practical and evidence-based solutions can be implemented into the treatment plans of individual patients.

The 2009 Expert Consensus Guidelines on Adherence Problems in Patients with Serious and Persistent Mental Illness divided variables related to nonadherence into 3 categories:

  • those that lie within the patient (intrinsic)
  • those that are related to the patient’s relationship with healthcare providers, family, or caregivers (extrinsic)
  • those that are related to the healthcare delivery system (extrinsic).7

Among intrinsic variables, studies have shown a correlation between nonadherence and education level, lower socioeconomic status, homelessness, and male sex.7 (The Expert Consensus Guidelines considered homelessness to be an intrinsic factor because it was used as a demographic variable in the studies.)

Cognitive and negative symptoms associated with schizophrenia are an intrinsic risk factor for nonadherence because patients might not remember when or how to take medication.7 In a study by Freudenreich and co-workers8 of 81 outpatients who had a diagnosis of schizophrenia, the presence of negative symptoms predicted a negative attitude toward psychotropic medications. Poor insight might be the result of cognitive dysfunction associated with schizophrenia, and often is due to a lack of awareness of the importance of taking medications.

Limited insight into the need for treatment can be problematic early in the course of the illness when it may be directly related to positive symptoms. Perkins and colleagues9 demonstrated that patients recovering from a first psychotic episode who had limited insight into their illness and lacked desire to seek treatment were less adherent with medication. In another study, 5% of psychiatrists surveyed thought that many of their patients with schizophrenia were nonadherent because those patients did not believe that medications were effective or useful.10

Comorbid substance abuse disorders can contribute to medication nonadherence. In an analysis of 6,731 patients with schizophrenia, Novick and co-workers reported that alcohol dependence and substance abuse in the previous month predicted medication nonadherence.11 Hunt and colleagues demonstrated that, among 99 nonadherent patients with schizophrenia, time to first readmission was shorter for patients with comorbid substance abuse disorders compared with patients who had a diagnosis of schizophrenia only. Over the 4-year study period, the 28 patients who had a dual diagnosis (schizophrenia and substance abuse) accounted for 57% of all hospital readmissions.12

Several variables that affect medication adherence are related to the patient’s relationship with healthcare providers, family, caregivers, and the service delivery system.7 These include:

  • the perceived stigma of being given a diagnosis of a serious mental illness
  • adverse effects related to medications
  • poor social and family support
  • difficulty gaining access to mental health services.7,10

Societal stigma associated with seeking treatment from a mental health professional may contribute to nonadherence in some patients. In 1 study,13 36% of people surveyed would not want to work closely with a person who has a serious mental illness.

Adverse effects contribute significantly to nonadherence

Limited treatment options (which may be expensive) can make it difficult to manage the adverse effects of antipsychotics. In a cross-sectional survey of 876 patients, investigators reported that: 1) <50% of patients were adherent with medication, and 2) 80% experienced ≥1 side effect that was reported to be “somewhat bothersome” in self-ratings (Table 1).14 Extrapyramidal symptoms (EPS) and agitation were most strongly associated with nonadherence; weight gain, akathisia, and sexual dysfunction also were associated with nonadherence.14 This study did not distinguish adverse effects associated with first-generation antipsychotics (FGAs) from those associated with second-generation antipsychotics (SGAs), even though 71.7% of patients studied were taking an SGA.

A meta-analysis by Leucht and co-workers15 compared 15 antipsychotics (the FGAs haloperidol and chlorpromazine and 13 SGAs) for efficacy and tolerability in schizophrenia. Haloperidol had the highest rate of discontinuation for any

cause; chlorpromazine was eighth, compared with all other drugs. Haloperidol and chlorpromazine were first and third, respectively, in terms of causing EPS. Haloperidol was the least likely to cause weight gain; only olanzapine, clozapine, and iloperidone caused more weight gain than chlorpromazine. Haloperidol was eighth in terms of sedation; only clozapine was reportedly more sedating than chlorpromazine.15

Antipsychotic binding affinities to dopamine 2 (D2), serotonin 2A (5-HT2A), histamine (H1), and other receptors have an impact on a medication’s side-effect profile. Because of individual patient characteristics, you might be faced with choosing a medication that has a lower risk of EPS but a higher risk of weight gain and metabolic complications—or the inverse. Understanding binding affinities, side-effect profiles, and how to minimize or utilize adverse effects (ie, giving a drug that is approved to treat schizophrenia and is associated with weight gain to a patient with schizophrenia who has lost weight) may lead to greater adherence (Table 216 and Table 317).

 

 

Adequate support is essential

The therapeutic alliance plays a key role in patients’ attitudes toward taking medication. Magura and colleagues18 found that one-third of psychiatric patients (13% of whom had a diagnosis of schizophrenia) reported that their psychiatrist did not spend enough time with them explaining side effects, and felt “rushed.”

Patients with schizophrenia often require access to social support systems provided by family members, friends, and community agencies that provide case management and attendant care services. Patients who are adherent to medication tend to have greater perceived family involvement in medication treatment, and tend to have been raised in a family that had more of a positive attitude toward medication.19

In our practice, we have observed that recent state and federal budget cuts have resulted in patients having greater difficulty gaining access to case management and attendant care services, which then leads to increased rates of medication nonadherence. Be aware that variables such as limited office hours, financial hardship, and cultural and language barriers can compromise a patient’s ability to seek and continue care.

In the following section, we lay out techniques for improving adherence in patients with schizophrenia.

Employ general and specific strategies to boost adherence

How can you raise medication adherence concerns with patients, keeping in mind that they often overestimate their adherence?

Ask. Some clinicians ask questions such as “Are you taking your medication?”, although a more effective approach might be to ask how the patient is taking his (her) medication. Asking questions such as “When do you take your medication?” and “In the past week, how many doses do you think you missed?” might be more effective ways to inquire about adherence.7

The Expert Consensus Guidelines recommend asking patients about medication adherence monthly for those who are stable, doing well, and believed to be adherent. For those who are new to a practice or who are not doing well, inquire about medication adherence at least weekly.7

In our practice, patients who are unstable but do not require inpatient hospitalization typically are seen more often in the clinic, or are referred to intensive outpatient or partial hospitalization programs. If an unstable patient is unable to come in for more frequent appointments, we arrange phone conferences between her and her provider. If a patient is not doing well and has a case manager, we often ask that case manager to visit the patient, in person, more often than he (she) would otherwise.

Take a nonjudgmental approach when raising these issues with patients. Questions such as “We all forget to take our medication sometimes; do you?” help to normalize nonadherence, and improve the therapeutic alliance, and might result in the patient being more honest with the clinician.7 Because patients may be apprehensive about discussing adverse events, clinicians must be proactive about improving the therapeutic alliance and making patients feel comfortable when discussing sensitive topics. Clinicians should try to convey the idea that, although adherence is a concern, so is quality of life. A clinicians’ willingness to take a flexible approach that is nonpunitive nor authoritarian can aid the therapeutic alliance and improve overall adherence.

Be sensitive to financial, cultural, and language variables that can affect access to care. The Expert Consensus Guidelines recommend asking patients if they can afford their medication. In our practice, we have seen patients with schizophrenia discharged from the hospital only to be readmitted 1 month later because they could not afford to fill their prescriptions.

It is important to have translation services available, in person or by phone, for patients who do not speak English. Furthermore, it is important to understand the limitations that your practice might place on access to care. Ask patients if they have ever had trouble making an appointment when they needed to be seen, or if they called the office with a question and did not receive an answer in a timely fashion; doing so allows you to assess the practice’s ability to meet patients’ needs and helps you build a therapeutic alliance.

Make objective assessments. It is important for practitioners to not base their assessment of medication adherence solely on subjective findings. Asking patients to bring in their medication bottles for pill counts and checking with the patients’ pharmacies for information about refill frequency can provide some objective data. Electronic monitoring systems use microprocessors inserted into bottle caps to record the occurrence and timing of each bottle opening. Studies show that these electronic monitoring systems are the gold standard for determining medication adherence and could be used in cases where it is unclear if the patient is taking his (her) medication.7,20 Such systems have successfully monitored medication adherence in clinical trials, but their use in clinical practice is complicated by ethical and legal considerations and cost issues.

 

 

Simplify the regimen. Using medications with once-daily dosing, for example, can help improve adherence. Pfeiffer and co-workers21 found that patients whose medication regimens were changed from once daily to more than once daily experienced a decrease in medication adherence. Conversely, a decrease in dosing frequency was significantly associated with improved adherence. More than once-daily dosing was only weakly associated with poorer adherence among patients already on a stable regimen.

Discussing positive and negative aspects of past medication trials with a patient and inquiring if she prefers a specific medication can be an effective way to build the therapeutic relationship and help with adherence.

Direct patients to psychosocial interventions. These can be broadly classified as:

  • educational approaches
  • group therapy approaches
  • family interventions
  • cognitive treatments
  • combination approaches.

Psychoeducational approaches have limited effect on improving adherence when delivered to individual patients. However, 1 study showed that psychoeducation was effective at improving adherence when extended to include the patient’s family.22

Motivational interviewing techniques, behavioral approaches, and family interventions are effective at increasing medication adherence. One study looked at the value of training a patient-identified informant to supervise and administer medication. This person, usually a family member or close support, was responsible for obtaining medication from the pharmacy, administering the medication, and recording adherence. After 1 year, 67% of patients who used an informant were adherent, compared with only 45% in the group that did not have informant support.22 Case managers, attendant care workers, home health nurses, and assertive community treatment (ACT) teams also can participate in this manner; it is important, therefore, for you to be aware of the resources available in your community and to understand your role as patient advocate.

Substance abuse is a strong risk factor for nonadherence among patients with schizophrenia,18 which makes it important to assess patients for substance use and encourage those who do abuse to seek treatment. Although 1 study showed no correlation between Alcoholics Anonymous (AA) attendance and medication adherence,12 many AA and Narcotics Anonymous groups do not discuss psychiatric medications during group meetings. Magura and colleagues encouraged the use of “dual focus” groups that involve mental health professionals and addiction treatment specialists discussing mental health and substance abuse issues at the same setting.18

Prescribe long-acting injectable antipsychotics. Typically, long-acting injectable antipsychotics (LAIs) are reserved for patients who have a history of nonadherence. In a small study (N = 97) comparing LAI risperidone and oral risperidone or oral haloperidol, patients treated with an LAI had significantly fewer all-cause discontinuations (26.0%, compared with 70.2%) at 24 months.23 The Adherence to Treatment and Therapeutic Strategies in Schizophrenic Patients study examined 1,848 patients with schizophrenia and reached similar findings regarding LAI antipsychotics.24 (Note: Aripiprazole, fluphenazine, haloperidol, olanzapine, and paliperidone also are available in an LAI formulation.)

Bottom Line

Antipsychotic nonadherence in schizophrenia is a major problem for patients, families, and society. Being able to identify patients at risk for nonadherence, understanding the reasons for their nonadherence, and seeking practical solutions to the problem are all the responsibility of the treating physician. Psychoeducation, addressing substance abuse, modifying dosing, and using long-acting injectable antipsychotics may help improve adherence.

Related Resources

  • Velligan DI, Weiden PJ, Sajatovic M, et al; Expert Consensus Panel on Adherence Problems in Serious and Persistent Mental Illness. Assessment of adherence problems in patients with serious and persistent mental illness: recommendations from the Expert Consensus Guidelines. J Clin Psychiatry. 2009;70(suppl 4):1-46.
  • National Alliance on Mental Illness. www.nami.org.
  • Assertive Community Treatment (ACT) Organization. www.actassociation.org.

Drug Brand Names

Aripiprazole • Abilify        Chlorpromazine • Thorazine      Clozapine • Clozaril       Fluphenazine • Permitil    Haloperidol • Haldol                  Iloperidone • Fanapt       Olanzapine • Zyprexa      Paliperidone • Invega               Perphenazine • Trilafon    Quetiapine • Seroquel      Risperidone • Risperdal            Ziprasidone • Geodon

Disclosures

Dr. Macaluso has been the principal investigator for clinical trials for AbbVie, Eisai, Envivo, Janssen, Naurex, and Pfizer. All clinical trial and study contracts and payments were made through the Kansas University Medical Center Research Institute. Dr. McKnight reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Sun SX, Liu GG, Christensen DB, et al. Review and analysis of hospitalization costs associated with antipsychotic nonadherence in the treatment of schizophrenia in the United States. Curr Med Res Opin. 2007;23:2305-2312.

2. Lacro JP, Dunn LB, Dolder CR, et al. Prevalence of and risk factors for medication nonadherence in patients with schizophrenia: a comprehensive review of recent literature. J Clin Psychiatry. 2002;63:892-909.

3. Fenton WS, Blyler CR, Heinssen RK. Determinants of medication compliance in schizophrenia: empirical and clinical findings. Schizophr Bull. 1997;23(4):637-651.

4. Olfson M, Mechanic D, Hansell S, et al. Predicting medication noncompliance after hospital discharge among patients with schizophrenia. Psychiatr Serv. 2000;51(2):216-222.

5. Herings RM, Erkens JA. Increased suicide attempt rate among patients interrupting use of atypical antipsychotics. Pharmacoepidemial Drug Saf. 2003;12(5):423-424.

6. Weiden PJ, Kozma C, Grogg A, et al. Partial compliance and risk of rehospitalization among California Medicaid patients with schizophrenia. Psychiatr Serv. 2004;55(8):886-891.

7. Velligan D, Weiden P, Sajatovic M, et al. Assessment of adherence problems in patients with serious and persistent mental illness. J Psychiatr Pract. 2010;16(1):34-45.

8. Freudenreich O, Cather C, Evins A, et al. Attitudes of schizophrenia outpatients toward psychiatric medications: relationship to clinical variables and insight. J Clin Psychiatry. 2004;65(10):1372-1376.

9. Perkins DO, Johnson JL, Hamer RM, et al. Predictors of antipsychotic medication adherence in patients recovering from a first psychotic episode. Schizophr Res. 2006;83(1):53-63.

10. Olivares JM, Alptekin K, Azorin JM, et al. Psychiatrists’ awareness of adherence to antipsychotic medication in patients with schizophrenia: results from a survey conducted across Europe, the Middle East, and Africa. Patient Prefer Adherence. 2013;7:121-132.

11. Novick D, Haro J, Suarez D, et al. Predictors and clinical consequences of nonadherence with antipsychotic medication in the outpatient treatment of schizophrenia. Psychiatry Res. 2010;176(2-3):109-113.

12. Hunt GE, Bergen J, Bashir M, et al. Medication compliance and comorbid substance abuse in schizophrenia: impact on community survival four years after a relapse. Schizophr Res. 2002;54(3):253-264.

13. McGinty EE, Webster DW, Barry CL. Effects of news media messages about mass shootings on attitudes toward persons with serious mental illness and public support for gun control policies. Am J Psychiatry. 2013;170(5):494-501.

14. DiBonaventura M, Gabriel S, Dupclay L, et al. A patient perspective of the impact of medication side effects on adherence: results of a cross-sectional nationwide survey of patients with schizophrenia. BMC Psychiatry. 2012;12:20.

15. Leucht S, Cipriani A, Spineli L, et al. Comparative efficacy and tolerability of 15 antipsychotic drugs in schizophrenia: a multiple-treatments meta-analysis. Lancet. 2013;1382(9896): 951-962.

16. Robinson DS. Antipsychotics: pharmacology and clinical decision making. Primary Psychiatry. 2007;14(10):23-25.

17. Robinson D, Correll CU, Kane JM, et al. Practical dosing strategies in the treatment of schizophrenia. CNS Spectr. 2010;15:4(suppl 6):1-16.

18. Magura S, Rosenblum A, Fong C. Factors associated with medication adherence among psychiatric outpatients at substance abuse risk. Open Addict J. 2011;4:58-64.

19. Baloush-Kleinman V, Levine SZ, Roe D, et al. Adherence to antipsychotic drug treatment in early-episode schizophrenia: a six-month naturalistic follow-up study. Schizophr Res. 2011;130(1-3):176-181.

20. Byerly M, Nakonezny P, Lescouflair E. Antipsychotic medication adherence in schizophrenia. Psychiatr Clin North Am. 2007;30:437-452.

21. Pfeiffer PN, Ganoczy D, Valenstein M. Dosing frequency and adherence to antipsychotic medications. Psychiatr Serv. 2008;59(10):1207-1210.

22. Farooq S, Nazar Z, Irfan M, et al. Schizophrenia medication adherence in a resource-poor setting: randomized controlled trial of supervised treatment in out-patients for schizophrenia (STOPS). Br J Psychiatry. 2011;199(6):467-472.

23. Emsley R, Oosthuizen P, Koen L, et al. Oral vs injectable antipsychotic treatment in early psychosis: post hoc comparison of two studies. Clin Ther. 2008;30(12):2378-2386.

24. Gutierrez-Casares JR, Canãs F, Rodriguez-Morales A, et al. Adherence to treatment and therapeutic strategies in schizophrenic patients: the ADHERE study. CNS Spectr. 2010;15(5):327-337.

References

1. Sun SX, Liu GG, Christensen DB, et al. Review and analysis of hospitalization costs associated with antipsychotic nonadherence in the treatment of schizophrenia in the United States. Curr Med Res Opin. 2007;23:2305-2312.

2. Lacro JP, Dunn LB, Dolder CR, et al. Prevalence of and risk factors for medication nonadherence in patients with schizophrenia: a comprehensive review of recent literature. J Clin Psychiatry. 2002;63:892-909.

3. Fenton WS, Blyler CR, Heinssen RK. Determinants of medication compliance in schizophrenia: empirical and clinical findings. Schizophr Bull. 1997;23(4):637-651.

4. Olfson M, Mechanic D, Hansell S, et al. Predicting medication noncompliance after hospital discharge among patients with schizophrenia. Psychiatr Serv. 2000;51(2):216-222.

5. Herings RM, Erkens JA. Increased suicide attempt rate among patients interrupting use of atypical antipsychotics. Pharmacoepidemial Drug Saf. 2003;12(5):423-424.

6. Weiden PJ, Kozma C, Grogg A, et al. Partial compliance and risk of rehospitalization among California Medicaid patients with schizophrenia. Psychiatr Serv. 2004;55(8):886-891.

7. Velligan D, Weiden P, Sajatovic M, et al. Assessment of adherence problems in patients with serious and persistent mental illness. J Psychiatr Pract. 2010;16(1):34-45.

8. Freudenreich O, Cather C, Evins A, et al. Attitudes of schizophrenia outpatients toward psychiatric medications: relationship to clinical variables and insight. J Clin Psychiatry. 2004;65(10):1372-1376.

9. Perkins DO, Johnson JL, Hamer RM, et al. Predictors of antipsychotic medication adherence in patients recovering from a first psychotic episode. Schizophr Res. 2006;83(1):53-63.

10. Olivares JM, Alptekin K, Azorin JM, et al. Psychiatrists’ awareness of adherence to antipsychotic medication in patients with schizophrenia: results from a survey conducted across Europe, the Middle East, and Africa. Patient Prefer Adherence. 2013;7:121-132.

11. Novick D, Haro J, Suarez D, et al. Predictors and clinical consequences of nonadherence with antipsychotic medication in the outpatient treatment of schizophrenia. Psychiatry Res. 2010;176(2-3):109-113.

12. Hunt GE, Bergen J, Bashir M, et al. Medication compliance and comorbid substance abuse in schizophrenia: impact on community survival four years after a relapse. Schizophr Res. 2002;54(3):253-264.

13. McGinty EE, Webster DW, Barry CL. Effects of news media messages about mass shootings on attitudes toward persons with serious mental illness and public support for gun control policies. Am J Psychiatry. 2013;170(5):494-501.

14. DiBonaventura M, Gabriel S, Dupclay L, et al. A patient perspective of the impact of medication side effects on adherence: results of a cross-sectional nationwide survey of patients with schizophrenia. BMC Psychiatry. 2012;12:20.

15. Leucht S, Cipriani A, Spineli L, et al. Comparative efficacy and tolerability of 15 antipsychotic drugs in schizophrenia: a multiple-treatments meta-analysis. Lancet. 2013;1382(9896): 951-962.

16. Robinson DS. Antipsychotics: pharmacology and clinical decision making. Primary Psychiatry. 2007;14(10):23-25.

17. Robinson D, Correll CU, Kane JM, et al. Practical dosing strategies in the treatment of schizophrenia. CNS Spectr. 2010;15:4(suppl 6):1-16.

18. Magura S, Rosenblum A, Fong C. Factors associated with medication adherence among psychiatric outpatients at substance abuse risk. Open Addict J. 2011;4:58-64.

19. Baloush-Kleinman V, Levine SZ, Roe D, et al. Adherence to antipsychotic drug treatment in early-episode schizophrenia: a six-month naturalistic follow-up study. Schizophr Res. 2011;130(1-3):176-181.

20. Byerly M, Nakonezny P, Lescouflair E. Antipsychotic medication adherence in schizophrenia. Psychiatr Clin North Am. 2007;30:437-452.

21. Pfeiffer PN, Ganoczy D, Valenstein M. Dosing frequency and adherence to antipsychotic medications. Psychiatr Serv. 2008;59(10):1207-1210.

22. Farooq S, Nazar Z, Irfan M, et al. Schizophrenia medication adherence in a resource-poor setting: randomized controlled trial of supervised treatment in out-patients for schizophrenia (STOPS). Br J Psychiatry. 2011;199(6):467-472.

23. Emsley R, Oosthuizen P, Koen L, et al. Oral vs injectable antipsychotic treatment in early psychosis: post hoc comparison of two studies. Clin Ther. 2008;30(12):2378-2386.

24. Gutierrez-Casares JR, Canãs F, Rodriguez-Morales A, et al. Adherence to treatment and therapeutic strategies in schizophrenic patients: the ADHERE study. CNS Spectr. 2010;15(5):327-337.

Issue
Current Psychiatry - 12(11)
Issue
Current Psychiatry - 12(11)
Page Number
14-20
Page Number
14-20
Publications
Publications
Topics
Article Type
Display Headline
Overcoming medication nonadherence in schizophrenia: Strategies that can reduce harm
Display Headline
Overcoming medication nonadherence in schizophrenia: Strategies that can reduce harm
Legacy Keywords
medication nonadherence, dosing strategies, psychoeducation, schizophrenia, substance abuse, antipsychotics, first-generation antipsychotics, FGAs, second-generation antipsychotics, SGAs
Legacy Keywords
medication nonadherence, dosing strategies, psychoeducation, schizophrenia, substance abuse, antipsychotics, first-generation antipsychotics, FGAs, second-generation antipsychotics, SGAs
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Off-label use of antipsychotics

Article Type
Changed
Wed, 09/26/2018 - 14:41
Display Headline
Off-label use of antipsychotics

Author and Disclosure Information

Dr. Macaluso is Assistant professor, Psychiatry snd Behavioral Sciences, University of Kansas School of Medicine-Witchita, Witchita, Kansas.

Issue
Current Psychiatry - 12(02)
Publications
Topics
Sections
Author and Disclosure Information

Dr. Macaluso is Assistant professor, Psychiatry snd Behavioral Sciences, University of Kansas School of Medicine-Witchita, Witchita, Kansas.

Author and Disclosure Information

Dr. Macaluso is Assistant professor, Psychiatry snd Behavioral Sciences, University of Kansas School of Medicine-Witchita, Witchita, Kansas.

Issue
Current Psychiatry - 12(02)
Issue
Current Psychiatry - 12(02)
Publications
Publications
Topics
Article Type
Display Headline
Off-label use of antipsychotics
Display Headline
Off-label use of antipsychotics
Sections
Disallow All Ads
Alternative CME
Use ProPublica

A mysterious physical and mental decline

Article Type
Changed
Tue, 12/11/2018 - 15:30
Display Headline
A mysterious physical and mental decline

HISTORY: ‘Not himself’

Mr. C, age 69, presents to the emergency department complaining of intermittent fever of about 100°F, hematuria, headache, weakness, fatigue, and decreased appetite over 2 months. Testing shows acute renal failure, elevated C-reactive protein, and increased sedimentation rate. The attending internist admits Mr. C with a working diagnosis of temporal arteritis and acute renal failure, administers corticosteroids for headache, and orders a right temporal artery biopsy, which shows no signs of vasculitis.

Family members report that Mr. C has not been himself—he has become increasingly withdrawn and “emotionless.” Mr. C’s wife says her husband has needed help with dressing and eating because of short-term memory loss over 9 months. She says he has lost 20 to 30 lb.

The patient’s cognitive function appears to have worsened since he developed these physical symptoms. Mrs. C also reports that he has had weakness and fatigue for 8 months.

One month earlier, the patient was admitted to a different hospital and treated for 2 weeks with IV antibiotics for fever of unknown origin. Results of lumbar puncture and extensive rheumatologic, infectious disease, urologic, and gastroenterologic evaluations were normal.

The internal medicine physician requests a psychiatric consultation. During our interview, Mr. C is cooperative, shows no signs of acute distress, is well groomed and dressed appropriately, and maintains eye contact. Speech rate and volume are low, with normal articulation and coherence, diminished spontaneity, and paucity of language. Mrs. C tells us her husband was lively and talkative before his recent illness. His mood is euthymic, and he is pleasant and cheerful during the evaluation.

The authors’ observations

Initially, we suspect an underlying medical condition is causing Mr. C’s psychiatric symptoms.

Mr. C’s wife reports that her husband stopped drinking 2 years ago after his family expressed concern about his health. Mr. C’s past alcohol use could not be quantified. He has not abused illicit drugs and has no personal or family history of dementia, trauma, or psychiatric or neurologic disorders.

EVALUATION: Impaired memory

Mr. C is afebrile during the initial physical examination, but fever returns within several days. Neurologic examination is normal, and negative rapid plasma reagin rules out syphilis. Vitamin B12 and folate levels are normal, as is thyroid function. Other laboratory findings are outside normal limits (Table).

Urine is cloudy with 2+ protein, 3+ blood, and trace leukocyte esterase. The presence of protein and blood suggests a glomerular disease such as a glomerulonephritis.

A positive leukocyte esterase test results from the presence of white blood cells, either as whole cells or as lysed cells. An abnormal number of leukocytes may appear with upper or lower urinary tract infection or in acute glomerulonephritis.

Chest radiography shows increased bilateral pulmonary vasculature, which can indicate pulmonary hypertension.

Mr. C shows variable and incongruent affect and mood, often shifting from labile to blunted. He denies depressed mood. At times he is disinhibited and makes inappropriate remarks. His thought processes are decreased but generally logical and goal-directed. Mr. C reports no hallucinations or suicidal thinking. He has concrete reasoning with regard to his medical condition and linear associations with fair to poor insight/judgment. He exhibits intact impulse control and is oriented to person, place, and time.

Mr. C has fair attention and concentration but impaired recent memory. He cannot recall yesterday’s events without help.

Mr. C’s Mini-Mental State Examination score of 21/30 suggests markedly impaired executive functioning and cognitive deficits. The attending psychiatrist recommends brain MRI.

Table

Mr. C’s laboratory findings

 ValueNormal range
WBC15.14.8 to 10.8 cells/μL
Hb913.8 to 17.5 g/dL
Hct25.9%40% to 54%
MCV89.780 to 94 fL
BUN1197 to 18 mg/dL
Cr12.10.7 to 1.3 mg/dL
Na125136 to 145 mmol/L
K6.53.5 to 5 mEq/dL
HCO313.522 to 29 mmol/L
ECR12530 mm/hr
WBC: white blood cell; Hb: hemoglobin; Hct: hematocrit; MCV: mean corpuscular volume; BUN: blood urea nitrogen; Cr: creatinine; Na: sodium; K: potassium; HCO3: bicarbonate; ESR: erythrocyte sedimentation rate

The authors’ observations

Mr. C shows markedly impaired cognitive function without significant impairment of attention and concentration despite his progressive deterioration and increasing disability. Urine toxicology shows no illicit substances. Given his lack of a previous mood disorder and his family’s description of him as formerly vibrant and cheerful, he likely does not have a mood disorder.

Based on the history of events, including the symptom pattern, we rule out delirium. We suspect that Mr. C has dementia secondary to a general medical condition. His symptoms seem to be directly related to his medical complaints and do not have a waxing and waning course. The internal medicine physician orders additional laboratory tests.

 

 

TESTING: Kidney, lung damage

Over 5 days, Mr. C’s intermittent low-grade fevers continue. Laboratory tests are negative for HIV antibody, hepatitis panel, and antinuclear antibodies (ANA). C-reactive protein is elevated at 27.8 mg/dL (normal range,

Renal ultrasound is normal, but preliminary renal biopsy shows rapidly progressive glomerulonephritis. The internist immediately starts dialysis, cyclophosphamide at 1.5 mg/kg, and prednisone, 1 mg/kg. The pathology report on the renal biopsy describes extensive crescentic glomerular destruction, with inflammatory cells present.

Ten days after admission, Mr. C develops hemoptysis, and chest radiography shows increasing alveolar infiltrates. The attending internist consults pulmonary and critical care services.

The consultant suspects a pulmonary-renal syndrome because of bilateral alveolar infiltrates (diffuse alveolar hemorrhage). The internal medicine team continues high-dose corticosteroids, followed by plasmapheresis.

Brain MRI shows subacute to chronic infarcts involving the right basal ganglia and corona radiate and mild to moderate small vessel ischemic changes. Old areas of hemorrhage are noted within both cerebellar lobes, left temporal lobe, right basal ganglia, right parietal lobe, and right frontal lobe.

During follow-up interviews, Mr. C often cannot recall recent dialysis or plasmapheresis and reports no physical symptoms. His short-term memory continues to deteriorate; he would forget to eat if not cued by family or nursing staff. He shows global cognitive deficits and is increasingly withdrawn and flat.

The authors’ observations

Mr. C’s overall condition and medical test results suggest Goodpasture’s syndrome—also called anti-GBM disease—a rare autoimmune condition characterized by rapid destruction of the kidneys and hemorrhaging of the lungs (Box).1-5 We diagnose Goodpasture’s syndrome based on Mr. C’s anti-GBM antibody and pANCA findings combined with crescentic glomerular destruction on renal biopsy.

Although few case reports have associated Goodpasture’s syndrome with neurobehavioral changes, the apparent relationship of Mr. C’s medical symptoms with the worsening of his cognitive impairment suggests a link.

Mr. C’s MRI findings also might suggest CNS vasculitis, which affects small arteries of the cerebral and spinal cord leptomeninges and parenchyma, leading to CNS dys-function.6-8 CNS vasculitis can result from primary nervous system involvement or from a secondary systemic process such as Goodpasture’s syndrome.9

We rule out lupus because Mr. C is ANA-negative; this test has 99% sensitivity for lupus.10

Box

Goodpasture’s syndrome: A rare autoimmune disease

Goodpasture’s syndrome, which afflicts 1 Patients typically present with alveolar bleeding, rapidly progressive acute renal failure with proteinuria,1 and pulmonary symptoms such as dyspnea and hemoptysis.2

Possible triggers include:

  • viral upper respiratory tract infection (20% to 60% of patients)3
  • exposure to hydrocarbon solvents (3,4

Mr. C was exposed to solvents during the 15 years he worked in a factory. Some researchers believe a noxious event among genetically susceptible persons damages basement membrane and exposes an antigen that triggers IgG auto-antibody production.3,4

Malaise, weight loss, and fever are atypical in Goodpasture’s syndrome but could suggest concomitant vasculitis.5

OUTCOME: Ongoing disability

Mr. C is hospitalized for 6 weeks. He receives cyclophosphamide, prednisone, and 10 sessions of plasmapheresis. We prescribe mirtazapine, 15 mg at bedtime, to treat mood symptoms. We chose mirtazapine because of the drug’s sleep-restoring and appetite-stimulating properties.

Mr. C’s fever resolves and pulmonary function soon improves, but his cognitive impairment persists. He has difficulties preparing meals, taking medications, and managing his money.

Mr. C is discharged with a referral to a psychiatrist. He continues taking mirtazapine and a lower dose of prednisone. He requires ongoing hemodialysis and assistance with activities of daily living.

The authors’ observations

Prompt multidisciplinary intervention is critical when patients present with concurrent cognitive and medical symptoms. A thorough psychiatric evaluation can help piece together the illness’ course. The psychiatrist’s role in a multidisciplinary assessment is to:

  • document neurocognitive changes
  • verify them through collateral information
  • correlate these changes with the timing of medical symptoms.
Interview the patient alone, then seek collateral information to assemble the clinical picture and help establish premorbid functioning. Ask family members about the patient’s cognitive function at home or at work and in high-risk situations such as while driving or operating machinery.

An underlying psychiatric condition can complicate the diagnosis. In these cases, careful interviewing and collateral information can help you discern the chronology of events.

References

1. Bolton WK. Goodpasture’s syndrome. Kidney Int 1996;50(5):1753-66.

2. Pusey CD. Anti-glomerular basement membrane disease. Kidney Int 2003;64(4):1535-50.

3. Humes HD, DuPont HL. eds. Kelley’s textbook of internal medicine. 4th ed. New York, NY: Lippincott Williams & Wilkins; 2000.

4. Stevenson A, Yaqoob M, Mason H, et al. Biochemical markers of basement membrane disturbances and occupational exposure to hydrocarbons and mixed solvents. QJM 1995;88(1):23-8.

5. Kluth DC, Rees AJ. Anti-glomerular basement membrane disease. J Am Soc Nephrol. 1999;10(11):2446-53.

6. Rydel JJ, Rodby RA. An 18-year-old man with Goodpasture’s syndrome and ANCA-negative central nervous system vasculitis. Am J Kidney Dis 1998;31(2):345-9.

7. Gittins N, Basu A, Eyre J, et al. Cerebral vasculitis in a teenager with Goodpasture’s syndrome. Nephrol Dial Transplant 2004;19(12):3168-71.

8. Garnier P, Deprele C. Cerebral angiitis and Goodpasture’s syndrome. Rev Neurol 2003;159(1):68-70.

9. Calabrese LH, Duna GF, Lie JT. Vasculitis in the central nervous system. Arthritis Rhem 1997;40(7):1189-201.

10. Edworthy SM, Zatarain E, McShane DJ, Bloch DA. Analysis of the 1982 ARA lupus criteria data set by recursive partitioning methodology: new insights into the relative merit of individual criteria. J Rheumatol 1988;15(10):1493-8.

Article PDF
Author and Disclosure Information

Matthew Macaluso, DO;
Christine Skotzko, MD;
Maria Rueda-Lara, MD
Dr. Macaluso is chief resident, department of psychiatry and behavioral sciences, University of Kansas School of Medicine, Wichita. Dr. Skotzko is director of psychosomatic medicine, Morristown Memorial Hospital, Morristown, NJ. Dr. Rueda-Lara is assistant professor, department of psychiatry, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ.

Issue
Current Psychiatry - 07(12)
Publications
Page Number
49-58
Legacy Keywords
Matthew Macaluso;Christine Skotzko;Maria Rueda-Lara; Goodpasture's syndrome;Goodpasture syndrome;anti-GBM disease;medical cause of psychiatric symptoms;kidney destruction;lung hemorrhaging;CNS vasculitis
Sections
Author and Disclosure Information

Matthew Macaluso, DO;
Christine Skotzko, MD;
Maria Rueda-Lara, MD
Dr. Macaluso is chief resident, department of psychiatry and behavioral sciences, University of Kansas School of Medicine, Wichita. Dr. Skotzko is director of psychosomatic medicine, Morristown Memorial Hospital, Morristown, NJ. Dr. Rueda-Lara is assistant professor, department of psychiatry, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ.

Author and Disclosure Information

Matthew Macaluso, DO;
Christine Skotzko, MD;
Maria Rueda-Lara, MD
Dr. Macaluso is chief resident, department of psychiatry and behavioral sciences, University of Kansas School of Medicine, Wichita. Dr. Skotzko is director of psychosomatic medicine, Morristown Memorial Hospital, Morristown, NJ. Dr. Rueda-Lara is assistant professor, department of psychiatry, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ.

Article PDF
Article PDF

HISTORY: ‘Not himself’

Mr. C, age 69, presents to the emergency department complaining of intermittent fever of about 100°F, hematuria, headache, weakness, fatigue, and decreased appetite over 2 months. Testing shows acute renal failure, elevated C-reactive protein, and increased sedimentation rate. The attending internist admits Mr. C with a working diagnosis of temporal arteritis and acute renal failure, administers corticosteroids for headache, and orders a right temporal artery biopsy, which shows no signs of vasculitis.

Family members report that Mr. C has not been himself—he has become increasingly withdrawn and “emotionless.” Mr. C’s wife says her husband has needed help with dressing and eating because of short-term memory loss over 9 months. She says he has lost 20 to 30 lb.

The patient’s cognitive function appears to have worsened since he developed these physical symptoms. Mrs. C also reports that he has had weakness and fatigue for 8 months.

One month earlier, the patient was admitted to a different hospital and treated for 2 weeks with IV antibiotics for fever of unknown origin. Results of lumbar puncture and extensive rheumatologic, infectious disease, urologic, and gastroenterologic evaluations were normal.

The internal medicine physician requests a psychiatric consultation. During our interview, Mr. C is cooperative, shows no signs of acute distress, is well groomed and dressed appropriately, and maintains eye contact. Speech rate and volume are low, with normal articulation and coherence, diminished spontaneity, and paucity of language. Mrs. C tells us her husband was lively and talkative before his recent illness. His mood is euthymic, and he is pleasant and cheerful during the evaluation.

The authors’ observations

Initially, we suspect an underlying medical condition is causing Mr. C’s psychiatric symptoms.

Mr. C’s wife reports that her husband stopped drinking 2 years ago after his family expressed concern about his health. Mr. C’s past alcohol use could not be quantified. He has not abused illicit drugs and has no personal or family history of dementia, trauma, or psychiatric or neurologic disorders.

EVALUATION: Impaired memory

Mr. C is afebrile during the initial physical examination, but fever returns within several days. Neurologic examination is normal, and negative rapid plasma reagin rules out syphilis. Vitamin B12 and folate levels are normal, as is thyroid function. Other laboratory findings are outside normal limits (Table).

Urine is cloudy with 2+ protein, 3+ blood, and trace leukocyte esterase. The presence of protein and blood suggests a glomerular disease such as a glomerulonephritis.

A positive leukocyte esterase test results from the presence of white blood cells, either as whole cells or as lysed cells. An abnormal number of leukocytes may appear with upper or lower urinary tract infection or in acute glomerulonephritis.

Chest radiography shows increased bilateral pulmonary vasculature, which can indicate pulmonary hypertension.

Mr. C shows variable and incongruent affect and mood, often shifting from labile to blunted. He denies depressed mood. At times he is disinhibited and makes inappropriate remarks. His thought processes are decreased but generally logical and goal-directed. Mr. C reports no hallucinations or suicidal thinking. He has concrete reasoning with regard to his medical condition and linear associations with fair to poor insight/judgment. He exhibits intact impulse control and is oriented to person, place, and time.

Mr. C has fair attention and concentration but impaired recent memory. He cannot recall yesterday’s events without help.

Mr. C’s Mini-Mental State Examination score of 21/30 suggests markedly impaired executive functioning and cognitive deficits. The attending psychiatrist recommends brain MRI.

Table

Mr. C’s laboratory findings

 ValueNormal range
WBC15.14.8 to 10.8 cells/μL
Hb913.8 to 17.5 g/dL
Hct25.9%40% to 54%
MCV89.780 to 94 fL
BUN1197 to 18 mg/dL
Cr12.10.7 to 1.3 mg/dL
Na125136 to 145 mmol/L
K6.53.5 to 5 mEq/dL
HCO313.522 to 29 mmol/L
ECR12530 mm/hr
WBC: white blood cell; Hb: hemoglobin; Hct: hematocrit; MCV: mean corpuscular volume; BUN: blood urea nitrogen; Cr: creatinine; Na: sodium; K: potassium; HCO3: bicarbonate; ESR: erythrocyte sedimentation rate

The authors’ observations

Mr. C shows markedly impaired cognitive function without significant impairment of attention and concentration despite his progressive deterioration and increasing disability. Urine toxicology shows no illicit substances. Given his lack of a previous mood disorder and his family’s description of him as formerly vibrant and cheerful, he likely does not have a mood disorder.

Based on the history of events, including the symptom pattern, we rule out delirium. We suspect that Mr. C has dementia secondary to a general medical condition. His symptoms seem to be directly related to his medical complaints and do not have a waxing and waning course. The internal medicine physician orders additional laboratory tests.

 

 

TESTING: Kidney, lung damage

Over 5 days, Mr. C’s intermittent low-grade fevers continue. Laboratory tests are negative for HIV antibody, hepatitis panel, and antinuclear antibodies (ANA). C-reactive protein is elevated at 27.8 mg/dL (normal range,

Renal ultrasound is normal, but preliminary renal biopsy shows rapidly progressive glomerulonephritis. The internist immediately starts dialysis, cyclophosphamide at 1.5 mg/kg, and prednisone, 1 mg/kg. The pathology report on the renal biopsy describes extensive crescentic glomerular destruction, with inflammatory cells present.

Ten days after admission, Mr. C develops hemoptysis, and chest radiography shows increasing alveolar infiltrates. The attending internist consults pulmonary and critical care services.

The consultant suspects a pulmonary-renal syndrome because of bilateral alveolar infiltrates (diffuse alveolar hemorrhage). The internal medicine team continues high-dose corticosteroids, followed by plasmapheresis.

Brain MRI shows subacute to chronic infarcts involving the right basal ganglia and corona radiate and mild to moderate small vessel ischemic changes. Old areas of hemorrhage are noted within both cerebellar lobes, left temporal lobe, right basal ganglia, right parietal lobe, and right frontal lobe.

During follow-up interviews, Mr. C often cannot recall recent dialysis or plasmapheresis and reports no physical symptoms. His short-term memory continues to deteriorate; he would forget to eat if not cued by family or nursing staff. He shows global cognitive deficits and is increasingly withdrawn and flat.

The authors’ observations

Mr. C’s overall condition and medical test results suggest Goodpasture’s syndrome—also called anti-GBM disease—a rare autoimmune condition characterized by rapid destruction of the kidneys and hemorrhaging of the lungs (Box).1-5 We diagnose Goodpasture’s syndrome based on Mr. C’s anti-GBM antibody and pANCA findings combined with crescentic glomerular destruction on renal biopsy.

Although few case reports have associated Goodpasture’s syndrome with neurobehavioral changes, the apparent relationship of Mr. C’s medical symptoms with the worsening of his cognitive impairment suggests a link.

Mr. C’s MRI findings also might suggest CNS vasculitis, which affects small arteries of the cerebral and spinal cord leptomeninges and parenchyma, leading to CNS dys-function.6-8 CNS vasculitis can result from primary nervous system involvement or from a secondary systemic process such as Goodpasture’s syndrome.9

We rule out lupus because Mr. C is ANA-negative; this test has 99% sensitivity for lupus.10

Box

Goodpasture’s syndrome: A rare autoimmune disease

Goodpasture’s syndrome, which afflicts 1 Patients typically present with alveolar bleeding, rapidly progressive acute renal failure with proteinuria,1 and pulmonary symptoms such as dyspnea and hemoptysis.2

Possible triggers include:

  • viral upper respiratory tract infection (20% to 60% of patients)3
  • exposure to hydrocarbon solvents (3,4

Mr. C was exposed to solvents during the 15 years he worked in a factory. Some researchers believe a noxious event among genetically susceptible persons damages basement membrane and exposes an antigen that triggers IgG auto-antibody production.3,4

Malaise, weight loss, and fever are atypical in Goodpasture’s syndrome but could suggest concomitant vasculitis.5

OUTCOME: Ongoing disability

Mr. C is hospitalized for 6 weeks. He receives cyclophosphamide, prednisone, and 10 sessions of plasmapheresis. We prescribe mirtazapine, 15 mg at bedtime, to treat mood symptoms. We chose mirtazapine because of the drug’s sleep-restoring and appetite-stimulating properties.

Mr. C’s fever resolves and pulmonary function soon improves, but his cognitive impairment persists. He has difficulties preparing meals, taking medications, and managing his money.

Mr. C is discharged with a referral to a psychiatrist. He continues taking mirtazapine and a lower dose of prednisone. He requires ongoing hemodialysis and assistance with activities of daily living.

The authors’ observations

Prompt multidisciplinary intervention is critical when patients present with concurrent cognitive and medical symptoms. A thorough psychiatric evaluation can help piece together the illness’ course. The psychiatrist’s role in a multidisciplinary assessment is to:

  • document neurocognitive changes
  • verify them through collateral information
  • correlate these changes with the timing of medical symptoms.
Interview the patient alone, then seek collateral information to assemble the clinical picture and help establish premorbid functioning. Ask family members about the patient’s cognitive function at home or at work and in high-risk situations such as while driving or operating machinery.

An underlying psychiatric condition can complicate the diagnosis. In these cases, careful interviewing and collateral information can help you discern the chronology of events.

HISTORY: ‘Not himself’

Mr. C, age 69, presents to the emergency department complaining of intermittent fever of about 100°F, hematuria, headache, weakness, fatigue, and decreased appetite over 2 months. Testing shows acute renal failure, elevated C-reactive protein, and increased sedimentation rate. The attending internist admits Mr. C with a working diagnosis of temporal arteritis and acute renal failure, administers corticosteroids for headache, and orders a right temporal artery biopsy, which shows no signs of vasculitis.

Family members report that Mr. C has not been himself—he has become increasingly withdrawn and “emotionless.” Mr. C’s wife says her husband has needed help with dressing and eating because of short-term memory loss over 9 months. She says he has lost 20 to 30 lb.

The patient’s cognitive function appears to have worsened since he developed these physical symptoms. Mrs. C also reports that he has had weakness and fatigue for 8 months.

One month earlier, the patient was admitted to a different hospital and treated for 2 weeks with IV antibiotics for fever of unknown origin. Results of lumbar puncture and extensive rheumatologic, infectious disease, urologic, and gastroenterologic evaluations were normal.

The internal medicine physician requests a psychiatric consultation. During our interview, Mr. C is cooperative, shows no signs of acute distress, is well groomed and dressed appropriately, and maintains eye contact. Speech rate and volume are low, with normal articulation and coherence, diminished spontaneity, and paucity of language. Mrs. C tells us her husband was lively and talkative before his recent illness. His mood is euthymic, and he is pleasant and cheerful during the evaluation.

The authors’ observations

Initially, we suspect an underlying medical condition is causing Mr. C’s psychiatric symptoms.

Mr. C’s wife reports that her husband stopped drinking 2 years ago after his family expressed concern about his health. Mr. C’s past alcohol use could not be quantified. He has not abused illicit drugs and has no personal or family history of dementia, trauma, or psychiatric or neurologic disorders.

EVALUATION: Impaired memory

Mr. C is afebrile during the initial physical examination, but fever returns within several days. Neurologic examination is normal, and negative rapid plasma reagin rules out syphilis. Vitamin B12 and folate levels are normal, as is thyroid function. Other laboratory findings are outside normal limits (Table).

Urine is cloudy with 2+ protein, 3+ blood, and trace leukocyte esterase. The presence of protein and blood suggests a glomerular disease such as a glomerulonephritis.

A positive leukocyte esterase test results from the presence of white blood cells, either as whole cells or as lysed cells. An abnormal number of leukocytes may appear with upper or lower urinary tract infection or in acute glomerulonephritis.

Chest radiography shows increased bilateral pulmonary vasculature, which can indicate pulmonary hypertension.

Mr. C shows variable and incongruent affect and mood, often shifting from labile to blunted. He denies depressed mood. At times he is disinhibited and makes inappropriate remarks. His thought processes are decreased but generally logical and goal-directed. Mr. C reports no hallucinations or suicidal thinking. He has concrete reasoning with regard to his medical condition and linear associations with fair to poor insight/judgment. He exhibits intact impulse control and is oriented to person, place, and time.

Mr. C has fair attention and concentration but impaired recent memory. He cannot recall yesterday’s events without help.

Mr. C’s Mini-Mental State Examination score of 21/30 suggests markedly impaired executive functioning and cognitive deficits. The attending psychiatrist recommends brain MRI.

Table

Mr. C’s laboratory findings

 ValueNormal range
WBC15.14.8 to 10.8 cells/μL
Hb913.8 to 17.5 g/dL
Hct25.9%40% to 54%
MCV89.780 to 94 fL
BUN1197 to 18 mg/dL
Cr12.10.7 to 1.3 mg/dL
Na125136 to 145 mmol/L
K6.53.5 to 5 mEq/dL
HCO313.522 to 29 mmol/L
ECR12530 mm/hr
WBC: white blood cell; Hb: hemoglobin; Hct: hematocrit; MCV: mean corpuscular volume; BUN: blood urea nitrogen; Cr: creatinine; Na: sodium; K: potassium; HCO3: bicarbonate; ESR: erythrocyte sedimentation rate

The authors’ observations

Mr. C shows markedly impaired cognitive function without significant impairment of attention and concentration despite his progressive deterioration and increasing disability. Urine toxicology shows no illicit substances. Given his lack of a previous mood disorder and his family’s description of him as formerly vibrant and cheerful, he likely does not have a mood disorder.

Based on the history of events, including the symptom pattern, we rule out delirium. We suspect that Mr. C has dementia secondary to a general medical condition. His symptoms seem to be directly related to his medical complaints and do not have a waxing and waning course. The internal medicine physician orders additional laboratory tests.

 

 

TESTING: Kidney, lung damage

Over 5 days, Mr. C’s intermittent low-grade fevers continue. Laboratory tests are negative for HIV antibody, hepatitis panel, and antinuclear antibodies (ANA). C-reactive protein is elevated at 27.8 mg/dL (normal range,

Renal ultrasound is normal, but preliminary renal biopsy shows rapidly progressive glomerulonephritis. The internist immediately starts dialysis, cyclophosphamide at 1.5 mg/kg, and prednisone, 1 mg/kg. The pathology report on the renal biopsy describes extensive crescentic glomerular destruction, with inflammatory cells present.

Ten days after admission, Mr. C develops hemoptysis, and chest radiography shows increasing alveolar infiltrates. The attending internist consults pulmonary and critical care services.

The consultant suspects a pulmonary-renal syndrome because of bilateral alveolar infiltrates (diffuse alveolar hemorrhage). The internal medicine team continues high-dose corticosteroids, followed by plasmapheresis.

Brain MRI shows subacute to chronic infarcts involving the right basal ganglia and corona radiate and mild to moderate small vessel ischemic changes. Old areas of hemorrhage are noted within both cerebellar lobes, left temporal lobe, right basal ganglia, right parietal lobe, and right frontal lobe.

During follow-up interviews, Mr. C often cannot recall recent dialysis or plasmapheresis and reports no physical symptoms. His short-term memory continues to deteriorate; he would forget to eat if not cued by family or nursing staff. He shows global cognitive deficits and is increasingly withdrawn and flat.

The authors’ observations

Mr. C’s overall condition and medical test results suggest Goodpasture’s syndrome—also called anti-GBM disease—a rare autoimmune condition characterized by rapid destruction of the kidneys and hemorrhaging of the lungs (Box).1-5 We diagnose Goodpasture’s syndrome based on Mr. C’s anti-GBM antibody and pANCA findings combined with crescentic glomerular destruction on renal biopsy.

Although few case reports have associated Goodpasture’s syndrome with neurobehavioral changes, the apparent relationship of Mr. C’s medical symptoms with the worsening of his cognitive impairment suggests a link.

Mr. C’s MRI findings also might suggest CNS vasculitis, which affects small arteries of the cerebral and spinal cord leptomeninges and parenchyma, leading to CNS dys-function.6-8 CNS vasculitis can result from primary nervous system involvement or from a secondary systemic process such as Goodpasture’s syndrome.9

We rule out lupus because Mr. C is ANA-negative; this test has 99% sensitivity for lupus.10

Box

Goodpasture’s syndrome: A rare autoimmune disease

Goodpasture’s syndrome, which afflicts 1 Patients typically present with alveolar bleeding, rapidly progressive acute renal failure with proteinuria,1 and pulmonary symptoms such as dyspnea and hemoptysis.2

Possible triggers include:

  • viral upper respiratory tract infection (20% to 60% of patients)3
  • exposure to hydrocarbon solvents (3,4

Mr. C was exposed to solvents during the 15 years he worked in a factory. Some researchers believe a noxious event among genetically susceptible persons damages basement membrane and exposes an antigen that triggers IgG auto-antibody production.3,4

Malaise, weight loss, and fever are atypical in Goodpasture’s syndrome but could suggest concomitant vasculitis.5

OUTCOME: Ongoing disability

Mr. C is hospitalized for 6 weeks. He receives cyclophosphamide, prednisone, and 10 sessions of plasmapheresis. We prescribe mirtazapine, 15 mg at bedtime, to treat mood symptoms. We chose mirtazapine because of the drug’s sleep-restoring and appetite-stimulating properties.

Mr. C’s fever resolves and pulmonary function soon improves, but his cognitive impairment persists. He has difficulties preparing meals, taking medications, and managing his money.

Mr. C is discharged with a referral to a psychiatrist. He continues taking mirtazapine and a lower dose of prednisone. He requires ongoing hemodialysis and assistance with activities of daily living.

The authors’ observations

Prompt multidisciplinary intervention is critical when patients present with concurrent cognitive and medical symptoms. A thorough psychiatric evaluation can help piece together the illness’ course. The psychiatrist’s role in a multidisciplinary assessment is to:

  • document neurocognitive changes
  • verify them through collateral information
  • correlate these changes with the timing of medical symptoms.
Interview the patient alone, then seek collateral information to assemble the clinical picture and help establish premorbid functioning. Ask family members about the patient’s cognitive function at home or at work and in high-risk situations such as while driving or operating machinery.

An underlying psychiatric condition can complicate the diagnosis. In these cases, careful interviewing and collateral information can help you discern the chronology of events.

References

1. Bolton WK. Goodpasture’s syndrome. Kidney Int 1996;50(5):1753-66.

2. Pusey CD. Anti-glomerular basement membrane disease. Kidney Int 2003;64(4):1535-50.

3. Humes HD, DuPont HL. eds. Kelley’s textbook of internal medicine. 4th ed. New York, NY: Lippincott Williams & Wilkins; 2000.

4. Stevenson A, Yaqoob M, Mason H, et al. Biochemical markers of basement membrane disturbances and occupational exposure to hydrocarbons and mixed solvents. QJM 1995;88(1):23-8.

5. Kluth DC, Rees AJ. Anti-glomerular basement membrane disease. J Am Soc Nephrol. 1999;10(11):2446-53.

6. Rydel JJ, Rodby RA. An 18-year-old man with Goodpasture’s syndrome and ANCA-negative central nervous system vasculitis. Am J Kidney Dis 1998;31(2):345-9.

7. Gittins N, Basu A, Eyre J, et al. Cerebral vasculitis in a teenager with Goodpasture’s syndrome. Nephrol Dial Transplant 2004;19(12):3168-71.

8. Garnier P, Deprele C. Cerebral angiitis and Goodpasture’s syndrome. Rev Neurol 2003;159(1):68-70.

9. Calabrese LH, Duna GF, Lie JT. Vasculitis in the central nervous system. Arthritis Rhem 1997;40(7):1189-201.

10. Edworthy SM, Zatarain E, McShane DJ, Bloch DA. Analysis of the 1982 ARA lupus criteria data set by recursive partitioning methodology: new insights into the relative merit of individual criteria. J Rheumatol 1988;15(10):1493-8.

References

1. Bolton WK. Goodpasture’s syndrome. Kidney Int 1996;50(5):1753-66.

2. Pusey CD. Anti-glomerular basement membrane disease. Kidney Int 2003;64(4):1535-50.

3. Humes HD, DuPont HL. eds. Kelley’s textbook of internal medicine. 4th ed. New York, NY: Lippincott Williams & Wilkins; 2000.

4. Stevenson A, Yaqoob M, Mason H, et al. Biochemical markers of basement membrane disturbances and occupational exposure to hydrocarbons and mixed solvents. QJM 1995;88(1):23-8.

5. Kluth DC, Rees AJ. Anti-glomerular basement membrane disease. J Am Soc Nephrol. 1999;10(11):2446-53.

6. Rydel JJ, Rodby RA. An 18-year-old man with Goodpasture’s syndrome and ANCA-negative central nervous system vasculitis. Am J Kidney Dis 1998;31(2):345-9.

7. Gittins N, Basu A, Eyre J, et al. Cerebral vasculitis in a teenager with Goodpasture’s syndrome. Nephrol Dial Transplant 2004;19(12):3168-71.

8. Garnier P, Deprele C. Cerebral angiitis and Goodpasture’s syndrome. Rev Neurol 2003;159(1):68-70.

9. Calabrese LH, Duna GF, Lie JT. Vasculitis in the central nervous system. Arthritis Rhem 1997;40(7):1189-201.

10. Edworthy SM, Zatarain E, McShane DJ, Bloch DA. Analysis of the 1982 ARA lupus criteria data set by recursive partitioning methodology: new insights into the relative merit of individual criteria. J Rheumatol 1988;15(10):1493-8.

Issue
Current Psychiatry - 07(12)
Issue
Current Psychiatry - 07(12)
Page Number
49-58
Page Number
49-58
Publications
Publications
Article Type
Display Headline
A mysterious physical and mental decline
Display Headline
A mysterious physical and mental decline
Legacy Keywords
Matthew Macaluso;Christine Skotzko;Maria Rueda-Lara; Goodpasture's syndrome;Goodpasture syndrome;anti-GBM disease;medical cause of psychiatric symptoms;kidney destruction;lung hemorrhaging;CNS vasculitis
Legacy Keywords
Matthew Macaluso;Christine Skotzko;Maria Rueda-Lara; Goodpasture's syndrome;Goodpasture syndrome;anti-GBM disease;medical cause of psychiatric symptoms;kidney destruction;lung hemorrhaging;CNS vasculitis
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media