Welcome to Current Psychiatry, a leading source of information, online and in print, for practitioners of psychiatry and its related subspecialties, including addiction psychiatry, child and adolescent psychiatry, and geriatric psychiatry. This Web site contains evidence-based reviews of the prevention, diagnosis, and treatment of mental illness and psychological disorders; case reports; updates on psychopharmacology; news about the specialty of psychiatry; pearls for practice; and other topics of interest and use to this audience.

Theme
medstat_cp
Top Sections
Evidence-Based Reviews
Cases That Test Your Skills
cp

Dear Drupal User: You're seeing this because you're logged in to Drupal, and not redirected to MDedge.com/psychiatry. 

Main menu
CP Main Menu
Explore menu
CP Explore Menu
Proclivity ID
18803001
Unpublish
Negative Keywords
Bipolar depression
Depression
adolescent depression
adolescent major depressive disorder
adolescent schizophrenia
adolescent with major depressive disorder
animals
autism
baby
brexpiprazole
child
child bipolar
child depression
child schizophrenia
children with bipolar disorder
children with depression
children with major depressive disorder
compulsive behaviors
cure
elderly bipolar
elderly depression
elderly major depressive disorder
elderly schizophrenia
elderly with dementia
first break
first episode
gambling
gaming
geriatric depression
geriatric major depressive disorder
geriatric schizophrenia
infant
kid
major depressive disorder
major depressive disorder in adolescents
major depressive disorder in children
parenting
pediatric
pediatric bipolar
pediatric depression
pediatric major depressive disorder
pediatric schizophrenia
pregnancy
pregnant
rexulti
skin care
teen
wine
Negative Keywords Excluded Elements
header[@id='header']
section[contains(@class, 'nav-hidden')]
footer[@id='footer']
div[contains(@class, 'pane-pub-article-current-psychiatry')]
div[contains(@class, 'pane-pub-home-current-psychiatry')]
div[contains(@class, 'pane-pub-topic-current-psychiatry')]
div[contains(@class, 'panel-panel-inner')]
div[contains(@class, 'pane-node-field-article-topics')]
section[contains(@class, 'footer-nav-section-wrapper')]
Altmetric
DSM Affiliated
Display in offset block
QuickLearn Excluded Topics/Sections
Best Practices
CME
CME Supplements
Education Center
Medical Education Library
Disqus Exclude
Best Practices
CE/CME
Education Center
Medical Education Library
Enable Disqus
Display Author and Disclosure Link
Publication Type
Clinical
Slot System
Top 25
Disable Sticky Ads
Disable Ad Block Mitigation
Featured Buckets Admin
Publication LayerRX Default ID
798,799
Show Ads on this Publication's Homepage
Consolidated Pub
Show Article Page Numbers on TOC

The larger mosaic; Same goal different method

Article Type
Changed
Thu, 03/28/2019 - 16:18
Display Headline
The larger mosaic; Same goal different method

The larger mosaic

Dr. Nasrallah paints a frightening vision in his April editorial. There are many schools of thought in psychiatry, including biological, psychodynamic, cognitive behavioral, relational, and humanistic approaches. All of these reflect a piece of the larger mosaic that makes us human and contributes to our mental health. Psychiatry is the only medical specialty where well and broadly trained clinicians can treat patients on any or all of these levels in an integrated fashion.

The desire to redefine psychiatry as the treatment of strictly neurologically based conditions may work well for illness such as schizophrenia or bipolar I disorder, but it does a disservice to patients with anxiety, depression, trauma, etc., who can benefit from an integrative approach that may include medication and a neuroscience perspective but does not deny the healing power of approaches that work with the subjective side of mental life.

David Aftergood, MD
Private Practice
White Plains, NY

Same goal, different method

I agree with Dr. Nasrallah that psychiatry is ready for creative destruction. We differ in the best way to achieve that goal. It is not enough to revolutionize current diagnostic schemes or the disastrously dysfunctional mental health bureaucracy. My suggestion is to get psychiatry out of the pocket of pharmaceutical manufacturers who support academic psychiatry and its publications. The April 2012 issue of Current Psychiatry has 78 pages; one-half are drug advertising. If we want to revolutionize our profession I suggest we wean ourselves from our dependency on pharmaceutical manufacturers’ support, and advocate for the elimination of direct-to-consumer advertising.

Carl Hammerschlag, MD
Chief of Community Mental Health
Gesundheit! Institute
Phoenix, AZ

Dr. Nasrallah responds

I thank my colleagues for their comments on my editorial, whether supportive or dismissive. Editorials represent my opinion, sometimes critical, sometimes aspirational, but always intended to provoke healthy discourse with Current Psychiatry ’s readers. My intent in this editorial was to urge psychiatrists to continuously question what we do and whether we can practice our art differently, better, or in a more scientifically valid manner.

Regarding the issue of laboratory testing to confirm a clinical diagnosis—which many were hoping would be part of DSM-5—I have no doubt that this will become a reality in the not-too-distant future. Testing will include a mix of blood, cerebrospinal fluid, neurophysiological, or neuroimaging tests—structural, functional, spectroscopic, and diffusion tensor imaging MRI. If this sounds unlikely right now, that’s what most people thought about our ability to land on the moon a mere decade before it happened.

When it comes to the future of psychiatry, I uphold 1 mantra: yes we can and yes we will!

Henry A. Nasrallah, MD
Editor-in-Chief

Article PDF
Author and Disclosure Information

Issue
Current Psychiatry - 11(06)
Publications
Topics
Page Number
19-64
Sections
Author and Disclosure Information

Author and Disclosure Information

Article PDF
Article PDF

The larger mosaic

Dr. Nasrallah paints a frightening vision in his April editorial. There are many schools of thought in psychiatry, including biological, psychodynamic, cognitive behavioral, relational, and humanistic approaches. All of these reflect a piece of the larger mosaic that makes us human and contributes to our mental health. Psychiatry is the only medical specialty where well and broadly trained clinicians can treat patients on any or all of these levels in an integrated fashion.

The desire to redefine psychiatry as the treatment of strictly neurologically based conditions may work well for illness such as schizophrenia or bipolar I disorder, but it does a disservice to patients with anxiety, depression, trauma, etc., who can benefit from an integrative approach that may include medication and a neuroscience perspective but does not deny the healing power of approaches that work with the subjective side of mental life.

David Aftergood, MD
Private Practice
White Plains, NY

Same goal, different method

I agree with Dr. Nasrallah that psychiatry is ready for creative destruction. We differ in the best way to achieve that goal. It is not enough to revolutionize current diagnostic schemes or the disastrously dysfunctional mental health bureaucracy. My suggestion is to get psychiatry out of the pocket of pharmaceutical manufacturers who support academic psychiatry and its publications. The April 2012 issue of Current Psychiatry has 78 pages; one-half are drug advertising. If we want to revolutionize our profession I suggest we wean ourselves from our dependency on pharmaceutical manufacturers’ support, and advocate for the elimination of direct-to-consumer advertising.

Carl Hammerschlag, MD
Chief of Community Mental Health
Gesundheit! Institute
Phoenix, AZ

Dr. Nasrallah responds

I thank my colleagues for their comments on my editorial, whether supportive or dismissive. Editorials represent my opinion, sometimes critical, sometimes aspirational, but always intended to provoke healthy discourse with Current Psychiatry ’s readers. My intent in this editorial was to urge psychiatrists to continuously question what we do and whether we can practice our art differently, better, or in a more scientifically valid manner.

Regarding the issue of laboratory testing to confirm a clinical diagnosis—which many were hoping would be part of DSM-5—I have no doubt that this will become a reality in the not-too-distant future. Testing will include a mix of blood, cerebrospinal fluid, neurophysiological, or neuroimaging tests—structural, functional, spectroscopic, and diffusion tensor imaging MRI. If this sounds unlikely right now, that’s what most people thought about our ability to land on the moon a mere decade before it happened.

When it comes to the future of psychiatry, I uphold 1 mantra: yes we can and yes we will!

Henry A. Nasrallah, MD
Editor-in-Chief

The larger mosaic

Dr. Nasrallah paints a frightening vision in his April editorial. There are many schools of thought in psychiatry, including biological, psychodynamic, cognitive behavioral, relational, and humanistic approaches. All of these reflect a piece of the larger mosaic that makes us human and contributes to our mental health. Psychiatry is the only medical specialty where well and broadly trained clinicians can treat patients on any or all of these levels in an integrated fashion.

The desire to redefine psychiatry as the treatment of strictly neurologically based conditions may work well for illness such as schizophrenia or bipolar I disorder, but it does a disservice to patients with anxiety, depression, trauma, etc., who can benefit from an integrative approach that may include medication and a neuroscience perspective but does not deny the healing power of approaches that work with the subjective side of mental life.

David Aftergood, MD
Private Practice
White Plains, NY

Same goal, different method

I agree with Dr. Nasrallah that psychiatry is ready for creative destruction. We differ in the best way to achieve that goal. It is not enough to revolutionize current diagnostic schemes or the disastrously dysfunctional mental health bureaucracy. My suggestion is to get psychiatry out of the pocket of pharmaceutical manufacturers who support academic psychiatry and its publications. The April 2012 issue of Current Psychiatry has 78 pages; one-half are drug advertising. If we want to revolutionize our profession I suggest we wean ourselves from our dependency on pharmaceutical manufacturers’ support, and advocate for the elimination of direct-to-consumer advertising.

Carl Hammerschlag, MD
Chief of Community Mental Health
Gesundheit! Institute
Phoenix, AZ

Dr. Nasrallah responds

I thank my colleagues for their comments on my editorial, whether supportive or dismissive. Editorials represent my opinion, sometimes critical, sometimes aspirational, but always intended to provoke healthy discourse with Current Psychiatry ’s readers. My intent in this editorial was to urge psychiatrists to continuously question what we do and whether we can practice our art differently, better, or in a more scientifically valid manner.

Regarding the issue of laboratory testing to confirm a clinical diagnosis—which many were hoping would be part of DSM-5—I have no doubt that this will become a reality in the not-too-distant future. Testing will include a mix of blood, cerebrospinal fluid, neurophysiological, or neuroimaging tests—structural, functional, spectroscopic, and diffusion tensor imaging MRI. If this sounds unlikely right now, that’s what most people thought about our ability to land on the moon a mere decade before it happened.

When it comes to the future of psychiatry, I uphold 1 mantra: yes we can and yes we will!

Henry A. Nasrallah, MD
Editor-in-Chief

Issue
Current Psychiatry - 11(06)
Issue
Current Psychiatry - 11(06)
Page Number
19-64
Page Number
19-64
Publications
Publications
Topics
Article Type
Display Headline
The larger mosaic; Same goal different method
Display Headline
The larger mosaic; Same goal different method
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

High-dose donepezil or memantine: Next step for Alzheimer’s disease?

Article Type
Changed
Tue, 12/11/2018 - 15:04
Display Headline
High-dose donepezil or memantine: Next step for Alzheimer’s disease?

Although cholinesterase inhibitors (ChEIs) and memantine at standard doses may slow the progression of Alzheimer’s disease (AD) as assessed by cognitive, functional, and global measures, this effect is relatively modest. For the estimated 5.4 million Americans with AD1—more than one-half of whom have moderate to severe disease2—there is a great need for new approaches to slow AD progression.

High doses of donepezil or memantine may be the next step in achieving better results than standard pharmacologic treatments for AD. This article presents the possible benefits and indications for high doses of donepezil (23 mg/d) and memantine (28 mg/d) for managing moderate to severe AD and their safety and tolerability profiles.

Current treatments offer modest benefits

AD treatments comprise 2 categories: ChEIs (donepezil, rivastigmine, and galantamine) and the N-methyl-D-aspartate (NMDA) receptor antagonist memantine (Table 1).3,4 All ChEIs are FDA-approved for mild to moderate AD; donepezil also is approved for severe AD. Memantine is approved for moderate to severe AD, either alone or in combination with ChEIs. Until recently, the maximum FDA-approved doses were donepezil, 10 mg/d, and memantine, 20 mg/d. However, these dosages are associated with only modest beneficial effects in managing cognitive deterioration in patients with moderate to severe dementia.5,6 Studies have reported that combining a ChEI, such as donepezil, and memantine is well tolerated and may result in synergistic benefits by affecting different neurotransmitters in patients with moderate to severe AD.7,8

Recently, the FDA approved higher daily doses of donepezil (23 mg) and memantine (28 mg) for moderate to severe AD on the basis of positive phase III trial results.9-11 Donepezil, 23 mg/d, currently is marketed in the United States; the availability date for memantine, 28 mg/d, was undetermined at press time.

Table 1

FDA-approved treatments for Alzheimer’s disease

DrugMaximum daily doseMechanism of actionIndicationCommon side effects/comments
Tacrine160 mg/dChEIMild to moderate ADNausea, vomiting, loss of appetite, diarrhea. First ChEI to be approved, but rarely used because of associated possible hepatotoxicity
Donepezil10 mg/dChEIAll stages of ADNausea, vomiting, loss of appetite, diarrhea, sleep disturbance
Rivastigmine12 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Galantamine24 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Memantine20 mg/dNMDA receptor antagonistModerate to severe ADDizziness, headache, constipation, confusion
Galantamine ER24 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Rivastigmine transdermal system9.5 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Donepezil 2323 mg/dChEIModerate to severe ADNausea, vomiting, diarrhea
Memantine ER28 mg/dNMDA receptor antagonistModerate to severe ADDizziness, headache, constipation, confusion
AD: Alzheimer’s disease; ChEI: cholinesterase inhibitor; ER: extended release; NMDA: N-methyl-D-aspartate
Source: References 3,4

High-dose donepezil (23 mg/d)

Cognitive decline with AD has been associated with increasing loss of cholinergic neurons and cholinergic activities, particularly in areas associated with memory/cognition and learning, including cortical areas involving the temporal lobe, hippocampus, and nucleus basalis of Meynert.12-14 In addition, evidence suggests that increasing levels of acetylcholine by using ChEIs can enhance cognitive function.13,15

Donepezil is a selective, reversible ChEI believed to enhance central cholinergic function.15 Randomized clinical trials assessing dose-response with donepezil, 5 mg/d and 10 mg/d, have demonstrated more benefit in cognition with either dose than placebo. The 10 mg/d dose was more effective than 5 mg/d in patients with mild to moderate and severe AD.16-18 In patients with advanced AD who are stable on 5 mg/d, increasing to 10 mg/d could slow the progression of cognitive decline.18

Rationale for higher doses. Positron emission tomography studies have shown that at stable doses of donepezil, 5 mg/d or 10 mg/d, average cortical acetylcholinesterase (AChE) inhibition was <30%.19,20 Based on these findings, researchers thought that cortical AChE inhibition may be suboptimal with donepezil, 10 mg/d, and that higher doses of ChEI may be required in patients with more advanced AD—and therefore more cholinergic loss—for adequate cholinesterase inhibition. In a pilot study of patients with mild to moderate AD, higher doses of donepezil (15 mg/d and 20 mg/d) were reported to be safe and well tolerated.21

The 23-mg/d donepezil formulation was developed to provide a higher dose administered once daily without a sharp rise in peak concentration. The FDA approved donepezil, 23 mg/d, for patients with moderate to severe AD on the basis of phase III trial results.9,22 In a randomized, double-blind, multicenter, head-to-head clinical trial, >1,400 patients with moderate to severe AD (Mini-Mental State Exam [MMSE]: 0 to 20) on a stable dose of donepezil, 10 mg/d, for ≥3 months were randomly assigned to receive high-dose donepezil (23 mg/d) or standard-dose donepezil (10 mg/d) for 24 weeks.9,22 Patients in the 23-mg/d group showed a statistically significant improvement in cognition compared with the 10-mg/d group. The difference between groups on a measure of global improvement was not significant.9,22 However, in a post-hoc analysis, it was demonstrated that a subgroup of patients with more severe cognitive impairment (baseline MMSE: 0 to 16), showed significant improvement in cognition as well as global functioning.9

 

 

Overall, treatment-emergent adverse events (TEAEs) during the study were higher in patients receiving 23 mg/d (74%) than those receiving 10 mg/d (64%). The most common TEAEs in the 23-mg/d and 10-mg/d groups were nausea (12% vs 3%, respectively), vomiting (9% vs 3%), and diarrhea (8% vs 5%) (Table 2).22 These gastrointestinal adverse effects were more frequent during the first month of treatment and were relatively infrequent beyond 1 month. Serious TEAEs, such as falls, urinary tract infection, pneumonia, syncope, aggression, and confusional state, were noted in a similar proportion of patients in the 23-mg/d and 10-mg/d groups; most of these were considered unrelated to treatment. No drug-related deaths occurred during the study. High-dose (23 mg/d) donepezil generally was well tolerated, with a typical ChEI safety profile but superior efficacy.

A recent commentary discussed the issue of effect size and whether a 2.2-point difference on a 100-point scale (the Severe Impairment Battery [SIB]) is clinically meaningful.23 As with all anti-dementia therapies, in any cohort some patients will gain considerably more than 2.2 points on the SIB, which is clinically significant. A 6-month trial is recommended to identify these optimal responders.

Table 2

High-dose vs standard-dose donepezil: Treatment-emergent adverse events

Adverse eventDonepezil, 23 mg/dDonepezil,10 mg/d
Nausea12%3%
Vomiting9%3%
Diarrhea8%5%
Anorexia5%2%
Dizziness5%3%
Weight decrease5%3%
Headache4%3%
Insomnia3%2%
Urinary incontinence3%1%
Fatigue2%1%
Weakness2%1%
Somnolence2%1%
Contusion2%0%
Source: Reference 22

High-dose memantine

Memantine is an NMDA receptor antagonist, which works on glutamate, an ubiquitous neurotransmitter in the brain that serves many functions. For reasons that are not fully understood, in AD glutamate becomes excitotoxic and causes neuronal death.

Some researchers have hypothesized that if safe and well tolerated, a memantine dose >20 mg/d may have better efficacy than a lower dose. Memantine’s manufacturer has developed an extended-release (ER), once-daily formulation of memantine, 28 mg/d, to improve adherence and possibly increase efficacy.10,11 Because of memantine ER’s relatively slow absorption rate and longer median Tmax, of 12 hours, there is minimal fluctuation in plasma levels during steady-state dosing intervals compared with the immediate-release (IR) formulation.10

In a phase I study of 24 healthy volunteers that investigated the safety, tolerability, and pharmacokinetics of memantine ER, 28 mg/d, TEAEs were mild; the most common were headache, somnolence, and dizziness.10 During memantine treatment, there were no serious adverse events, potential significant changes in patients’ vital signs, or deaths.

Memantine ER plus ChEI. A multicenter, multinational, randomized, double-blind study compared memantine ER, 28 mg/d, and placebo in patients with moderate to severe AD (MMSE: 3 to 14).11 All patients were receiving concurrent, stable ChEI treatment (donepezil, rivastigmine, or galantamine) for ≥3 months before the study. Patients treated with memantine ER, 28 mg/d, and ChEI (n = 342) showed a significant improvement compared with the placebo/ChEI group (n = 335) in cognition and global functioning. Patients receiving memantine/ChEI also showed statistically significant benefits on behavior and verbal fluency testing compared with patients receiving placebo/ChEI. Memantine was well tolerated; most adverse events were mild or moderate. The most common adverse events in the memantine/ChEI group that occurred at a higher rate relative to the placebo/ChEI group were headache (5.6% vs 5.1%, respectively), diarrhea (5.0% vs 3.9%), and dizziness (4.7% vs 1.5%). There were no deaths related to memantine (Table 3).11

Memantine ER, 28 mg/d, may be tolerated better than the IR formulation because of less plasma level fluctuation during the steady-state dosing interval. Also, memantine ER, 28 mg/d, may offer better efficacy over memantine IR, 20 mg/d, because of dose-dependent cognitive, global, and behavioral effects. In addition, once-daily dosing of memantine ER may improve adherence compared with the IR formulation.24

In patients with severe renal impairment, dosage of memantine IR should be reduced from 20 mg/d to 10 mg/d.25 However, there is no available information regarding the dosing, safety, and tolerability of memantine ER, 28 mg/d, in patients with renal disease.

Table3

High-dose memantine: Treatment-emergent adverse eventsa

Adverse eventPlacebo (n = 335)Memantine ER (n = 341)
Any TEAE214 (63.9%)214 (62.8%)
Fall26 (7.8%)19 (5.6%)
Urinary tract infection24 (7.2%)19 (5.6%)
Headache17 (5.1%)19 (5.6%)
Diarrhea13 (3.9%)17 (5.0%)
Dizziness5 (1.5%)16 (4.7%)
Influenza9 (2.7%)15 (4.4%)
Insomnia16 (4.8%)14 (4.1%)
Agitation15 (4.5%)14 (4.1%)
Hypertension8 (2.4%)13 (3.8%)
Anxiety9 (2.7%)12 (3.5%)
Depression5 (1.5%)11 (3.2%)
Weight increased3 (0.9%)11 (3.2%)
Constipation4 (1.2%)10 (2.9%)
Somnolence4 (1.2%)10 (2.9%)
Back pain2 (0.6%)9 (2.6%)
Aggression5 (1.5%)8 (2.3%)
Hypotension5 (1.5%)7 (2.1%)
Vomiting4 (1.2%)7 (2.1%)
Abdominal pain2 (0.6%)7 (2.1%)
Nasopharyngitis10 (3.0%)6 (1.8%)
Confusional state7 (2.1%)6 (1.8%)
Weight decreased11 (3.3%)5 (1.5%)
Nausea7 (2.1%)5 (1.5%)
Irritability8 (2.4%)4 (1.2%)
Cough8 (2.4%)3 (0.9%)
aData [n (%)] include all adverse events experienced by ≥2% patients in either group (safety population). Adverse events that were experienced at twice the rate in 1 group compared with the other are indicated by bold type
ER: extended-release (28 mg); TEAE: treatment-emergent adverse event
Source: Reference 11
 

 

Recommendations

Because there are few FDA-approved treatments for AD, higher doses of donepezil or memantine may be an option for patients who have “maxed out” on their AD therapy or no longer respond to lower doses. Higher doses of donepezil (23 mg/d) and memantine (28 mg/d) could improve medication adherence because both are once-daily preparations. In clinical trials, donepezil, 23 mg/d, was more effective than donepezil, 10 mg/d.9 Whether memantine ER, 28 mg/d, is superior to memantine IR, 20 mg/d, needs to be investigated in head-to-head, double-blind, controlled studies.

For patients with moderate to severe AD, donepezil, 23 mg, is associated with greater benefits in cognition compared with donepezil, 10 mg/d.9 Similarly, because of potentially superior efficacy because of a higher dose, memantine ER, 28 mg, might best help patients with moderate to severe AD, specifically those who either don’t respond or lose response to memantine IR, 20 mg/d. Combining a ChEI, such as donepezil, with memantine is associated with slower cognitive decline and short and long-term benefits on measures of cognition, activities of daily living, global outcome, and behavior.7,26 However, additional clinical trials are needed to assess the safety, tolerability, and efficacy of combination therapy with higher doses of donepezil and memantine ER.

Related Resources

  • Alzheimer’s Disease Education and Referral Center. www.nia.nih.gov/Alzheimers.
  • Lleó A, Greenberg SM, Growdon JH. Current pharmacotherapy for Alzheimer’s disease. Annu Rev Med. 2006;57:513-533.

Drug Brand Names

  • Donepezil • Aricept
  • Galantamine • Razadyne
  • Memantine • Namenda
  • Rivastigmine • Exelon
  • Tacrine • Cognex

Disclosures

Dr. Grossberg’s academic department has received research funding from Forest Pharmaceuticals and Pfizer Inc. Dr. Grossberg has received grant/research support from Baxter BioScience, Forest Pharmaceuticals, Janssen, the National Institutes of Health, Novartis, and Pfizer, Inc.; is a consultant to Baxter BioScience, Forest Pharmaceuticals, Merck, Novartis, and Otsuka; and is on the Safety Monitoring Committee for Merck.

Dr. Singh reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Alzheimer’s Association, Thies W, Bleiler L. 2011 Alzheimer’s disease facts and figures. Alzheimers Dement. 2011;7(2):208-244.

2. Hebert LE, Scherr PA, Bienias JL, et al. Alzheimer disease in the US population: prevalence estimates using the 2000 census. Arch Neurol. 2003;60(8):1119-1122.

3. Alzheimer’s Disease Education and Referral Center. Alzheimer’s disease medications. http://www.nia.nih.gov/alzheimers/publication/alzheimers-disease-medications-fact-sheet. Accessed May 10 2012.

4. Osborn GG, Saunders AV. Current treatments for patients with Alzheimer disease. J Am Osteopath Assoc. 2010;110(9 suppl 8):S16-S26.

5. Raina P, Santaguida P, Ismaila A, et al. Effectiveness of cholinesterase inhibitors and memantine for treating dementia: evidence review for a clinical practice guideline. Ann Intern Med. 2008;148(5):379-397.

6. Cummings JL. Alzheimer’s disease. N Engl J Med. 2004;351(1):56-67.

7. Tariot PN, Farlow MR, Grossberg GT, et al. Memantine Study Group. Memantine treatment in patients with moderate to severe Alzheimer disease already receiving donepezil: a randomized controlled trial. JAMA. 2004;291(3):317-324.

8. Xiong G, Doraiswamy PM. Combination drug therapy for Alzheimer’s disease: what is evidence-based and what is not? Geriatrics. 2005;60(6):22-26.

9. Farlow MR, Salloway S, Tariot PN, et al. Effectiveness and tolerability of high (23 mg/d) versus standard-dose (10 mg/d) donepezil in moderate to severe Alzheimer’s disease: a 24-week, randomized, double-blind study. Clin Ther. 2010;32(7):1234-1251.

10. Periclou A, Hu Y. Extended-release memantine capsule (28 mg once daily): a multiple dose, open-label study evaluating steady-state pharmacokinetics in healthy volunteers. Poster presented at 11th International Conference on Alzheimer’s Disease; July 26-31, 2008; Chicago, IL.

11. Grossberg GT, Manes F, Allegri R, et al. A multinational, randomized, double-blind, placebo-controlled, parallel-group trial of memantine extended-release capsule (28 mg, once daily) in patients with moderate to severe Alzheimer’s disease. Poster presented at 11th International Conference on Alzheimer’s Disease; July 26-31, 2008; Chicago, IL.

12. Geula C, Mesulam MM. Systematic regional variations in the loss of cortical cholinergic fibers in Alzheimer’s disease. Cereb Cortex. 1996;6(2):165-177.

13. Whitehouse PJ. The cholinergic deficit in Alzheimer’s disease. J Clin Psychiatry. 1998;59(suppl 13):19-22.

14. Teipel SJ, Flatz WH, Heinsen H, et al. Measurement of basal forebrain atrophy in Alzheimer’s disease using MRI. Brain. 2005;128(11):2626-2644.

15. Shintani EY, Uchida KM. Donepezil: an anticholinesterase inhibitor for Alzheimer’s disease. Am J Health Syst Pharm. 1997;54(24):2805-2810.

16. Homma A, Imai Y, Tago H, et al. Donepezil treatment of patients with severe Alzheimer’s disease in a Japanese population: results from a 24-week, double-blind, placebo-controlled, randomized trial. Dement Geriatr Cogn Disord. 2008;25(5):399-407.

17. Whitehead A, Perdomo C, Pratt RD, et al. Donepezil for the symptomatic treatment of patients with mild to moderate Alzheimer’s disease: a meta-analysis of individual patient data from randomised controlled trials. Int J Geriatr Psychiatry. 2004;19(7):624-633.

18. Nozawa M, Ichimiya Y, Nozawa E, et al. Clinical effects of high oral dose of donepezil for patients with Alzheimer’s disease in Japan. Psychogeriatrics. 2009;9(2):50-55.

19. Kuhl DE, Minoshima S, Frey KA, et al. Limited donepezil inhibition of acetylcholinesterase measured with positron emission tomography in living Alzheimer cerebral cortex. Ann Neurol. 2000;48(3):391-395.

20. Bohnen NI, Kaufer DI, Hendrickson R, et al. Degree of inhibition of cortical acetylcholinesterase activity and cognitive effects by donepezil treatment in Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 2005;76(3):315-319.

21. Doody RS, Corey-Bloom J, Zhang R, et al. Safety and tolerability of donepezil at doses up to 20 mg/day: results from a pilot study in patients with Alzheimer’s disease. Drugs Aging. 2008;25(2):163-174.

22. Aricept [package insert]. Woodcliff Lake NJ: Eisai Co.; 2012.

23. Schwartz LM, Woloshin S. How the FDA forgot the evidence: the case of donepezil 23 mg. BMJ. 2012;344:e1086.-doi: 10.1136/bmj.e1086.

24. Saini SD, Schoenfeld P, Kaulback K, et al. Effect of medication dosing frequency on adherence in chronic diseases. Am J Manag Care. 2009;15(6):e22-e33.

25. Periclou A, Ventura D, Rao N, et al. Pharmacokinetic study of memantine in healthy and renally impaired subjects. Clin Pharmacol Ther. 2006;79(1):134-143.

26. Atri A, Shaughnessy LW, Locascio JJ, et al. Long-term course and effectiveness of combination therapy in Alzheimer disease. Alzheimer Dis Assoc Disord. 2008;22(3):209-221.

Article PDF
Author and Disclosure Information

Indrapal Singh, MD
Assistant Professor, Department of Geriatric Mental Health, Chhattrapati Shahuji Maharaj Medical University, Lucknow, Uttar Pradesh, India
George T. Grossberg, MD
Samuel W. Fordyce Professor, Department of Neurology and Psychiatry, Division of Geriatric Psychiatry, Saint Louis University School of Medicine, St. Louis, MO

Issue
Current Psychiatry - 11(06)
Publications
Topics
Page Number
20-29
Sections
Author and Disclosure Information

Indrapal Singh, MD
Assistant Professor, Department of Geriatric Mental Health, Chhattrapati Shahuji Maharaj Medical University, Lucknow, Uttar Pradesh, India
George T. Grossberg, MD
Samuel W. Fordyce Professor, Department of Neurology and Psychiatry, Division of Geriatric Psychiatry, Saint Louis University School of Medicine, St. Louis, MO

Author and Disclosure Information

Indrapal Singh, MD
Assistant Professor, Department of Geriatric Mental Health, Chhattrapati Shahuji Maharaj Medical University, Lucknow, Uttar Pradesh, India
George T. Grossberg, MD
Samuel W. Fordyce Professor, Department of Neurology and Psychiatry, Division of Geriatric Psychiatry, Saint Louis University School of Medicine, St. Louis, MO

Article PDF
Article PDF

Although cholinesterase inhibitors (ChEIs) and memantine at standard doses may slow the progression of Alzheimer’s disease (AD) as assessed by cognitive, functional, and global measures, this effect is relatively modest. For the estimated 5.4 million Americans with AD1—more than one-half of whom have moderate to severe disease2—there is a great need for new approaches to slow AD progression.

High doses of donepezil or memantine may be the next step in achieving better results than standard pharmacologic treatments for AD. This article presents the possible benefits and indications for high doses of donepezil (23 mg/d) and memantine (28 mg/d) for managing moderate to severe AD and their safety and tolerability profiles.

Current treatments offer modest benefits

AD treatments comprise 2 categories: ChEIs (donepezil, rivastigmine, and galantamine) and the N-methyl-D-aspartate (NMDA) receptor antagonist memantine (Table 1).3,4 All ChEIs are FDA-approved for mild to moderate AD; donepezil also is approved for severe AD. Memantine is approved for moderate to severe AD, either alone or in combination with ChEIs. Until recently, the maximum FDA-approved doses were donepezil, 10 mg/d, and memantine, 20 mg/d. However, these dosages are associated with only modest beneficial effects in managing cognitive deterioration in patients with moderate to severe dementia.5,6 Studies have reported that combining a ChEI, such as donepezil, and memantine is well tolerated and may result in synergistic benefits by affecting different neurotransmitters in patients with moderate to severe AD.7,8

Recently, the FDA approved higher daily doses of donepezil (23 mg) and memantine (28 mg) for moderate to severe AD on the basis of positive phase III trial results.9-11 Donepezil, 23 mg/d, currently is marketed in the United States; the availability date for memantine, 28 mg/d, was undetermined at press time.

Table 1

FDA-approved treatments for Alzheimer’s disease

DrugMaximum daily doseMechanism of actionIndicationCommon side effects/comments
Tacrine160 mg/dChEIMild to moderate ADNausea, vomiting, loss of appetite, diarrhea. First ChEI to be approved, but rarely used because of associated possible hepatotoxicity
Donepezil10 mg/dChEIAll stages of ADNausea, vomiting, loss of appetite, diarrhea, sleep disturbance
Rivastigmine12 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Galantamine24 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Memantine20 mg/dNMDA receptor antagonistModerate to severe ADDizziness, headache, constipation, confusion
Galantamine ER24 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Rivastigmine transdermal system9.5 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Donepezil 2323 mg/dChEIModerate to severe ADNausea, vomiting, diarrhea
Memantine ER28 mg/dNMDA receptor antagonistModerate to severe ADDizziness, headache, constipation, confusion
AD: Alzheimer’s disease; ChEI: cholinesterase inhibitor; ER: extended release; NMDA: N-methyl-D-aspartate
Source: References 3,4

High-dose donepezil (23 mg/d)

Cognitive decline with AD has been associated with increasing loss of cholinergic neurons and cholinergic activities, particularly in areas associated with memory/cognition and learning, including cortical areas involving the temporal lobe, hippocampus, and nucleus basalis of Meynert.12-14 In addition, evidence suggests that increasing levels of acetylcholine by using ChEIs can enhance cognitive function.13,15

Donepezil is a selective, reversible ChEI believed to enhance central cholinergic function.15 Randomized clinical trials assessing dose-response with donepezil, 5 mg/d and 10 mg/d, have demonstrated more benefit in cognition with either dose than placebo. The 10 mg/d dose was more effective than 5 mg/d in patients with mild to moderate and severe AD.16-18 In patients with advanced AD who are stable on 5 mg/d, increasing to 10 mg/d could slow the progression of cognitive decline.18

Rationale for higher doses. Positron emission tomography studies have shown that at stable doses of donepezil, 5 mg/d or 10 mg/d, average cortical acetylcholinesterase (AChE) inhibition was <30%.19,20 Based on these findings, researchers thought that cortical AChE inhibition may be suboptimal with donepezil, 10 mg/d, and that higher doses of ChEI may be required in patients with more advanced AD—and therefore more cholinergic loss—for adequate cholinesterase inhibition. In a pilot study of patients with mild to moderate AD, higher doses of donepezil (15 mg/d and 20 mg/d) were reported to be safe and well tolerated.21

The 23-mg/d donepezil formulation was developed to provide a higher dose administered once daily without a sharp rise in peak concentration. The FDA approved donepezil, 23 mg/d, for patients with moderate to severe AD on the basis of phase III trial results.9,22 In a randomized, double-blind, multicenter, head-to-head clinical trial, >1,400 patients with moderate to severe AD (Mini-Mental State Exam [MMSE]: 0 to 20) on a stable dose of donepezil, 10 mg/d, for ≥3 months were randomly assigned to receive high-dose donepezil (23 mg/d) or standard-dose donepezil (10 mg/d) for 24 weeks.9,22 Patients in the 23-mg/d group showed a statistically significant improvement in cognition compared with the 10-mg/d group. The difference between groups on a measure of global improvement was not significant.9,22 However, in a post-hoc analysis, it was demonstrated that a subgroup of patients with more severe cognitive impairment (baseline MMSE: 0 to 16), showed significant improvement in cognition as well as global functioning.9

 

 

Overall, treatment-emergent adverse events (TEAEs) during the study were higher in patients receiving 23 mg/d (74%) than those receiving 10 mg/d (64%). The most common TEAEs in the 23-mg/d and 10-mg/d groups were nausea (12% vs 3%, respectively), vomiting (9% vs 3%), and diarrhea (8% vs 5%) (Table 2).22 These gastrointestinal adverse effects were more frequent during the first month of treatment and were relatively infrequent beyond 1 month. Serious TEAEs, such as falls, urinary tract infection, pneumonia, syncope, aggression, and confusional state, were noted in a similar proportion of patients in the 23-mg/d and 10-mg/d groups; most of these were considered unrelated to treatment. No drug-related deaths occurred during the study. High-dose (23 mg/d) donepezil generally was well tolerated, with a typical ChEI safety profile but superior efficacy.

A recent commentary discussed the issue of effect size and whether a 2.2-point difference on a 100-point scale (the Severe Impairment Battery [SIB]) is clinically meaningful.23 As with all anti-dementia therapies, in any cohort some patients will gain considerably more than 2.2 points on the SIB, which is clinically significant. A 6-month trial is recommended to identify these optimal responders.

Table 2

High-dose vs standard-dose donepezil: Treatment-emergent adverse events

Adverse eventDonepezil, 23 mg/dDonepezil,10 mg/d
Nausea12%3%
Vomiting9%3%
Diarrhea8%5%
Anorexia5%2%
Dizziness5%3%
Weight decrease5%3%
Headache4%3%
Insomnia3%2%
Urinary incontinence3%1%
Fatigue2%1%
Weakness2%1%
Somnolence2%1%
Contusion2%0%
Source: Reference 22

High-dose memantine

Memantine is an NMDA receptor antagonist, which works on glutamate, an ubiquitous neurotransmitter in the brain that serves many functions. For reasons that are not fully understood, in AD glutamate becomes excitotoxic and causes neuronal death.

Some researchers have hypothesized that if safe and well tolerated, a memantine dose >20 mg/d may have better efficacy than a lower dose. Memantine’s manufacturer has developed an extended-release (ER), once-daily formulation of memantine, 28 mg/d, to improve adherence and possibly increase efficacy.10,11 Because of memantine ER’s relatively slow absorption rate and longer median Tmax, of 12 hours, there is minimal fluctuation in plasma levels during steady-state dosing intervals compared with the immediate-release (IR) formulation.10

In a phase I study of 24 healthy volunteers that investigated the safety, tolerability, and pharmacokinetics of memantine ER, 28 mg/d, TEAEs were mild; the most common were headache, somnolence, and dizziness.10 During memantine treatment, there were no serious adverse events, potential significant changes in patients’ vital signs, or deaths.

Memantine ER plus ChEI. A multicenter, multinational, randomized, double-blind study compared memantine ER, 28 mg/d, and placebo in patients with moderate to severe AD (MMSE: 3 to 14).11 All patients were receiving concurrent, stable ChEI treatment (donepezil, rivastigmine, or galantamine) for ≥3 months before the study. Patients treated with memantine ER, 28 mg/d, and ChEI (n = 342) showed a significant improvement compared with the placebo/ChEI group (n = 335) in cognition and global functioning. Patients receiving memantine/ChEI also showed statistically significant benefits on behavior and verbal fluency testing compared with patients receiving placebo/ChEI. Memantine was well tolerated; most adverse events were mild or moderate. The most common adverse events in the memantine/ChEI group that occurred at a higher rate relative to the placebo/ChEI group were headache (5.6% vs 5.1%, respectively), diarrhea (5.0% vs 3.9%), and dizziness (4.7% vs 1.5%). There were no deaths related to memantine (Table 3).11

Memantine ER, 28 mg/d, may be tolerated better than the IR formulation because of less plasma level fluctuation during the steady-state dosing interval. Also, memantine ER, 28 mg/d, may offer better efficacy over memantine IR, 20 mg/d, because of dose-dependent cognitive, global, and behavioral effects. In addition, once-daily dosing of memantine ER may improve adherence compared with the IR formulation.24

In patients with severe renal impairment, dosage of memantine IR should be reduced from 20 mg/d to 10 mg/d.25 However, there is no available information regarding the dosing, safety, and tolerability of memantine ER, 28 mg/d, in patients with renal disease.

Table3

High-dose memantine: Treatment-emergent adverse eventsa

Adverse eventPlacebo (n = 335)Memantine ER (n = 341)
Any TEAE214 (63.9%)214 (62.8%)
Fall26 (7.8%)19 (5.6%)
Urinary tract infection24 (7.2%)19 (5.6%)
Headache17 (5.1%)19 (5.6%)
Diarrhea13 (3.9%)17 (5.0%)
Dizziness5 (1.5%)16 (4.7%)
Influenza9 (2.7%)15 (4.4%)
Insomnia16 (4.8%)14 (4.1%)
Agitation15 (4.5%)14 (4.1%)
Hypertension8 (2.4%)13 (3.8%)
Anxiety9 (2.7%)12 (3.5%)
Depression5 (1.5%)11 (3.2%)
Weight increased3 (0.9%)11 (3.2%)
Constipation4 (1.2%)10 (2.9%)
Somnolence4 (1.2%)10 (2.9%)
Back pain2 (0.6%)9 (2.6%)
Aggression5 (1.5%)8 (2.3%)
Hypotension5 (1.5%)7 (2.1%)
Vomiting4 (1.2%)7 (2.1%)
Abdominal pain2 (0.6%)7 (2.1%)
Nasopharyngitis10 (3.0%)6 (1.8%)
Confusional state7 (2.1%)6 (1.8%)
Weight decreased11 (3.3%)5 (1.5%)
Nausea7 (2.1%)5 (1.5%)
Irritability8 (2.4%)4 (1.2%)
Cough8 (2.4%)3 (0.9%)
aData [n (%)] include all adverse events experienced by ≥2% patients in either group (safety population). Adverse events that were experienced at twice the rate in 1 group compared with the other are indicated by bold type
ER: extended-release (28 mg); TEAE: treatment-emergent adverse event
Source: Reference 11
 

 

Recommendations

Because there are few FDA-approved treatments for AD, higher doses of donepezil or memantine may be an option for patients who have “maxed out” on their AD therapy or no longer respond to lower doses. Higher doses of donepezil (23 mg/d) and memantine (28 mg/d) could improve medication adherence because both are once-daily preparations. In clinical trials, donepezil, 23 mg/d, was more effective than donepezil, 10 mg/d.9 Whether memantine ER, 28 mg/d, is superior to memantine IR, 20 mg/d, needs to be investigated in head-to-head, double-blind, controlled studies.

For patients with moderate to severe AD, donepezil, 23 mg, is associated with greater benefits in cognition compared with donepezil, 10 mg/d.9 Similarly, because of potentially superior efficacy because of a higher dose, memantine ER, 28 mg, might best help patients with moderate to severe AD, specifically those who either don’t respond or lose response to memantine IR, 20 mg/d. Combining a ChEI, such as donepezil, with memantine is associated with slower cognitive decline and short and long-term benefits on measures of cognition, activities of daily living, global outcome, and behavior.7,26 However, additional clinical trials are needed to assess the safety, tolerability, and efficacy of combination therapy with higher doses of donepezil and memantine ER.

Related Resources

  • Alzheimer’s Disease Education and Referral Center. www.nia.nih.gov/Alzheimers.
  • Lleó A, Greenberg SM, Growdon JH. Current pharmacotherapy for Alzheimer’s disease. Annu Rev Med. 2006;57:513-533.

Drug Brand Names

  • Donepezil • Aricept
  • Galantamine • Razadyne
  • Memantine • Namenda
  • Rivastigmine • Exelon
  • Tacrine • Cognex

Disclosures

Dr. Grossberg’s academic department has received research funding from Forest Pharmaceuticals and Pfizer Inc. Dr. Grossberg has received grant/research support from Baxter BioScience, Forest Pharmaceuticals, Janssen, the National Institutes of Health, Novartis, and Pfizer, Inc.; is a consultant to Baxter BioScience, Forest Pharmaceuticals, Merck, Novartis, and Otsuka; and is on the Safety Monitoring Committee for Merck.

Dr. Singh reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Although cholinesterase inhibitors (ChEIs) and memantine at standard doses may slow the progression of Alzheimer’s disease (AD) as assessed by cognitive, functional, and global measures, this effect is relatively modest. For the estimated 5.4 million Americans with AD1—more than one-half of whom have moderate to severe disease2—there is a great need for new approaches to slow AD progression.

High doses of donepezil or memantine may be the next step in achieving better results than standard pharmacologic treatments for AD. This article presents the possible benefits and indications for high doses of donepezil (23 mg/d) and memantine (28 mg/d) for managing moderate to severe AD and their safety and tolerability profiles.

Current treatments offer modest benefits

AD treatments comprise 2 categories: ChEIs (donepezil, rivastigmine, and galantamine) and the N-methyl-D-aspartate (NMDA) receptor antagonist memantine (Table 1).3,4 All ChEIs are FDA-approved for mild to moderate AD; donepezil also is approved for severe AD. Memantine is approved for moderate to severe AD, either alone or in combination with ChEIs. Until recently, the maximum FDA-approved doses were donepezil, 10 mg/d, and memantine, 20 mg/d. However, these dosages are associated with only modest beneficial effects in managing cognitive deterioration in patients with moderate to severe dementia.5,6 Studies have reported that combining a ChEI, such as donepezil, and memantine is well tolerated and may result in synergistic benefits by affecting different neurotransmitters in patients with moderate to severe AD.7,8

Recently, the FDA approved higher daily doses of donepezil (23 mg) and memantine (28 mg) for moderate to severe AD on the basis of positive phase III trial results.9-11 Donepezil, 23 mg/d, currently is marketed in the United States; the availability date for memantine, 28 mg/d, was undetermined at press time.

Table 1

FDA-approved treatments for Alzheimer’s disease

DrugMaximum daily doseMechanism of actionIndicationCommon side effects/comments
Tacrine160 mg/dChEIMild to moderate ADNausea, vomiting, loss of appetite, diarrhea. First ChEI to be approved, but rarely used because of associated possible hepatotoxicity
Donepezil10 mg/dChEIAll stages of ADNausea, vomiting, loss of appetite, diarrhea, sleep disturbance
Rivastigmine12 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Galantamine24 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Memantine20 mg/dNMDA receptor antagonistModerate to severe ADDizziness, headache, constipation, confusion
Galantamine ER24 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Rivastigmine transdermal system9.5 mg/dChEIMild to moderate ADNausea, vomiting, diarrhea, weight loss, loss of appetite
Donepezil 2323 mg/dChEIModerate to severe ADNausea, vomiting, diarrhea
Memantine ER28 mg/dNMDA receptor antagonistModerate to severe ADDizziness, headache, constipation, confusion
AD: Alzheimer’s disease; ChEI: cholinesterase inhibitor; ER: extended release; NMDA: N-methyl-D-aspartate
Source: References 3,4

High-dose donepezil (23 mg/d)

Cognitive decline with AD has been associated with increasing loss of cholinergic neurons and cholinergic activities, particularly in areas associated with memory/cognition and learning, including cortical areas involving the temporal lobe, hippocampus, and nucleus basalis of Meynert.12-14 In addition, evidence suggests that increasing levels of acetylcholine by using ChEIs can enhance cognitive function.13,15

Donepezil is a selective, reversible ChEI believed to enhance central cholinergic function.15 Randomized clinical trials assessing dose-response with donepezil, 5 mg/d and 10 mg/d, have demonstrated more benefit in cognition with either dose than placebo. The 10 mg/d dose was more effective than 5 mg/d in patients with mild to moderate and severe AD.16-18 In patients with advanced AD who are stable on 5 mg/d, increasing to 10 mg/d could slow the progression of cognitive decline.18

Rationale for higher doses. Positron emission tomography studies have shown that at stable doses of donepezil, 5 mg/d or 10 mg/d, average cortical acetylcholinesterase (AChE) inhibition was <30%.19,20 Based on these findings, researchers thought that cortical AChE inhibition may be suboptimal with donepezil, 10 mg/d, and that higher doses of ChEI may be required in patients with more advanced AD—and therefore more cholinergic loss—for adequate cholinesterase inhibition. In a pilot study of patients with mild to moderate AD, higher doses of donepezil (15 mg/d and 20 mg/d) were reported to be safe and well tolerated.21

The 23-mg/d donepezil formulation was developed to provide a higher dose administered once daily without a sharp rise in peak concentration. The FDA approved donepezil, 23 mg/d, for patients with moderate to severe AD on the basis of phase III trial results.9,22 In a randomized, double-blind, multicenter, head-to-head clinical trial, >1,400 patients with moderate to severe AD (Mini-Mental State Exam [MMSE]: 0 to 20) on a stable dose of donepezil, 10 mg/d, for ≥3 months were randomly assigned to receive high-dose donepezil (23 mg/d) or standard-dose donepezil (10 mg/d) for 24 weeks.9,22 Patients in the 23-mg/d group showed a statistically significant improvement in cognition compared with the 10-mg/d group. The difference between groups on a measure of global improvement was not significant.9,22 However, in a post-hoc analysis, it was demonstrated that a subgroup of patients with more severe cognitive impairment (baseline MMSE: 0 to 16), showed significant improvement in cognition as well as global functioning.9

 

 

Overall, treatment-emergent adverse events (TEAEs) during the study were higher in patients receiving 23 mg/d (74%) than those receiving 10 mg/d (64%). The most common TEAEs in the 23-mg/d and 10-mg/d groups were nausea (12% vs 3%, respectively), vomiting (9% vs 3%), and diarrhea (8% vs 5%) (Table 2).22 These gastrointestinal adverse effects were more frequent during the first month of treatment and were relatively infrequent beyond 1 month. Serious TEAEs, such as falls, urinary tract infection, pneumonia, syncope, aggression, and confusional state, were noted in a similar proportion of patients in the 23-mg/d and 10-mg/d groups; most of these were considered unrelated to treatment. No drug-related deaths occurred during the study. High-dose (23 mg/d) donepezil generally was well tolerated, with a typical ChEI safety profile but superior efficacy.

A recent commentary discussed the issue of effect size and whether a 2.2-point difference on a 100-point scale (the Severe Impairment Battery [SIB]) is clinically meaningful.23 As with all anti-dementia therapies, in any cohort some patients will gain considerably more than 2.2 points on the SIB, which is clinically significant. A 6-month trial is recommended to identify these optimal responders.

Table 2

High-dose vs standard-dose donepezil: Treatment-emergent adverse events

Adverse eventDonepezil, 23 mg/dDonepezil,10 mg/d
Nausea12%3%
Vomiting9%3%
Diarrhea8%5%
Anorexia5%2%
Dizziness5%3%
Weight decrease5%3%
Headache4%3%
Insomnia3%2%
Urinary incontinence3%1%
Fatigue2%1%
Weakness2%1%
Somnolence2%1%
Contusion2%0%
Source: Reference 22

High-dose memantine

Memantine is an NMDA receptor antagonist, which works on glutamate, an ubiquitous neurotransmitter in the brain that serves many functions. For reasons that are not fully understood, in AD glutamate becomes excitotoxic and causes neuronal death.

Some researchers have hypothesized that if safe and well tolerated, a memantine dose >20 mg/d may have better efficacy than a lower dose. Memantine’s manufacturer has developed an extended-release (ER), once-daily formulation of memantine, 28 mg/d, to improve adherence and possibly increase efficacy.10,11 Because of memantine ER’s relatively slow absorption rate and longer median Tmax, of 12 hours, there is minimal fluctuation in plasma levels during steady-state dosing intervals compared with the immediate-release (IR) formulation.10

In a phase I study of 24 healthy volunteers that investigated the safety, tolerability, and pharmacokinetics of memantine ER, 28 mg/d, TEAEs were mild; the most common were headache, somnolence, and dizziness.10 During memantine treatment, there were no serious adverse events, potential significant changes in patients’ vital signs, or deaths.

Memantine ER plus ChEI. A multicenter, multinational, randomized, double-blind study compared memantine ER, 28 mg/d, and placebo in patients with moderate to severe AD (MMSE: 3 to 14).11 All patients were receiving concurrent, stable ChEI treatment (donepezil, rivastigmine, or galantamine) for ≥3 months before the study. Patients treated with memantine ER, 28 mg/d, and ChEI (n = 342) showed a significant improvement compared with the placebo/ChEI group (n = 335) in cognition and global functioning. Patients receiving memantine/ChEI also showed statistically significant benefits on behavior and verbal fluency testing compared with patients receiving placebo/ChEI. Memantine was well tolerated; most adverse events were mild or moderate. The most common adverse events in the memantine/ChEI group that occurred at a higher rate relative to the placebo/ChEI group were headache (5.6% vs 5.1%, respectively), diarrhea (5.0% vs 3.9%), and dizziness (4.7% vs 1.5%). There were no deaths related to memantine (Table 3).11

Memantine ER, 28 mg/d, may be tolerated better than the IR formulation because of less plasma level fluctuation during the steady-state dosing interval. Also, memantine ER, 28 mg/d, may offer better efficacy over memantine IR, 20 mg/d, because of dose-dependent cognitive, global, and behavioral effects. In addition, once-daily dosing of memantine ER may improve adherence compared with the IR formulation.24

In patients with severe renal impairment, dosage of memantine IR should be reduced from 20 mg/d to 10 mg/d.25 However, there is no available information regarding the dosing, safety, and tolerability of memantine ER, 28 mg/d, in patients with renal disease.

Table3

High-dose memantine: Treatment-emergent adverse eventsa

Adverse eventPlacebo (n = 335)Memantine ER (n = 341)
Any TEAE214 (63.9%)214 (62.8%)
Fall26 (7.8%)19 (5.6%)
Urinary tract infection24 (7.2%)19 (5.6%)
Headache17 (5.1%)19 (5.6%)
Diarrhea13 (3.9%)17 (5.0%)
Dizziness5 (1.5%)16 (4.7%)
Influenza9 (2.7%)15 (4.4%)
Insomnia16 (4.8%)14 (4.1%)
Agitation15 (4.5%)14 (4.1%)
Hypertension8 (2.4%)13 (3.8%)
Anxiety9 (2.7%)12 (3.5%)
Depression5 (1.5%)11 (3.2%)
Weight increased3 (0.9%)11 (3.2%)
Constipation4 (1.2%)10 (2.9%)
Somnolence4 (1.2%)10 (2.9%)
Back pain2 (0.6%)9 (2.6%)
Aggression5 (1.5%)8 (2.3%)
Hypotension5 (1.5%)7 (2.1%)
Vomiting4 (1.2%)7 (2.1%)
Abdominal pain2 (0.6%)7 (2.1%)
Nasopharyngitis10 (3.0%)6 (1.8%)
Confusional state7 (2.1%)6 (1.8%)
Weight decreased11 (3.3%)5 (1.5%)
Nausea7 (2.1%)5 (1.5%)
Irritability8 (2.4%)4 (1.2%)
Cough8 (2.4%)3 (0.9%)
aData [n (%)] include all adverse events experienced by ≥2% patients in either group (safety population). Adverse events that were experienced at twice the rate in 1 group compared with the other are indicated by bold type
ER: extended-release (28 mg); TEAE: treatment-emergent adverse event
Source: Reference 11
 

 

Recommendations

Because there are few FDA-approved treatments for AD, higher doses of donepezil or memantine may be an option for patients who have “maxed out” on their AD therapy or no longer respond to lower doses. Higher doses of donepezil (23 mg/d) and memantine (28 mg/d) could improve medication adherence because both are once-daily preparations. In clinical trials, donepezil, 23 mg/d, was more effective than donepezil, 10 mg/d.9 Whether memantine ER, 28 mg/d, is superior to memantine IR, 20 mg/d, needs to be investigated in head-to-head, double-blind, controlled studies.

For patients with moderate to severe AD, donepezil, 23 mg, is associated with greater benefits in cognition compared with donepezil, 10 mg/d.9 Similarly, because of potentially superior efficacy because of a higher dose, memantine ER, 28 mg, might best help patients with moderate to severe AD, specifically those who either don’t respond or lose response to memantine IR, 20 mg/d. Combining a ChEI, such as donepezil, with memantine is associated with slower cognitive decline and short and long-term benefits on measures of cognition, activities of daily living, global outcome, and behavior.7,26 However, additional clinical trials are needed to assess the safety, tolerability, and efficacy of combination therapy with higher doses of donepezil and memantine ER.

Related Resources

  • Alzheimer’s Disease Education and Referral Center. www.nia.nih.gov/Alzheimers.
  • Lleó A, Greenberg SM, Growdon JH. Current pharmacotherapy for Alzheimer’s disease. Annu Rev Med. 2006;57:513-533.

Drug Brand Names

  • Donepezil • Aricept
  • Galantamine • Razadyne
  • Memantine • Namenda
  • Rivastigmine • Exelon
  • Tacrine • Cognex

Disclosures

Dr. Grossberg’s academic department has received research funding from Forest Pharmaceuticals and Pfizer Inc. Dr. Grossberg has received grant/research support from Baxter BioScience, Forest Pharmaceuticals, Janssen, the National Institutes of Health, Novartis, and Pfizer, Inc.; is a consultant to Baxter BioScience, Forest Pharmaceuticals, Merck, Novartis, and Otsuka; and is on the Safety Monitoring Committee for Merck.

Dr. Singh reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Alzheimer’s Association, Thies W, Bleiler L. 2011 Alzheimer’s disease facts and figures. Alzheimers Dement. 2011;7(2):208-244.

2. Hebert LE, Scherr PA, Bienias JL, et al. Alzheimer disease in the US population: prevalence estimates using the 2000 census. Arch Neurol. 2003;60(8):1119-1122.

3. Alzheimer’s Disease Education and Referral Center. Alzheimer’s disease medications. http://www.nia.nih.gov/alzheimers/publication/alzheimers-disease-medications-fact-sheet. Accessed May 10 2012.

4. Osborn GG, Saunders AV. Current treatments for patients with Alzheimer disease. J Am Osteopath Assoc. 2010;110(9 suppl 8):S16-S26.

5. Raina P, Santaguida P, Ismaila A, et al. Effectiveness of cholinesterase inhibitors and memantine for treating dementia: evidence review for a clinical practice guideline. Ann Intern Med. 2008;148(5):379-397.

6. Cummings JL. Alzheimer’s disease. N Engl J Med. 2004;351(1):56-67.

7. Tariot PN, Farlow MR, Grossberg GT, et al. Memantine Study Group. Memantine treatment in patients with moderate to severe Alzheimer disease already receiving donepezil: a randomized controlled trial. JAMA. 2004;291(3):317-324.

8. Xiong G, Doraiswamy PM. Combination drug therapy for Alzheimer’s disease: what is evidence-based and what is not? Geriatrics. 2005;60(6):22-26.

9. Farlow MR, Salloway S, Tariot PN, et al. Effectiveness and tolerability of high (23 mg/d) versus standard-dose (10 mg/d) donepezil in moderate to severe Alzheimer’s disease: a 24-week, randomized, double-blind study. Clin Ther. 2010;32(7):1234-1251.

10. Periclou A, Hu Y. Extended-release memantine capsule (28 mg once daily): a multiple dose, open-label study evaluating steady-state pharmacokinetics in healthy volunteers. Poster presented at 11th International Conference on Alzheimer’s Disease; July 26-31, 2008; Chicago, IL.

11. Grossberg GT, Manes F, Allegri R, et al. A multinational, randomized, double-blind, placebo-controlled, parallel-group trial of memantine extended-release capsule (28 mg, once daily) in patients with moderate to severe Alzheimer’s disease. Poster presented at 11th International Conference on Alzheimer’s Disease; July 26-31, 2008; Chicago, IL.

12. Geula C, Mesulam MM. Systematic regional variations in the loss of cortical cholinergic fibers in Alzheimer’s disease. Cereb Cortex. 1996;6(2):165-177.

13. Whitehouse PJ. The cholinergic deficit in Alzheimer’s disease. J Clin Psychiatry. 1998;59(suppl 13):19-22.

14. Teipel SJ, Flatz WH, Heinsen H, et al. Measurement of basal forebrain atrophy in Alzheimer’s disease using MRI. Brain. 2005;128(11):2626-2644.

15. Shintani EY, Uchida KM. Donepezil: an anticholinesterase inhibitor for Alzheimer’s disease. Am J Health Syst Pharm. 1997;54(24):2805-2810.

16. Homma A, Imai Y, Tago H, et al. Donepezil treatment of patients with severe Alzheimer’s disease in a Japanese population: results from a 24-week, double-blind, placebo-controlled, randomized trial. Dement Geriatr Cogn Disord. 2008;25(5):399-407.

17. Whitehead A, Perdomo C, Pratt RD, et al. Donepezil for the symptomatic treatment of patients with mild to moderate Alzheimer’s disease: a meta-analysis of individual patient data from randomised controlled trials. Int J Geriatr Psychiatry. 2004;19(7):624-633.

18. Nozawa M, Ichimiya Y, Nozawa E, et al. Clinical effects of high oral dose of donepezil for patients with Alzheimer’s disease in Japan. Psychogeriatrics. 2009;9(2):50-55.

19. Kuhl DE, Minoshima S, Frey KA, et al. Limited donepezil inhibition of acetylcholinesterase measured with positron emission tomography in living Alzheimer cerebral cortex. Ann Neurol. 2000;48(3):391-395.

20. Bohnen NI, Kaufer DI, Hendrickson R, et al. Degree of inhibition of cortical acetylcholinesterase activity and cognitive effects by donepezil treatment in Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 2005;76(3):315-319.

21. Doody RS, Corey-Bloom J, Zhang R, et al. Safety and tolerability of donepezil at doses up to 20 mg/day: results from a pilot study in patients with Alzheimer’s disease. Drugs Aging. 2008;25(2):163-174.

22. Aricept [package insert]. Woodcliff Lake NJ: Eisai Co.; 2012.

23. Schwartz LM, Woloshin S. How the FDA forgot the evidence: the case of donepezil 23 mg. BMJ. 2012;344:e1086.-doi: 10.1136/bmj.e1086.

24. Saini SD, Schoenfeld P, Kaulback K, et al. Effect of medication dosing frequency on adherence in chronic diseases. Am J Manag Care. 2009;15(6):e22-e33.

25. Periclou A, Ventura D, Rao N, et al. Pharmacokinetic study of memantine in healthy and renally impaired subjects. Clin Pharmacol Ther. 2006;79(1):134-143.

26. Atri A, Shaughnessy LW, Locascio JJ, et al. Long-term course and effectiveness of combination therapy in Alzheimer disease. Alzheimer Dis Assoc Disord. 2008;22(3):209-221.

References

1. Alzheimer’s Association, Thies W, Bleiler L. 2011 Alzheimer’s disease facts and figures. Alzheimers Dement. 2011;7(2):208-244.

2. Hebert LE, Scherr PA, Bienias JL, et al. Alzheimer disease in the US population: prevalence estimates using the 2000 census. Arch Neurol. 2003;60(8):1119-1122.

3. Alzheimer’s Disease Education and Referral Center. Alzheimer’s disease medications. http://www.nia.nih.gov/alzheimers/publication/alzheimers-disease-medications-fact-sheet. Accessed May 10 2012.

4. Osborn GG, Saunders AV. Current treatments for patients with Alzheimer disease. J Am Osteopath Assoc. 2010;110(9 suppl 8):S16-S26.

5. Raina P, Santaguida P, Ismaila A, et al. Effectiveness of cholinesterase inhibitors and memantine for treating dementia: evidence review for a clinical practice guideline. Ann Intern Med. 2008;148(5):379-397.

6. Cummings JL. Alzheimer’s disease. N Engl J Med. 2004;351(1):56-67.

7. Tariot PN, Farlow MR, Grossberg GT, et al. Memantine Study Group. Memantine treatment in patients with moderate to severe Alzheimer disease already receiving donepezil: a randomized controlled trial. JAMA. 2004;291(3):317-324.

8. Xiong G, Doraiswamy PM. Combination drug therapy for Alzheimer’s disease: what is evidence-based and what is not? Geriatrics. 2005;60(6):22-26.

9. Farlow MR, Salloway S, Tariot PN, et al. Effectiveness and tolerability of high (23 mg/d) versus standard-dose (10 mg/d) donepezil in moderate to severe Alzheimer’s disease: a 24-week, randomized, double-blind study. Clin Ther. 2010;32(7):1234-1251.

10. Periclou A, Hu Y. Extended-release memantine capsule (28 mg once daily): a multiple dose, open-label study evaluating steady-state pharmacokinetics in healthy volunteers. Poster presented at 11th International Conference on Alzheimer’s Disease; July 26-31, 2008; Chicago, IL.

11. Grossberg GT, Manes F, Allegri R, et al. A multinational, randomized, double-blind, placebo-controlled, parallel-group trial of memantine extended-release capsule (28 mg, once daily) in patients with moderate to severe Alzheimer’s disease. Poster presented at 11th International Conference on Alzheimer’s Disease; July 26-31, 2008; Chicago, IL.

12. Geula C, Mesulam MM. Systematic regional variations in the loss of cortical cholinergic fibers in Alzheimer’s disease. Cereb Cortex. 1996;6(2):165-177.

13. Whitehouse PJ. The cholinergic deficit in Alzheimer’s disease. J Clin Psychiatry. 1998;59(suppl 13):19-22.

14. Teipel SJ, Flatz WH, Heinsen H, et al. Measurement of basal forebrain atrophy in Alzheimer’s disease using MRI. Brain. 2005;128(11):2626-2644.

15. Shintani EY, Uchida KM. Donepezil: an anticholinesterase inhibitor for Alzheimer’s disease. Am J Health Syst Pharm. 1997;54(24):2805-2810.

16. Homma A, Imai Y, Tago H, et al. Donepezil treatment of patients with severe Alzheimer’s disease in a Japanese population: results from a 24-week, double-blind, placebo-controlled, randomized trial. Dement Geriatr Cogn Disord. 2008;25(5):399-407.

17. Whitehead A, Perdomo C, Pratt RD, et al. Donepezil for the symptomatic treatment of patients with mild to moderate Alzheimer’s disease: a meta-analysis of individual patient data from randomised controlled trials. Int J Geriatr Psychiatry. 2004;19(7):624-633.

18. Nozawa M, Ichimiya Y, Nozawa E, et al. Clinical effects of high oral dose of donepezil for patients with Alzheimer’s disease in Japan. Psychogeriatrics. 2009;9(2):50-55.

19. Kuhl DE, Minoshima S, Frey KA, et al. Limited donepezil inhibition of acetylcholinesterase measured with positron emission tomography in living Alzheimer cerebral cortex. Ann Neurol. 2000;48(3):391-395.

20. Bohnen NI, Kaufer DI, Hendrickson R, et al. Degree of inhibition of cortical acetylcholinesterase activity and cognitive effects by donepezil treatment in Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 2005;76(3):315-319.

21. Doody RS, Corey-Bloom J, Zhang R, et al. Safety and tolerability of donepezil at doses up to 20 mg/day: results from a pilot study in patients with Alzheimer’s disease. Drugs Aging. 2008;25(2):163-174.

22. Aricept [package insert]. Woodcliff Lake NJ: Eisai Co.; 2012.

23. Schwartz LM, Woloshin S. How the FDA forgot the evidence: the case of donepezil 23 mg. BMJ. 2012;344:e1086.-doi: 10.1136/bmj.e1086.

24. Saini SD, Schoenfeld P, Kaulback K, et al. Effect of medication dosing frequency on adherence in chronic diseases. Am J Manag Care. 2009;15(6):e22-e33.

25. Periclou A, Ventura D, Rao N, et al. Pharmacokinetic study of memantine in healthy and renally impaired subjects. Clin Pharmacol Ther. 2006;79(1):134-143.

26. Atri A, Shaughnessy LW, Locascio JJ, et al. Long-term course and effectiveness of combination therapy in Alzheimer disease. Alzheimer Dis Assoc Disord. 2008;22(3):209-221.

Issue
Current Psychiatry - 11(06)
Issue
Current Psychiatry - 11(06)
Page Number
20-29
Page Number
20-29
Publications
Publications
Topics
Article Type
Display Headline
High-dose donepezil or memantine: Next step for Alzheimer’s disease?
Display Headline
High-dose donepezil or memantine: Next step for Alzheimer’s disease?
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Does bupropion exacerbate anxiety?

Article Type
Changed
Fri, 01/24/2020 - 11:00
Display Headline
Does bupropion exacerbate anxiety?

For many clinicians, bupropion is the “go-to” medication for treating depressed patients who smoke, have concerns about sexual dysfunction side effects, and/or worry about weight gain. Bupropion is FDA-approved for preventing seasonal major depressive episodes in patients with seasonal affective disorder and is indicated as a smoking cessation aid.

“Anxious depression”—defined as depression with high levels of anxiety—is associated with poorer outcomes than “non-anxious” depression.1 Prescribing medications for these patients can be challenging. Some clinicians believe that bupropion exacerbates anxiety and should not be used to treat patients who experience both anxiety and depression.

Reports from our patients and our cumulative clinical experience are key factors in developing expertise in selecting appropriate medications. When informing our patients about what to expect from medications, however, it can be useful to combine anecdotal evidence with knowledge of the facts or lack thereof. Are there data to support or contradict the idea that bupropion can cause anxiety while treating depression?

What the research shows

The drug manufacturer reports a “substantial proportion of patients treated with Wellbutrin experience some degree of increased restlessness, agitation, anxiety, and insomnia, especially shortly after initiation of treatment.”2

In 2001, Rush et al3 published the results of a 16-week study (n=248) assessing pre-treatment anxiety levels and response to sertraline or bupropion. The authors concluded that anxious and depressed patients who received sertraline didn’t experience a superior anxiolytic or antidepressant response compared with bupropion.3 The same authors came to similar conclusions in a retrospective analysis of a pair of 8-week randomized, controlled, double-blind trials of selective serotonin reuptake inhibitors (SSRIs) and bupropion.4

In 2001, Nieuwstraten et al5 compared bupropion with SSRIs for treating depression by reviewing several randomized, double-blind, controlled trials. The relative risk of developing “anxiety/agitation” was 1.32 (95% confidence interval, 0.85 to 2.04), which was not statistically significant.

In a 2008 meta-analysis, Papakostas et al6 pooled individual patient data from 10 randomized, double-blind, placebo-controlled trials. Their aim was to compare the efficacy of bupropion to SSRIs in treating “anxious depression.” They found no difference in timing or degree of improvement in anxiety symptoms between groups based on Hamilton Anxiety Scale or Hamilton Depression Rating Scale—Anxiety-Somatization (HDRS-AS) scores. The authors recommended that antidepressant choice should not be based on concerns about worsening anxiety symptoms in depressed patients.6

Another meta-analysis by Papakostas et al7 of the same 10 randomized, double-blind, placebo-controlled trials suggested SSRIs may confer an advantage over bupropion in treating a subset of patients with “anxious depression,” which they defined as a HDRS-AS score ≥7. The authors noted the advantage was statistically significant, although “modest.”

Other smaller studies suggest that bupropion does not increase anxiety.8,9 A pilot study (N = 24, no placebo control) concluded that bupropion XL was comparable to escitalopram in treating anxiety in outpatients with generalized anxiety disorder.8

Because designing and executing drug trials can be expensive, it is not surprising that most of the evidence cited above derives from pharmaceutical company-sponsored or industry-affiliated work. As such, we should evaluate available evidence within the context of what we hear from and observe in our patients.

Our opinion

When assessing patients with depression and anxiety, we must carefully evaluate symptoms to distinguish between depression with associated anxiety symptoms and depression with a comorbid anxiety disorder.

If a patient suffers from depression with associated anxiety symptoms (“anxious depression”), keep in mind that although some data demonstrate a superior response to SSRIs, other studies show no difference in effect. Some research—albeit smaller, less compelling studies—suggests that bupropion may decrease anxiety.

If your patient suffers from comorbid depression and an anxiety disorder, bupropion would not be a first-line choice because it is not FDA-approved to treat anxiety disorders. Although it is possible that anxiety/agitation could result from bupropion use, there is not sufficient data to support its reputation as ”anxiogenic.”

What is your experience?

Do you agree with the authors? Send comments to [email protected] or share your thoughts on http://www.facebook.com/CurrentPsychiatry.

 

Related Resource

 

  • American Psychiatric Association. Mixed anxiety-depressive disorder. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington, DC: American Psychiatric Association; 2000:780-781.

Drug Brand Names

 

  • Bupropion • Wellbutrin, Zyban
  • Escitalopram • Lexapro
  • Sertraline • Zoloft

Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

 

1. Fava M, Rush AJ, Alpert JE, et al. Difference in treatment outcome in outpatients with anxious versus nonanxious depression: a STAR*D report. Am J Psychiatry. 2008;165(3):342-351.

2. Wellbutrin [package insert]. Research Triangle Park NC: GlaxoSmithKline; 2008.

3. Rush AJ, Trivedi MH, Carmody TJ, et al. Response in relation to baseline anxiety levels in major depressive disorder treated with bupropion sustained release or sertraline. Neuropsychopharmacology. 2001;25(1):131-138.

4. Trivedi MH, Rush AJ, Carmody TJ, et al. Do bupropion SR and sertraline differ in their effects on anxiety in depressed patients? J Clin Psychiatry. 2001;62(10):776-781.

5. Nieuwstraten CE, Dolovich LR. Bupropion versus selective serotonin-reuptake inhibitors for treatment of depression. Ann Pharmacother. 2001;35(12):1608-1613.

6. Papakostas GI, Trivedi MH, Alpert JE, et al. Efficacy of bupropion and the selective serotonin reuptake inhibitors in the treatment of anxiety symptoms in major depressive disorder: a meta-analysis of individual patient data from 10 double-blind, randomized clinical trials. J Psychiatr Res. 2008;42(2):134-140.

7. Papakostas GI, Stahl SM, Krishen A, et al. Efficacy of bupropion and the selective serotonin reuptake inhibitors in the treatment of major depressive disorder with high levels of anxiety (anxious depression): a pooled analysis of 10 studies. J Clin Psychiatry. 2008;69(8):1287-1292.

8. Bystritsky A, Kerwin L, Feusner JD, et al. A pilot controlled trial of bupropion XL versus escitalopram in generalized anxiety disorder. Psychopharmacol Bull. 2008;41(1):46-51.

9. Feighner JP, Gardner EA, Johnston JA, et al. Double-blind comparison of bupropion and fluoxetine in depressed outpatients. J Clin Psychiatry. 1991;52(8):329-335.

Article PDF
Author and Disclosure Information

 

Courtney Nemeth Wiseman, MD, MPH
Dr. Wiseman is Instructor, Clinical, Contributing Services Faculty, Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL

Jessica L. Gören, PharmD, BCPP
Dr. Gören is Associate Professor, Department of Pharmacy Practice, University of Rhode Island, Kingston, RI; Senior Clinical Pharmacist Specialist, Department of Pharmacy, Cambridge Health Alliance; and Instructor in Psychiatry, Harvard Medical School, Boston, MA

Issue
Current Psychiatry - 11(06)
Publications
Topics
Page Number
E3-E4
Sections
Author and Disclosure Information

 

Courtney Nemeth Wiseman, MD, MPH
Dr. Wiseman is Instructor, Clinical, Contributing Services Faculty, Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL

Jessica L. Gören, PharmD, BCPP
Dr. Gören is Associate Professor, Department of Pharmacy Practice, University of Rhode Island, Kingston, RI; Senior Clinical Pharmacist Specialist, Department of Pharmacy, Cambridge Health Alliance; and Instructor in Psychiatry, Harvard Medical School, Boston, MA

Author and Disclosure Information

 

Courtney Nemeth Wiseman, MD, MPH
Dr. Wiseman is Instructor, Clinical, Contributing Services Faculty, Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL

Jessica L. Gören, PharmD, BCPP
Dr. Gören is Associate Professor, Department of Pharmacy Practice, University of Rhode Island, Kingston, RI; Senior Clinical Pharmacist Specialist, Department of Pharmacy, Cambridge Health Alliance; and Instructor in Psychiatry, Harvard Medical School, Boston, MA

Article PDF
Article PDF

For many clinicians, bupropion is the “go-to” medication for treating depressed patients who smoke, have concerns about sexual dysfunction side effects, and/or worry about weight gain. Bupropion is FDA-approved for preventing seasonal major depressive episodes in patients with seasonal affective disorder and is indicated as a smoking cessation aid.

“Anxious depression”—defined as depression with high levels of anxiety—is associated with poorer outcomes than “non-anxious” depression.1 Prescribing medications for these patients can be challenging. Some clinicians believe that bupropion exacerbates anxiety and should not be used to treat patients who experience both anxiety and depression.

Reports from our patients and our cumulative clinical experience are key factors in developing expertise in selecting appropriate medications. When informing our patients about what to expect from medications, however, it can be useful to combine anecdotal evidence with knowledge of the facts or lack thereof. Are there data to support or contradict the idea that bupropion can cause anxiety while treating depression?

What the research shows

The drug manufacturer reports a “substantial proportion of patients treated with Wellbutrin experience some degree of increased restlessness, agitation, anxiety, and insomnia, especially shortly after initiation of treatment.”2

In 2001, Rush et al3 published the results of a 16-week study (n=248) assessing pre-treatment anxiety levels and response to sertraline or bupropion. The authors concluded that anxious and depressed patients who received sertraline didn’t experience a superior anxiolytic or antidepressant response compared with bupropion.3 The same authors came to similar conclusions in a retrospective analysis of a pair of 8-week randomized, controlled, double-blind trials of selective serotonin reuptake inhibitors (SSRIs) and bupropion.4

In 2001, Nieuwstraten et al5 compared bupropion with SSRIs for treating depression by reviewing several randomized, double-blind, controlled trials. The relative risk of developing “anxiety/agitation” was 1.32 (95% confidence interval, 0.85 to 2.04), which was not statistically significant.

In a 2008 meta-analysis, Papakostas et al6 pooled individual patient data from 10 randomized, double-blind, placebo-controlled trials. Their aim was to compare the efficacy of bupropion to SSRIs in treating “anxious depression.” They found no difference in timing or degree of improvement in anxiety symptoms between groups based on Hamilton Anxiety Scale or Hamilton Depression Rating Scale—Anxiety-Somatization (HDRS-AS) scores. The authors recommended that antidepressant choice should not be based on concerns about worsening anxiety symptoms in depressed patients.6

Another meta-analysis by Papakostas et al7 of the same 10 randomized, double-blind, placebo-controlled trials suggested SSRIs may confer an advantage over bupropion in treating a subset of patients with “anxious depression,” which they defined as a HDRS-AS score ≥7. The authors noted the advantage was statistically significant, although “modest.”

Other smaller studies suggest that bupropion does not increase anxiety.8,9 A pilot study (N = 24, no placebo control) concluded that bupropion XL was comparable to escitalopram in treating anxiety in outpatients with generalized anxiety disorder.8

Because designing and executing drug trials can be expensive, it is not surprising that most of the evidence cited above derives from pharmaceutical company-sponsored or industry-affiliated work. As such, we should evaluate available evidence within the context of what we hear from and observe in our patients.

Our opinion

When assessing patients with depression and anxiety, we must carefully evaluate symptoms to distinguish between depression with associated anxiety symptoms and depression with a comorbid anxiety disorder.

If a patient suffers from depression with associated anxiety symptoms (“anxious depression”), keep in mind that although some data demonstrate a superior response to SSRIs, other studies show no difference in effect. Some research—albeit smaller, less compelling studies—suggests that bupropion may decrease anxiety.

If your patient suffers from comorbid depression and an anxiety disorder, bupropion would not be a first-line choice because it is not FDA-approved to treat anxiety disorders. Although it is possible that anxiety/agitation could result from bupropion use, there is not sufficient data to support its reputation as ”anxiogenic.”

What is your experience?

Do you agree with the authors? Send comments to [email protected] or share your thoughts on http://www.facebook.com/CurrentPsychiatry.

 

Related Resource

 

  • American Psychiatric Association. Mixed anxiety-depressive disorder. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington, DC: American Psychiatric Association; 2000:780-781.

Drug Brand Names

 

  • Bupropion • Wellbutrin, Zyban
  • Escitalopram • Lexapro
  • Sertraline • Zoloft

Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

For many clinicians, bupropion is the “go-to” medication for treating depressed patients who smoke, have concerns about sexual dysfunction side effects, and/or worry about weight gain. Bupropion is FDA-approved for preventing seasonal major depressive episodes in patients with seasonal affective disorder and is indicated as a smoking cessation aid.

“Anxious depression”—defined as depression with high levels of anxiety—is associated with poorer outcomes than “non-anxious” depression.1 Prescribing medications for these patients can be challenging. Some clinicians believe that bupropion exacerbates anxiety and should not be used to treat patients who experience both anxiety and depression.

Reports from our patients and our cumulative clinical experience are key factors in developing expertise in selecting appropriate medications. When informing our patients about what to expect from medications, however, it can be useful to combine anecdotal evidence with knowledge of the facts or lack thereof. Are there data to support or contradict the idea that bupropion can cause anxiety while treating depression?

What the research shows

The drug manufacturer reports a “substantial proportion of patients treated with Wellbutrin experience some degree of increased restlessness, agitation, anxiety, and insomnia, especially shortly after initiation of treatment.”2

In 2001, Rush et al3 published the results of a 16-week study (n=248) assessing pre-treatment anxiety levels and response to sertraline or bupropion. The authors concluded that anxious and depressed patients who received sertraline didn’t experience a superior anxiolytic or antidepressant response compared with bupropion.3 The same authors came to similar conclusions in a retrospective analysis of a pair of 8-week randomized, controlled, double-blind trials of selective serotonin reuptake inhibitors (SSRIs) and bupropion.4

In 2001, Nieuwstraten et al5 compared bupropion with SSRIs for treating depression by reviewing several randomized, double-blind, controlled trials. The relative risk of developing “anxiety/agitation” was 1.32 (95% confidence interval, 0.85 to 2.04), which was not statistically significant.

In a 2008 meta-analysis, Papakostas et al6 pooled individual patient data from 10 randomized, double-blind, placebo-controlled trials. Their aim was to compare the efficacy of bupropion to SSRIs in treating “anxious depression.” They found no difference in timing or degree of improvement in anxiety symptoms between groups based on Hamilton Anxiety Scale or Hamilton Depression Rating Scale—Anxiety-Somatization (HDRS-AS) scores. The authors recommended that antidepressant choice should not be based on concerns about worsening anxiety symptoms in depressed patients.6

Another meta-analysis by Papakostas et al7 of the same 10 randomized, double-blind, placebo-controlled trials suggested SSRIs may confer an advantage over bupropion in treating a subset of patients with “anxious depression,” which they defined as a HDRS-AS score ≥7. The authors noted the advantage was statistically significant, although “modest.”

Other smaller studies suggest that bupropion does not increase anxiety.8,9 A pilot study (N = 24, no placebo control) concluded that bupropion XL was comparable to escitalopram in treating anxiety in outpatients with generalized anxiety disorder.8

Because designing and executing drug trials can be expensive, it is not surprising that most of the evidence cited above derives from pharmaceutical company-sponsored or industry-affiliated work. As such, we should evaluate available evidence within the context of what we hear from and observe in our patients.

Our opinion

When assessing patients with depression and anxiety, we must carefully evaluate symptoms to distinguish between depression with associated anxiety symptoms and depression with a comorbid anxiety disorder.

If a patient suffers from depression with associated anxiety symptoms (“anxious depression”), keep in mind that although some data demonstrate a superior response to SSRIs, other studies show no difference in effect. Some research—albeit smaller, less compelling studies—suggests that bupropion may decrease anxiety.

If your patient suffers from comorbid depression and an anxiety disorder, bupropion would not be a first-line choice because it is not FDA-approved to treat anxiety disorders. Although it is possible that anxiety/agitation could result from bupropion use, there is not sufficient data to support its reputation as ”anxiogenic.”

What is your experience?

Do you agree with the authors? Send comments to [email protected] or share your thoughts on http://www.facebook.com/CurrentPsychiatry.

 

Related Resource

 

  • American Psychiatric Association. Mixed anxiety-depressive disorder. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington, DC: American Psychiatric Association; 2000:780-781.

Drug Brand Names

 

  • Bupropion • Wellbutrin, Zyban
  • Escitalopram • Lexapro
  • Sertraline • Zoloft

Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

 

1. Fava M, Rush AJ, Alpert JE, et al. Difference in treatment outcome in outpatients with anxious versus nonanxious depression: a STAR*D report. Am J Psychiatry. 2008;165(3):342-351.

2. Wellbutrin [package insert]. Research Triangle Park NC: GlaxoSmithKline; 2008.

3. Rush AJ, Trivedi MH, Carmody TJ, et al. Response in relation to baseline anxiety levels in major depressive disorder treated with bupropion sustained release or sertraline. Neuropsychopharmacology. 2001;25(1):131-138.

4. Trivedi MH, Rush AJ, Carmody TJ, et al. Do bupropion SR and sertraline differ in their effects on anxiety in depressed patients? J Clin Psychiatry. 2001;62(10):776-781.

5. Nieuwstraten CE, Dolovich LR. Bupropion versus selective serotonin-reuptake inhibitors for treatment of depression. Ann Pharmacother. 2001;35(12):1608-1613.

6. Papakostas GI, Trivedi MH, Alpert JE, et al. Efficacy of bupropion and the selective serotonin reuptake inhibitors in the treatment of anxiety symptoms in major depressive disorder: a meta-analysis of individual patient data from 10 double-blind, randomized clinical trials. J Psychiatr Res. 2008;42(2):134-140.

7. Papakostas GI, Stahl SM, Krishen A, et al. Efficacy of bupropion and the selective serotonin reuptake inhibitors in the treatment of major depressive disorder with high levels of anxiety (anxious depression): a pooled analysis of 10 studies. J Clin Psychiatry. 2008;69(8):1287-1292.

8. Bystritsky A, Kerwin L, Feusner JD, et al. A pilot controlled trial of bupropion XL versus escitalopram in generalized anxiety disorder. Psychopharmacol Bull. 2008;41(1):46-51.

9. Feighner JP, Gardner EA, Johnston JA, et al. Double-blind comparison of bupropion and fluoxetine in depressed outpatients. J Clin Psychiatry. 1991;52(8):329-335.

References

 

1. Fava M, Rush AJ, Alpert JE, et al. Difference in treatment outcome in outpatients with anxious versus nonanxious depression: a STAR*D report. Am J Psychiatry. 2008;165(3):342-351.

2. Wellbutrin [package insert]. Research Triangle Park NC: GlaxoSmithKline; 2008.

3. Rush AJ, Trivedi MH, Carmody TJ, et al. Response in relation to baseline anxiety levels in major depressive disorder treated with bupropion sustained release or sertraline. Neuropsychopharmacology. 2001;25(1):131-138.

4. Trivedi MH, Rush AJ, Carmody TJ, et al. Do bupropion SR and sertraline differ in their effects on anxiety in depressed patients? J Clin Psychiatry. 2001;62(10):776-781.

5. Nieuwstraten CE, Dolovich LR. Bupropion versus selective serotonin-reuptake inhibitors for treatment of depression. Ann Pharmacother. 2001;35(12):1608-1613.

6. Papakostas GI, Trivedi MH, Alpert JE, et al. Efficacy of bupropion and the selective serotonin reuptake inhibitors in the treatment of anxiety symptoms in major depressive disorder: a meta-analysis of individual patient data from 10 double-blind, randomized clinical trials. J Psychiatr Res. 2008;42(2):134-140.

7. Papakostas GI, Stahl SM, Krishen A, et al. Efficacy of bupropion and the selective serotonin reuptake inhibitors in the treatment of major depressive disorder with high levels of anxiety (anxious depression): a pooled analysis of 10 studies. J Clin Psychiatry. 2008;69(8):1287-1292.

8. Bystritsky A, Kerwin L, Feusner JD, et al. A pilot controlled trial of bupropion XL versus escitalopram in generalized anxiety disorder. Psychopharmacol Bull. 2008;41(1):46-51.

9. Feighner JP, Gardner EA, Johnston JA, et al. Double-blind comparison of bupropion and fluoxetine in depressed outpatients. J Clin Psychiatry. 1991;52(8):329-335.

Issue
Current Psychiatry - 11(06)
Issue
Current Psychiatry - 11(06)
Page Number
E3-E4
Page Number
E3-E4
Publications
Publications
Topics
Article Type
Display Headline
Does bupropion exacerbate anxiety?
Display Headline
Does bupropion exacerbate anxiety?
Sections
PURLs Copyright

Disallow All Ads
Alternative CME
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Innovative approaches to treatment-resistant depression

Article Type
Changed
Mon, 04/16/2018 - 14:27
Display Headline
Innovative approaches to treatment-resistant depression

Depression treatment seems to be in a “funk” these days. Armchair critics, some of whom have never treated a depressed patient, are flooding the media and Internet with allegations that antidepressants—which have helped patients for decades—are no better than placebo.

Several major pharmaceutical companies have pared down or shut down their CNS discovery, research, and development operations. There is a lack of new antidepressants with novel mechanisms of action, and the proportion of refractory or treatment-resistant depression (TRD) seems to be growing. In short, the status of clinical depression seems rather depressing.

Yet nothing can be further from the truth! A vibrant set of innovative, even radical, solutions for TRD are on the horizon. Here are some of the paradigm shifts in the neurobiology and therapeutics of major depression that gradually are toppling decades-old tenets and creeds related to this serious psychiatric disorder:

From genes vs environment to gene × environment interaction. For decades, it was assumed that some people have “endogenous” depression due to genetic determinants, while others are afflicted with “exogenous” depression caused by stressful life events. The new model shows that the environment interacts with genes to produce depression, and that having only risk genes or only environmental stress does not necessarily lead to depression.1

From ‘chemical imbalance’ to ‘inflammatory process.’ Evidence is accumulating that inflammation may be underpinning depression,2 and studies show levels of inflammatory cytokines and interleukins rise during a depressive episode and decline when the depression remits.

From ‘neurotransmitters’ to ‘neuroplasticity and neurotropic factors.’ Current treatments were developed to increase neurotransmitter activity in the brain, but new research reveals that depression is associated with a significant drop in neurotropic factors such as brain derived neurotropic factor (BDNF) or fibroblast growth factor with a concomitant decline in hippocampal neurogenesis.3

From serotonin, norepinephrine, and dopamine to glutamate. Over the past few years, glutamate pathways and the glutamate N-methyl-D-aspartate (NMDA) receptor have emerged as possibly of central importance to the neurobiology of depression.4 The link between the strong therapeutic effects of antagonizing the NMDA receptor in depression and the increase in BDNF and neuroplasticity has emerged as a fresh model of depression.

From pills to intravenous infusions. Studies have shown that a single infusion of the NMDA receptor antagonist ketamine produces a very robust response, including full remission, in treatment–resistant unipolar or bipolar depression within 1 to 2 hours!5 The mechanism of action has been attributed to a surge of BDNF and immediate neuroplastic changes4 following NMDA receptor blockade. This abrupt reversal of severe depression, like turning on a switch, is a total and pleasant surprise.

From monotherapy to augmentation strategies. The rather imprudent notion that a single medication can be effective in a heterogeneous spectrum of disorders such as depression gradually is yielding to intelligent polypharmacy, using evidence-based augmentation strategies that might include lithium, thyroid hormone, another antidepressant (particularly mirtazapine), atypical antipsychotics, anti-inflammatory agents (including omega-3 fatty acids), antioxidants (especially N-acetylcysteine), L-methylfolate, and exercise.

From pharmacotherapy to neuromodulation. Because they disseminate to all organs and not just the brain, medications can cause undesirable side effects. Neuromodulation is used to treat depression by stimulating specific brain regions. Electroconvulsive therapy has a tarnished image but well established efficacy in severe depression. Repetitive transcranial magnetic stimulation and vagus nerve stimulation are FDA-approved for treating depression, while other forms of neuromodulation, such as cranial electrical stimulation, epidural cortical stimulation, focused ultrasound, low field magnetic stimulation, magnetic seizure therapy, near infrared light therapy, and transcranial direct current stimulation, still are in development.6 Deep brain stimulation (DBS) has been shown in several recent studies to reverse TRD,7,8 especially when stimulating the subgenual anterior cingulated region. In the future, DBS may become as commonly used in depression as it currently is in Parkinson’s disease.

A remarkable transformation is underway to reinvent the causes and treatments of depression that will reignite optimism about what psychiatry can do and eliminate the disability associated with major depression. Our patients can hardly wait.

References

 

1. Raison CL, Lowry CA, Rook GA. Inflammation, sanitation, and consternation: loss of contact with coevolved, tolerogenic microorganisms and the pathophysiology and treatment of major depression. Arch Gen Psychiatry. 2010;67(12):1211-1224.

2. Caspi A, Sugden K, Moffitt TE, et al. Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science. 2003;301(5631):386-389.

3. Duman RS, Monteggia LM. A neurotrophic model for stress-related mood disorders. Biol Psychiatry. 2006;59(12):1116-1127.

4. Li N, Lee B, Liu RJ, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science. 2010;329(5994):959-964.

5. Zarate CA, Jr, Singh JB, Carlson PJ, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63(8):856-864.

6. Rosa MA, Lisanby SH. Somatic treatments for mood disorders. Neuropsychopharmacology. 2012;37(1):102-116.

7. Mayberg HS, Lozano AM, Voon V, et al. Deep brain stimulation for treatment-resistant depression. Neuron. 2005;45(5):651-660.

8. Holtzheimer PE, Kelley ME, Gross RE, et al. Subcallosal cingulate deep brain stimulation for treatment-resistant unipolar and bipolar depression. Arch Gen Psychiatry. 2012;69(2):150-158.

Article PDF
Author and Disclosure Information

 


Henry A. Nasrallah, MD
Editor-in-Chief
To comment on this editorial or other topics of interest, visit http://www.facebook.com/CurrentPsychiatry, or click on the “Send Letters” link.

Issue
Current Psychiatry - 11(06)
Publications
Topics
Page Number
4-5
Sections
Author and Disclosure Information

 


Henry A. Nasrallah, MD
Editor-in-Chief
To comment on this editorial or other topics of interest, visit http://www.facebook.com/CurrentPsychiatry, or click on the “Send Letters” link.

Author and Disclosure Information

 


Henry A. Nasrallah, MD
Editor-in-Chief
To comment on this editorial or other topics of interest, visit http://www.facebook.com/CurrentPsychiatry, or click on the “Send Letters” link.

Article PDF
Article PDF

Depression treatment seems to be in a “funk” these days. Armchair critics, some of whom have never treated a depressed patient, are flooding the media and Internet with allegations that antidepressants—which have helped patients for decades—are no better than placebo.

Several major pharmaceutical companies have pared down or shut down their CNS discovery, research, and development operations. There is a lack of new antidepressants with novel mechanisms of action, and the proportion of refractory or treatment-resistant depression (TRD) seems to be growing. In short, the status of clinical depression seems rather depressing.

Yet nothing can be further from the truth! A vibrant set of innovative, even radical, solutions for TRD are on the horizon. Here are some of the paradigm shifts in the neurobiology and therapeutics of major depression that gradually are toppling decades-old tenets and creeds related to this serious psychiatric disorder:

From genes vs environment to gene × environment interaction. For decades, it was assumed that some people have “endogenous” depression due to genetic determinants, while others are afflicted with “exogenous” depression caused by stressful life events. The new model shows that the environment interacts with genes to produce depression, and that having only risk genes or only environmental stress does not necessarily lead to depression.1

From ‘chemical imbalance’ to ‘inflammatory process.’ Evidence is accumulating that inflammation may be underpinning depression,2 and studies show levels of inflammatory cytokines and interleukins rise during a depressive episode and decline when the depression remits.

From ‘neurotransmitters’ to ‘neuroplasticity and neurotropic factors.’ Current treatments were developed to increase neurotransmitter activity in the brain, but new research reveals that depression is associated with a significant drop in neurotropic factors such as brain derived neurotropic factor (BDNF) or fibroblast growth factor with a concomitant decline in hippocampal neurogenesis.3

From serotonin, norepinephrine, and dopamine to glutamate. Over the past few years, glutamate pathways and the glutamate N-methyl-D-aspartate (NMDA) receptor have emerged as possibly of central importance to the neurobiology of depression.4 The link between the strong therapeutic effects of antagonizing the NMDA receptor in depression and the increase in BDNF and neuroplasticity has emerged as a fresh model of depression.

From pills to intravenous infusions. Studies have shown that a single infusion of the NMDA receptor antagonist ketamine produces a very robust response, including full remission, in treatment–resistant unipolar or bipolar depression within 1 to 2 hours!5 The mechanism of action has been attributed to a surge of BDNF and immediate neuroplastic changes4 following NMDA receptor blockade. This abrupt reversal of severe depression, like turning on a switch, is a total and pleasant surprise.

From monotherapy to augmentation strategies. The rather imprudent notion that a single medication can be effective in a heterogeneous spectrum of disorders such as depression gradually is yielding to intelligent polypharmacy, using evidence-based augmentation strategies that might include lithium, thyroid hormone, another antidepressant (particularly mirtazapine), atypical antipsychotics, anti-inflammatory agents (including omega-3 fatty acids), antioxidants (especially N-acetylcysteine), L-methylfolate, and exercise.

From pharmacotherapy to neuromodulation. Because they disseminate to all organs and not just the brain, medications can cause undesirable side effects. Neuromodulation is used to treat depression by stimulating specific brain regions. Electroconvulsive therapy has a tarnished image but well established efficacy in severe depression. Repetitive transcranial magnetic stimulation and vagus nerve stimulation are FDA-approved for treating depression, while other forms of neuromodulation, such as cranial electrical stimulation, epidural cortical stimulation, focused ultrasound, low field magnetic stimulation, magnetic seizure therapy, near infrared light therapy, and transcranial direct current stimulation, still are in development.6 Deep brain stimulation (DBS) has been shown in several recent studies to reverse TRD,7,8 especially when stimulating the subgenual anterior cingulated region. In the future, DBS may become as commonly used in depression as it currently is in Parkinson’s disease.

A remarkable transformation is underway to reinvent the causes and treatments of depression that will reignite optimism about what psychiatry can do and eliminate the disability associated with major depression. Our patients can hardly wait.

Depression treatment seems to be in a “funk” these days. Armchair critics, some of whom have never treated a depressed patient, are flooding the media and Internet with allegations that antidepressants—which have helped patients for decades—are no better than placebo.

Several major pharmaceutical companies have pared down or shut down their CNS discovery, research, and development operations. There is a lack of new antidepressants with novel mechanisms of action, and the proportion of refractory or treatment-resistant depression (TRD) seems to be growing. In short, the status of clinical depression seems rather depressing.

Yet nothing can be further from the truth! A vibrant set of innovative, even radical, solutions for TRD are on the horizon. Here are some of the paradigm shifts in the neurobiology and therapeutics of major depression that gradually are toppling decades-old tenets and creeds related to this serious psychiatric disorder:

From genes vs environment to gene × environment interaction. For decades, it was assumed that some people have “endogenous” depression due to genetic determinants, while others are afflicted with “exogenous” depression caused by stressful life events. The new model shows that the environment interacts with genes to produce depression, and that having only risk genes or only environmental stress does not necessarily lead to depression.1

From ‘chemical imbalance’ to ‘inflammatory process.’ Evidence is accumulating that inflammation may be underpinning depression,2 and studies show levels of inflammatory cytokines and interleukins rise during a depressive episode and decline when the depression remits.

From ‘neurotransmitters’ to ‘neuroplasticity and neurotropic factors.’ Current treatments were developed to increase neurotransmitter activity in the brain, but new research reveals that depression is associated with a significant drop in neurotropic factors such as brain derived neurotropic factor (BDNF) or fibroblast growth factor with a concomitant decline in hippocampal neurogenesis.3

From serotonin, norepinephrine, and dopamine to glutamate. Over the past few years, glutamate pathways and the glutamate N-methyl-D-aspartate (NMDA) receptor have emerged as possibly of central importance to the neurobiology of depression.4 The link between the strong therapeutic effects of antagonizing the NMDA receptor in depression and the increase in BDNF and neuroplasticity has emerged as a fresh model of depression.

From pills to intravenous infusions. Studies have shown that a single infusion of the NMDA receptor antagonist ketamine produces a very robust response, including full remission, in treatment–resistant unipolar or bipolar depression within 1 to 2 hours!5 The mechanism of action has been attributed to a surge of BDNF and immediate neuroplastic changes4 following NMDA receptor blockade. This abrupt reversal of severe depression, like turning on a switch, is a total and pleasant surprise.

From monotherapy to augmentation strategies. The rather imprudent notion that a single medication can be effective in a heterogeneous spectrum of disorders such as depression gradually is yielding to intelligent polypharmacy, using evidence-based augmentation strategies that might include lithium, thyroid hormone, another antidepressant (particularly mirtazapine), atypical antipsychotics, anti-inflammatory agents (including omega-3 fatty acids), antioxidants (especially N-acetylcysteine), L-methylfolate, and exercise.

From pharmacotherapy to neuromodulation. Because they disseminate to all organs and not just the brain, medications can cause undesirable side effects. Neuromodulation is used to treat depression by stimulating specific brain regions. Electroconvulsive therapy has a tarnished image but well established efficacy in severe depression. Repetitive transcranial magnetic stimulation and vagus nerve stimulation are FDA-approved for treating depression, while other forms of neuromodulation, such as cranial electrical stimulation, epidural cortical stimulation, focused ultrasound, low field magnetic stimulation, magnetic seizure therapy, near infrared light therapy, and transcranial direct current stimulation, still are in development.6 Deep brain stimulation (DBS) has been shown in several recent studies to reverse TRD,7,8 especially when stimulating the subgenual anterior cingulated region. In the future, DBS may become as commonly used in depression as it currently is in Parkinson’s disease.

A remarkable transformation is underway to reinvent the causes and treatments of depression that will reignite optimism about what psychiatry can do and eliminate the disability associated with major depression. Our patients can hardly wait.

References

 

1. Raison CL, Lowry CA, Rook GA. Inflammation, sanitation, and consternation: loss of contact with coevolved, tolerogenic microorganisms and the pathophysiology and treatment of major depression. Arch Gen Psychiatry. 2010;67(12):1211-1224.

2. Caspi A, Sugden K, Moffitt TE, et al. Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science. 2003;301(5631):386-389.

3. Duman RS, Monteggia LM. A neurotrophic model for stress-related mood disorders. Biol Psychiatry. 2006;59(12):1116-1127.

4. Li N, Lee B, Liu RJ, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science. 2010;329(5994):959-964.

5. Zarate CA, Jr, Singh JB, Carlson PJ, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63(8):856-864.

6. Rosa MA, Lisanby SH. Somatic treatments for mood disorders. Neuropsychopharmacology. 2012;37(1):102-116.

7. Mayberg HS, Lozano AM, Voon V, et al. Deep brain stimulation for treatment-resistant depression. Neuron. 2005;45(5):651-660.

8. Holtzheimer PE, Kelley ME, Gross RE, et al. Subcallosal cingulate deep brain stimulation for treatment-resistant unipolar and bipolar depression. Arch Gen Psychiatry. 2012;69(2):150-158.

References

 

1. Raison CL, Lowry CA, Rook GA. Inflammation, sanitation, and consternation: loss of contact with coevolved, tolerogenic microorganisms and the pathophysiology and treatment of major depression. Arch Gen Psychiatry. 2010;67(12):1211-1224.

2. Caspi A, Sugden K, Moffitt TE, et al. Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science. 2003;301(5631):386-389.

3. Duman RS, Monteggia LM. A neurotrophic model for stress-related mood disorders. Biol Psychiatry. 2006;59(12):1116-1127.

4. Li N, Lee B, Liu RJ, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science. 2010;329(5994):959-964.

5. Zarate CA, Jr, Singh JB, Carlson PJ, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry. 2006;63(8):856-864.

6. Rosa MA, Lisanby SH. Somatic treatments for mood disorders. Neuropsychopharmacology. 2012;37(1):102-116.

7. Mayberg HS, Lozano AM, Voon V, et al. Deep brain stimulation for treatment-resistant depression. Neuron. 2005;45(5):651-660.

8. Holtzheimer PE, Kelley ME, Gross RE, et al. Subcallosal cingulate deep brain stimulation for treatment-resistant unipolar and bipolar depression. Arch Gen Psychiatry. 2012;69(2):150-158.

Issue
Current Psychiatry - 11(06)
Issue
Current Psychiatry - 11(06)
Page Number
4-5
Page Number
4-5
Publications
Publications
Topics
Article Type
Display Headline
Innovative approaches to treatment-resistant depression
Display Headline
Innovative approaches to treatment-resistant depression
Sections
Article PDF Media

Recognizing mimics of depression: The ‘8 Ds’

Article Type
Changed
Tue, 12/11/2018 - 15:04
Display Headline
Recognizing mimics of depression: The ‘8 Ds’

Discuss this article at www.facebook.com/CurrentPsychiatry

Dr. Rackley: How to spot conditions that may masquerade as depression

Many psychiatric and medical illnesses—as well as normal reactions to stressors—have symptoms that overlap with those of depressive disorders, including outwardly sad or dysphoric appearance, irritability, apathy or amotivation, fatigue, difficulty making decisions, social withdrawal, and sleep disturbances. This cluster of symptoms forms a readily observable behavioral phenotype that clinicians may label as depression before considering a broader differential diagnosis.

To better understand what other conditions belong in the differential diagnosis, we reviewed a sample of 100 consecutive medical/surgical inpatients referred to our consultation-liaison psychiatry practice for evaluation of “depression.” Ultimately, only 29 of these patients received a depression diagnosis. Many of the other diagnoses given in our sample required attention during inpatient medical or surgical care because they were potentially life-threatening if left unaddressed—such as delirium—or they interfered with managing the primary medical or surgical condition for which the patient was hospitalized.

Hurried or uncertain primary care clinicians frequently use “depression” as a catch-all term when requesting psychiatric consultation for patients who seem depressed. A wide range of conditions can mimic depression, and the art of psychosomatic psychiatry includes considering protean possibilities when assessing a patient. We identified 7 diagnoses that mimic major depression and developed our “8 D” differential to help clinicians properly diagnose “depressed” patients who have something other than a depressive disorder. Although our sample consisted of hospitalized patients, these mimics of depression may be found among patients referred from other clinical settings for evaluation of possible depression.

The perils of misdiagnosis

Depression is common among patients hospitalized with medical or surgical conditions. DSM-IV-TR diagnostic criteria for a major depressive episode (MDE) include the presence of low mood and/or anhedonia, plus ≥4 other depressive symptoms for ≥2 weeks.1 Growing evidence suggests that the relationship between depression and morbidity and mortality in medical illness is bidirectional, and nonpsychiatrists are becoming increasingly aware of major depression’s serious impact on their patients’ physical health.2-5

Although improving nonpsychiatrists’ recognition of depression in medically ill patients is laudable, it comes with a high false-positive rate. In a study of primary care outpatients, Berardi et al found that 45% of patients labeled “depressed” did not meet ICD-10 criteria for major depression, but >25% of those patients were prescribed an antidepressant.6 In a large retrospective study, Boland et al found that approximately 40% of patients referred to an inpatient psychiatric consultation service for depression did not meet criteria for a depressive illness, and primary medical services often confused organic syndromes such as delirium and dementia with depression.7 Similarly, Clarke et al found that 26% of medical and surgical inpatients referred to psychiatry with “depression” had another diagnosis—commonly delirium—that better accounted for their symptoms.8

What is the harm in overdiagnosing depression? Missing a serious or life-threatening diagnosis is a primary concern. For example, unrecognized delirium, which frequently was misdiagnosed as depression in the Berardi,6 Boland,7 and Clarke8 studies, is associated with myriad difficulties, including higher morbidity and mortality.9 Substance use disorders, which also commonly masquerade as depression, frequently are comorbid with medical illness. Delays in appropriate treatment of withdrawal syndromes—particularly of alcohol and sedative/hypnotic medications—are risk factors for increased mortality in these illnesses.10

Inappropriate, potentially harmful interventions are another concern. Many patients diagnosed with depression are prescribed antidepressants, but this is not always a benign intervention. Smith et al found that >10% of adult medical inpatients referred to a psychiatry consultation service who were started on an antidepressant had an adverse drug reaction severe enough to warrant discontinuing the medication.11 Antidepressant side effects relevant to medically ill patients include hyponatremia, serotonin syndrome, and exacerbation of delirium.12

Polypharmacy in medically ill patients increases the risk for serious drug-drug interactions. For example, serotonergic antidepressants can increase the risk for serotonin syndrome when combined with the analgesic tramadol, which has serotonergic activity,13 or the antibiotic linezolid, which is a reversible monoamine oxidase inhibitor.14 Many antidepressants—including paroxetine, fluoxetine, bupropion, sertraline, and duloxetine—are moderate to strong inhibitors of cytochrome P450 2D6 and therefore affect metabolism of many medications, including several beta blockers and antiarrhythmics, as well as the anti-estrogen tamoxifen. In the case of tamoxifen, which is a prodrug converted to active form by 2D6, concomitant use of a 2D6 inhibitor can substantially reduce the medication’s in vivo efficacy and lead to higher morbidity and mortality in breast cancer patients.15 As with any treatment, a decision to prescribe antidepressants needs to carefully be weighed in light of individual risks and benefits. This analysis starts by ensuring that an antidepressant is indicated.

 

 

Another concern is failing to recognize immediate human suffering for what it is. Hospitals and doctors’ offices are places of pain and loss as patients encounter morbidity and mortality in themselves and their loved ones. Rushing to pathologize the psychological or social manifestations of this pain can be invalidating to patients and may impair the doctor-patient relationship.

The 8 Ds

To determine what these “depression lookalike” syndromes could be, we identified 100 consecutive consultations to our adult inpatient psychiatry consultation-liaison team with a question of “depression.” We reviewed each patient’s chart, and recorded the diagnosis the psychiatrist gave to explain the patient’s depressed appearance. Data were recorded without patient identifiers, and the Mayo Clinic institutional review board (IRB) determined this study was exempt from IRB review.

Our sample included 45 men and 55 women with an average age of 48 (range: 18 to 91). On evaluation, 3 patients were given no psychiatric diagnosis, 29 were categorized as depressed, and 68 fell into one of 7 other “D” categories we describe below.

Depressed. These patients met criteria for a MDE in the context of major depressive disorder (MDD) or bipolar disorder, dysthymic disorder, mood disorder due to a general medical condition, substance-induced mood disorder, or depressive disorder not otherwise specified.

Demoralized. Patients who had difficulty adjusting to or coping with illness, and received a DSM-IV-TR diagnosis of adjustment disorder with the illness as the inciting stressor were placed in this category. Consistent with adjustment disorder criteria, these patients did not have depressive symptoms of sufficient intensity or duration to meet criteria for MDD or another primary mood disorder.

Difficult. For these patients, the primary issue was a breakdown in the therapeutic alliance with their treatment team. They received DSM-IV-TR diagnoses of personality disorder, noncompliance with treatment, or adult antisocial behavior.

Drugged. Patients in this category appeared depressed as a result of illicit substance use or misuse of alcohol or pharmaceuticals. DSM-IV-TR diagnoses included substance intoxication or withdrawal and substance abuse or dependence.

Delirious. This group consisted of patients with acute disruption in attention and level of consciousness that met DSM-IV-TR criteria for delirium. Patients whose delirious appearance was the result of illicit substance use or pharmaceutical misuse were categorized as “Drugged” rather than “Delirious.”

Disaffiliated. Patients in this category had dysphoria not commensurate with a full-blown mood disorder but attributable to grief from losing a major relationship to death, separation, or divorce. These patients received a DSM-IV-TR diagnosis of bereavement or a partner relational problem.

Delusional. These patients demonstrated amotivation and affective blunting as a result of a primary psychotic disorder such as schizophrenia. In preparation for emergent surgery, these patients had been prevented from taking anything orally, including antipsychotics, and their antipsychotics had not been restarted, which precipitated a gradual return of psychotic symptoms in the days after surgery.

Dulled. Two patients in our sample had irreversible cognitive deficits that explained their withdrawal and blunted affect; 1 had dementia and the other had mental retardation.

Managing the other Ds

In our sample, the most commonly misdiagnosed patients were those having difficulty adjusting to illness (Demoralized) or to other life events (Disaffiliated) (Table 1). In these cases, misdiagnosis has substantial treatment implications because these patients are better served by acute, illness-specific interventions that bolster coping skills, rather than pharmacotherapy or psychotherapy that targets entrenched depressive symptoms. For these patients, psychiatrists may “prescribe” interventions such as visits with a chaplain or other spiritual advisor, telephone calls or visits from family, friends, and other social supports, participation in physical or occupational therapy to improve adaptive functioning, or connecting with other patients in similar situations. Often, the key with these patients is to identify ways they have managed previous stressors and creatively use those resources to adapt to their new situation.

A second large group in our sample consisted of patients actively or passively fighting with their treatment team—the Difficult (Table 2). The treatment team or the patient’s caregivers and loved ones often are more distressed by the “difficult” patient’s symptoms than the patient, who may instead focus on his or her disappointment with caregivers who are unable to meet the patient’s unreasonable expectations. These challenges typically can be addressed by clarifying the salient issues for both the patient and team and establishing a liaison between patient and team to improve communication among all parties. Multidisciplinary care conferences can be an excellent way to ensure that the care team provides the patient with consistent communication and care.

 

 

A third group had potentially life-threatening conditions such as substance abuse/withdrawal or delirium as the cause of their “depressive” symptoms—the Drugged and the Delirious (Table 3). Recognizing an organic etiology of mood or behavioral symptoms is important because managing the underlying problem is the primary treatment strategy, not psychopharmacologic or psychotherapeutic intervention. Early identification and appropriate management of these patients could prevent further deterioration, improve medical outcomes, and shorten length of hospital stay.

A final group of patients was those whose chronic psychiatric and cognitive issues may go unrecognized or unappreciated until they interfere with the patient’s medical care—the Delusional and the Dulled (Table 2). In these cases, the correct diagnosis often hinges on obtaining a thorough history through collateral sources. The consulting psychiatrist can be crucial in co-managing these patients by establishing a liaison with outpatient providers, suggesting in-hospital management strategies such as alternate routes of administration of antipsychotics for patients with psychotic disorders, and connecting patients with outpatient supports after hospitalization. Continuity between inpatient and outpatient management is necessary to ensure a successful medical and psychiatric outcome.

Our 8 Ds are limited to the subset of patients referred by their medical teams with a question of depression. These referrals may have been motivated by a variety of patient, family, and team factors above and beyond the categories discussed in this article, and therefore may not accurately represent all patients who present with depressive symptoms in an inpatient setting. However, we hope that providing a mnemonic that suggests an extensive differential for a depressed phenotype may improve identification and management of these issues.

Table 1

Psychological crises that may look like depression

CategoryPercentage of our sampleDistinguishing featuresSuggested interventions
“Depressed” patients met DSM-IV-TR criteria for a depressive disorder29%Emotional symptoms: Depressed mood, anhedonia
Cognitive symptoms: concentration problems, indecisiveness, negative thoughts, irrational guilt
Physical symptoms: changes in sleep, appetite, energy
Initiate psychotherapy with or without antidepressants
“Demoralized” patients had difficulty coping with a medical illness23%Close temporal association with illness. Few neurovegetative symptoms. Able to maintain future orientation/hopeProvide compassion, recognition, and normalization. Connect patients with illness-specific supports (groups, social work, chaplaincy). Implement interventions to improve functioning (eg, PT/OT). Encourage patients to engage in activities that have helped them cope in the past
“Disaffiliated” patients had dysphoria attributable to grief from losing a major relationship3%Few neurovegetative symptoms. Able to maintain future orientation/hope. Improvement typical as time since loss increasesEncourage patients to connect with other supportive relationships. Refer patients to grief resources (eg, hospice, spiritual supports)
OT: occupational therapy; PT: physical therapy

Table 2

Differentiating patients with social challenges from those with depression

CategoryPercentage of our sampleDistinguishing featuresSuggested interventions
“Difficult” patients have a breakdown in the therapeutic alliance with their treatment team15%Mood changes often intense, immediate, and reactive to situation. Frequent breakdowns in communication with care team. Care team more distressed by patient’s symptoms than the patientEstablish frequent communication among care team members. Use multidisciplinary care conferences to clarify salient issues for patients and their team. Provide patients with consistent information and expectations
“Delusional” patients had affective blunting as a result of a psychotic disorder2%Suspicious about care team/procedures. Seems frightened or scans the room. On antipsychotics at admission. Slowly developing symptoms over several days after home medications are heldAcquire collateral history (an assigned community case manager or social worker can be an important source). Establish a plan for administering psychotropics in chronically mentally ill patients; consider IM or orally disintegrating formulations
“Dulled” patients had irreversible cognitive deficits2%Baseline impairments in memory and/or independent functioningAcquire collateral history. Perform a safety assessment of home environment with attention to need for additional supports
IM: intramuscular

Table 3

Substance abuse and delirium can mimic depression

CategoryPercentage of our sampleDistinguishing featuresSuggested interventions
“Drugged” patients appeared depressed as a result of substance use/ withdrawal12%Acute presentation closely mimicking mood, anxiety, or psychotic disorders. Emotional symptoms present when intoxicated or withdrawing and resolved during sobrietyImplement safety interventions to prevent self-harm or aggression during acute phase. Support and monitor withdrawal as indicated. Reassess mood state and symptoms once the patient is sober. Refer for chemical dependency evaluation
“Delirious” patients met DSM-IV-TR criteria for delirium11%Disoriented and inattentive. Onset over hours to days. Waxing and waning throughout the day. Possible hallucinations (often visual or tactile)Identify and correct underlying medical cause(s). Restore the patient’s sleep-wake cycle. Provide frequent reorientation and reassurance

Related Resources

  • Stern TA, Fricchione GL, Cassem NH, et al, eds. Massachusetts General Hospital handbook of general hospital psychiatry, 6th ed. Philadelphia, PA: Saunders Elsevier; 2010.
  • Levenson JL, ed. The American Psychiatric Publishing textbook of psychosomatic medicine. 2nd ed. Arlington, VA: American Psychiatric Publishing, Inc.; 2011.
  • Academy of Psychosomatic Medicine. www.apm.org.
  • Caplan JP, Stern TA. Mnemonics in a mnutshell: 32 aids to psychiatric diagnosis. Current Psychiatry. 2008;7(10):27-33.
 

 

Drug Brand Names

  • Bupropion • Wellbutrin, Zyban
  • Duloxetine • Cymbalta
  • Fluoxetine • Prozac
  • Linezolid • Zyvox
  • Paroxetine • Paxil
  • Sertraline • Zoloft
  • Tamoxifen • Nolvadex
  • Tramadol • Ultracet

Disclosures

Dr. Bostwick reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Rackley receives research/grant support from the Maternal and Child Health Bureau, Health Resources and Services Administration, U.S. Department of Health and Human Services, for a Collaborative Office Rounds program with primary care pediatricians.

References

1. Diagnostic and statistical manual of mental disorders 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.

2. Hansen MS, Fink P, Frydenberg M, et al. Use of health services, mental illness, and self-rated disability and health in medical inpatients. Psychosom Med. 2002;64(4):668-675.

3. Hosaka T, Aoki T, Watanabe T, et al. Comorbidity of depression among physically ill patients and its effect on the length of hospital stay. Psychiatry Clin Neurosci. 1999;53(4):491-495.

4. McCusker J, Cole M, Ciampi A, et al. Major depression in older medical inpatients predicts poor physical and mental health status over 12 months. Gen Hosp Psychiatry. 2007;29(4):340-348.

5. McCusker J, Cole M, Dufouil C, et al. The prevalence and correlates of major and minor depression in older medical inpatients. J Am Geriatr Soc. 2005;53(8):1344-1353.

6. Berardi D, Menchetti M, Cevenini N, et al. Increased recognition of depression in primary care. Comparison between primary-care physician and ICD-10 diagnosis of depression. Psychother Psychosom. 2005;74(4):225-230.

7. Boland RJ, Diaz S, Lamdan RM, et al. Overdiagnosis of depression in the general hospital. Gen Hosp Psychiatry. 1996;18(1):28-35.

8. Clarke DM, McKenzie DP, Smith GC. The recognition of depression in patients referred to a consultation-liaison service. J Psychosom Res. 1995;39(3):327-334.

9. Siddiqi N, House AO, Holmes JD. Occurrence and outcome of delirium in medical in-patients: a systematic literature review. Age Ageing. 2006;35(4):350-364.

10. Franklin JE, Levenson JL, McCance-Katz EF. Substance-related disorders. In: Levenson JL, ed. The American Psychiatric Publishing textbook of psychosomatic medicine. Washington, DC: American Psychiatric Publishing, Inc.; 2005:387–420.

11. Smith GC, Clarke DM, Handrinos D, et al. Consultation-liaison psychiatrists’ use of antidepressants in the physically ill. Psychosomatics. 2002;43(3):221-227.

12. Robinson MJ, Owen JA. Psychopharmacology. In: Levenson JL, ed. The American Psychiatric Publishing textbook of psychosomatic medicine. Washington, DC: American Psychiatric Publishing, Inc.; 2005:387–420.

13. Hersh EV, Pinto A, Moore PA. Adverse drug interactions involving common prescription and over-the-counter analgesic agents. Clin Ther. 2007;29(suppl):2477-2497.

14. Sola CL, Bostwick JM, Hart DA, et al. Anticipating potential linezolid-SSRI interactions in the general hospital setting: an MAOI in disguise. Mayo Clin Proc. 2006;81(3):330-334.

15. Stearns V, Johnson MD, Rae JM, et al. Active tamoxifen metabolite plasma concentrations after coadministration of tamoxifen and the selective serotonin reuptake inhibitor paroxetine. J Natl Cancer Inst. 2003;95(23):1758-1764.

Article PDF
Author and Disclosure Information

Michael J. Bostwick, MD
Professor of Psychiatry, Mayo Clinic College of Medicine, Rochester, MN
Sandra Rackley, MD
Director of Psychiatry Consultation-Liaison and Emergency Services, Children’s National Medical Center, Assistant Professor of Psychiatry and Pediatrics, The George Washington University Medical Center, Washington, DC

Issue
Current Psychiatry - 11(06)
Publications
Topics
Page Number
31-36
Sections
Author and Disclosure Information

Michael J. Bostwick, MD
Professor of Psychiatry, Mayo Clinic College of Medicine, Rochester, MN
Sandra Rackley, MD
Director of Psychiatry Consultation-Liaison and Emergency Services, Children’s National Medical Center, Assistant Professor of Psychiatry and Pediatrics, The George Washington University Medical Center, Washington, DC

Author and Disclosure Information

Michael J. Bostwick, MD
Professor of Psychiatry, Mayo Clinic College of Medicine, Rochester, MN
Sandra Rackley, MD
Director of Psychiatry Consultation-Liaison and Emergency Services, Children’s National Medical Center, Assistant Professor of Psychiatry and Pediatrics, The George Washington University Medical Center, Washington, DC

Article PDF
Article PDF

Discuss this article at www.facebook.com/CurrentPsychiatry

Dr. Rackley: How to spot conditions that may masquerade as depression

Many psychiatric and medical illnesses—as well as normal reactions to stressors—have symptoms that overlap with those of depressive disorders, including outwardly sad or dysphoric appearance, irritability, apathy or amotivation, fatigue, difficulty making decisions, social withdrawal, and sleep disturbances. This cluster of symptoms forms a readily observable behavioral phenotype that clinicians may label as depression before considering a broader differential diagnosis.

To better understand what other conditions belong in the differential diagnosis, we reviewed a sample of 100 consecutive medical/surgical inpatients referred to our consultation-liaison psychiatry practice for evaluation of “depression.” Ultimately, only 29 of these patients received a depression diagnosis. Many of the other diagnoses given in our sample required attention during inpatient medical or surgical care because they were potentially life-threatening if left unaddressed—such as delirium—or they interfered with managing the primary medical or surgical condition for which the patient was hospitalized.

Hurried or uncertain primary care clinicians frequently use “depression” as a catch-all term when requesting psychiatric consultation for patients who seem depressed. A wide range of conditions can mimic depression, and the art of psychosomatic psychiatry includes considering protean possibilities when assessing a patient. We identified 7 diagnoses that mimic major depression and developed our “8 D” differential to help clinicians properly diagnose “depressed” patients who have something other than a depressive disorder. Although our sample consisted of hospitalized patients, these mimics of depression may be found among patients referred from other clinical settings for evaluation of possible depression.

The perils of misdiagnosis

Depression is common among patients hospitalized with medical or surgical conditions. DSM-IV-TR diagnostic criteria for a major depressive episode (MDE) include the presence of low mood and/or anhedonia, plus ≥4 other depressive symptoms for ≥2 weeks.1 Growing evidence suggests that the relationship between depression and morbidity and mortality in medical illness is bidirectional, and nonpsychiatrists are becoming increasingly aware of major depression’s serious impact on their patients’ physical health.2-5

Although improving nonpsychiatrists’ recognition of depression in medically ill patients is laudable, it comes with a high false-positive rate. In a study of primary care outpatients, Berardi et al found that 45% of patients labeled “depressed” did not meet ICD-10 criteria for major depression, but >25% of those patients were prescribed an antidepressant.6 In a large retrospective study, Boland et al found that approximately 40% of patients referred to an inpatient psychiatric consultation service for depression did not meet criteria for a depressive illness, and primary medical services often confused organic syndromes such as delirium and dementia with depression.7 Similarly, Clarke et al found that 26% of medical and surgical inpatients referred to psychiatry with “depression” had another diagnosis—commonly delirium—that better accounted for their symptoms.8

What is the harm in overdiagnosing depression? Missing a serious or life-threatening diagnosis is a primary concern. For example, unrecognized delirium, which frequently was misdiagnosed as depression in the Berardi,6 Boland,7 and Clarke8 studies, is associated with myriad difficulties, including higher morbidity and mortality.9 Substance use disorders, which also commonly masquerade as depression, frequently are comorbid with medical illness. Delays in appropriate treatment of withdrawal syndromes—particularly of alcohol and sedative/hypnotic medications—are risk factors for increased mortality in these illnesses.10

Inappropriate, potentially harmful interventions are another concern. Many patients diagnosed with depression are prescribed antidepressants, but this is not always a benign intervention. Smith et al found that >10% of adult medical inpatients referred to a psychiatry consultation service who were started on an antidepressant had an adverse drug reaction severe enough to warrant discontinuing the medication.11 Antidepressant side effects relevant to medically ill patients include hyponatremia, serotonin syndrome, and exacerbation of delirium.12

Polypharmacy in medically ill patients increases the risk for serious drug-drug interactions. For example, serotonergic antidepressants can increase the risk for serotonin syndrome when combined with the analgesic tramadol, which has serotonergic activity,13 or the antibiotic linezolid, which is a reversible monoamine oxidase inhibitor.14 Many antidepressants—including paroxetine, fluoxetine, bupropion, sertraline, and duloxetine—are moderate to strong inhibitors of cytochrome P450 2D6 and therefore affect metabolism of many medications, including several beta blockers and antiarrhythmics, as well as the anti-estrogen tamoxifen. In the case of tamoxifen, which is a prodrug converted to active form by 2D6, concomitant use of a 2D6 inhibitor can substantially reduce the medication’s in vivo efficacy and lead to higher morbidity and mortality in breast cancer patients.15 As with any treatment, a decision to prescribe antidepressants needs to carefully be weighed in light of individual risks and benefits. This analysis starts by ensuring that an antidepressant is indicated.

 

 

Another concern is failing to recognize immediate human suffering for what it is. Hospitals and doctors’ offices are places of pain and loss as patients encounter morbidity and mortality in themselves and their loved ones. Rushing to pathologize the psychological or social manifestations of this pain can be invalidating to patients and may impair the doctor-patient relationship.

The 8 Ds

To determine what these “depression lookalike” syndromes could be, we identified 100 consecutive consultations to our adult inpatient psychiatry consultation-liaison team with a question of “depression.” We reviewed each patient’s chart, and recorded the diagnosis the psychiatrist gave to explain the patient’s depressed appearance. Data were recorded without patient identifiers, and the Mayo Clinic institutional review board (IRB) determined this study was exempt from IRB review.

Our sample included 45 men and 55 women with an average age of 48 (range: 18 to 91). On evaluation, 3 patients were given no psychiatric diagnosis, 29 were categorized as depressed, and 68 fell into one of 7 other “D” categories we describe below.

Depressed. These patients met criteria for a MDE in the context of major depressive disorder (MDD) or bipolar disorder, dysthymic disorder, mood disorder due to a general medical condition, substance-induced mood disorder, or depressive disorder not otherwise specified.

Demoralized. Patients who had difficulty adjusting to or coping with illness, and received a DSM-IV-TR diagnosis of adjustment disorder with the illness as the inciting stressor were placed in this category. Consistent with adjustment disorder criteria, these patients did not have depressive symptoms of sufficient intensity or duration to meet criteria for MDD or another primary mood disorder.

Difficult. For these patients, the primary issue was a breakdown in the therapeutic alliance with their treatment team. They received DSM-IV-TR diagnoses of personality disorder, noncompliance with treatment, or adult antisocial behavior.

Drugged. Patients in this category appeared depressed as a result of illicit substance use or misuse of alcohol or pharmaceuticals. DSM-IV-TR diagnoses included substance intoxication or withdrawal and substance abuse or dependence.

Delirious. This group consisted of patients with acute disruption in attention and level of consciousness that met DSM-IV-TR criteria for delirium. Patients whose delirious appearance was the result of illicit substance use or pharmaceutical misuse were categorized as “Drugged” rather than “Delirious.”

Disaffiliated. Patients in this category had dysphoria not commensurate with a full-blown mood disorder but attributable to grief from losing a major relationship to death, separation, or divorce. These patients received a DSM-IV-TR diagnosis of bereavement or a partner relational problem.

Delusional. These patients demonstrated amotivation and affective blunting as a result of a primary psychotic disorder such as schizophrenia. In preparation for emergent surgery, these patients had been prevented from taking anything orally, including antipsychotics, and their antipsychotics had not been restarted, which precipitated a gradual return of psychotic symptoms in the days after surgery.

Dulled. Two patients in our sample had irreversible cognitive deficits that explained their withdrawal and blunted affect; 1 had dementia and the other had mental retardation.

Managing the other Ds

In our sample, the most commonly misdiagnosed patients were those having difficulty adjusting to illness (Demoralized) or to other life events (Disaffiliated) (Table 1). In these cases, misdiagnosis has substantial treatment implications because these patients are better served by acute, illness-specific interventions that bolster coping skills, rather than pharmacotherapy or psychotherapy that targets entrenched depressive symptoms. For these patients, psychiatrists may “prescribe” interventions such as visits with a chaplain or other spiritual advisor, telephone calls or visits from family, friends, and other social supports, participation in physical or occupational therapy to improve adaptive functioning, or connecting with other patients in similar situations. Often, the key with these patients is to identify ways they have managed previous stressors and creatively use those resources to adapt to their new situation.

A second large group in our sample consisted of patients actively or passively fighting with their treatment team—the Difficult (Table 2). The treatment team or the patient’s caregivers and loved ones often are more distressed by the “difficult” patient’s symptoms than the patient, who may instead focus on his or her disappointment with caregivers who are unable to meet the patient’s unreasonable expectations. These challenges typically can be addressed by clarifying the salient issues for both the patient and team and establishing a liaison between patient and team to improve communication among all parties. Multidisciplinary care conferences can be an excellent way to ensure that the care team provides the patient with consistent communication and care.

 

 

A third group had potentially life-threatening conditions such as substance abuse/withdrawal or delirium as the cause of their “depressive” symptoms—the Drugged and the Delirious (Table 3). Recognizing an organic etiology of mood or behavioral symptoms is important because managing the underlying problem is the primary treatment strategy, not psychopharmacologic or psychotherapeutic intervention. Early identification and appropriate management of these patients could prevent further deterioration, improve medical outcomes, and shorten length of hospital stay.

A final group of patients was those whose chronic psychiatric and cognitive issues may go unrecognized or unappreciated until they interfere with the patient’s medical care—the Delusional and the Dulled (Table 2). In these cases, the correct diagnosis often hinges on obtaining a thorough history through collateral sources. The consulting psychiatrist can be crucial in co-managing these patients by establishing a liaison with outpatient providers, suggesting in-hospital management strategies such as alternate routes of administration of antipsychotics for patients with psychotic disorders, and connecting patients with outpatient supports after hospitalization. Continuity between inpatient and outpatient management is necessary to ensure a successful medical and psychiatric outcome.

Our 8 Ds are limited to the subset of patients referred by their medical teams with a question of depression. These referrals may have been motivated by a variety of patient, family, and team factors above and beyond the categories discussed in this article, and therefore may not accurately represent all patients who present with depressive symptoms in an inpatient setting. However, we hope that providing a mnemonic that suggests an extensive differential for a depressed phenotype may improve identification and management of these issues.

Table 1

Psychological crises that may look like depression

CategoryPercentage of our sampleDistinguishing featuresSuggested interventions
“Depressed” patients met DSM-IV-TR criteria for a depressive disorder29%Emotional symptoms: Depressed mood, anhedonia
Cognitive symptoms: concentration problems, indecisiveness, negative thoughts, irrational guilt
Physical symptoms: changes in sleep, appetite, energy
Initiate psychotherapy with or without antidepressants
“Demoralized” patients had difficulty coping with a medical illness23%Close temporal association with illness. Few neurovegetative symptoms. Able to maintain future orientation/hopeProvide compassion, recognition, and normalization. Connect patients with illness-specific supports (groups, social work, chaplaincy). Implement interventions to improve functioning (eg, PT/OT). Encourage patients to engage in activities that have helped them cope in the past
“Disaffiliated” patients had dysphoria attributable to grief from losing a major relationship3%Few neurovegetative symptoms. Able to maintain future orientation/hope. Improvement typical as time since loss increasesEncourage patients to connect with other supportive relationships. Refer patients to grief resources (eg, hospice, spiritual supports)
OT: occupational therapy; PT: physical therapy

Table 2

Differentiating patients with social challenges from those with depression

CategoryPercentage of our sampleDistinguishing featuresSuggested interventions
“Difficult” patients have a breakdown in the therapeutic alliance with their treatment team15%Mood changes often intense, immediate, and reactive to situation. Frequent breakdowns in communication with care team. Care team more distressed by patient’s symptoms than the patientEstablish frequent communication among care team members. Use multidisciplinary care conferences to clarify salient issues for patients and their team. Provide patients with consistent information and expectations
“Delusional” patients had affective blunting as a result of a psychotic disorder2%Suspicious about care team/procedures. Seems frightened or scans the room. On antipsychotics at admission. Slowly developing symptoms over several days after home medications are heldAcquire collateral history (an assigned community case manager or social worker can be an important source). Establish a plan for administering psychotropics in chronically mentally ill patients; consider IM or orally disintegrating formulations
“Dulled” patients had irreversible cognitive deficits2%Baseline impairments in memory and/or independent functioningAcquire collateral history. Perform a safety assessment of home environment with attention to need for additional supports
IM: intramuscular

Table 3

Substance abuse and delirium can mimic depression

CategoryPercentage of our sampleDistinguishing featuresSuggested interventions
“Drugged” patients appeared depressed as a result of substance use/ withdrawal12%Acute presentation closely mimicking mood, anxiety, or psychotic disorders. Emotional symptoms present when intoxicated or withdrawing and resolved during sobrietyImplement safety interventions to prevent self-harm or aggression during acute phase. Support and monitor withdrawal as indicated. Reassess mood state and symptoms once the patient is sober. Refer for chemical dependency evaluation
“Delirious” patients met DSM-IV-TR criteria for delirium11%Disoriented and inattentive. Onset over hours to days. Waxing and waning throughout the day. Possible hallucinations (often visual or tactile)Identify and correct underlying medical cause(s). Restore the patient’s sleep-wake cycle. Provide frequent reorientation and reassurance

Related Resources

  • Stern TA, Fricchione GL, Cassem NH, et al, eds. Massachusetts General Hospital handbook of general hospital psychiatry, 6th ed. Philadelphia, PA: Saunders Elsevier; 2010.
  • Levenson JL, ed. The American Psychiatric Publishing textbook of psychosomatic medicine. 2nd ed. Arlington, VA: American Psychiatric Publishing, Inc.; 2011.
  • Academy of Psychosomatic Medicine. www.apm.org.
  • Caplan JP, Stern TA. Mnemonics in a mnutshell: 32 aids to psychiatric diagnosis. Current Psychiatry. 2008;7(10):27-33.
 

 

Drug Brand Names

  • Bupropion • Wellbutrin, Zyban
  • Duloxetine • Cymbalta
  • Fluoxetine • Prozac
  • Linezolid • Zyvox
  • Paroxetine • Paxil
  • Sertraline • Zoloft
  • Tamoxifen • Nolvadex
  • Tramadol • Ultracet

Disclosures

Dr. Bostwick reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Rackley receives research/grant support from the Maternal and Child Health Bureau, Health Resources and Services Administration, U.S. Department of Health and Human Services, for a Collaborative Office Rounds program with primary care pediatricians.

Discuss this article at www.facebook.com/CurrentPsychiatry

Dr. Rackley: How to spot conditions that may masquerade as depression

Many psychiatric and medical illnesses—as well as normal reactions to stressors—have symptoms that overlap with those of depressive disorders, including outwardly sad or dysphoric appearance, irritability, apathy or amotivation, fatigue, difficulty making decisions, social withdrawal, and sleep disturbances. This cluster of symptoms forms a readily observable behavioral phenotype that clinicians may label as depression before considering a broader differential diagnosis.

To better understand what other conditions belong in the differential diagnosis, we reviewed a sample of 100 consecutive medical/surgical inpatients referred to our consultation-liaison psychiatry practice for evaluation of “depression.” Ultimately, only 29 of these patients received a depression diagnosis. Many of the other diagnoses given in our sample required attention during inpatient medical or surgical care because they were potentially life-threatening if left unaddressed—such as delirium—or they interfered with managing the primary medical or surgical condition for which the patient was hospitalized.

Hurried or uncertain primary care clinicians frequently use “depression” as a catch-all term when requesting psychiatric consultation for patients who seem depressed. A wide range of conditions can mimic depression, and the art of psychosomatic psychiatry includes considering protean possibilities when assessing a patient. We identified 7 diagnoses that mimic major depression and developed our “8 D” differential to help clinicians properly diagnose “depressed” patients who have something other than a depressive disorder. Although our sample consisted of hospitalized patients, these mimics of depression may be found among patients referred from other clinical settings for evaluation of possible depression.

The perils of misdiagnosis

Depression is common among patients hospitalized with medical or surgical conditions. DSM-IV-TR diagnostic criteria for a major depressive episode (MDE) include the presence of low mood and/or anhedonia, plus ≥4 other depressive symptoms for ≥2 weeks.1 Growing evidence suggests that the relationship between depression and morbidity and mortality in medical illness is bidirectional, and nonpsychiatrists are becoming increasingly aware of major depression’s serious impact on their patients’ physical health.2-5

Although improving nonpsychiatrists’ recognition of depression in medically ill patients is laudable, it comes with a high false-positive rate. In a study of primary care outpatients, Berardi et al found that 45% of patients labeled “depressed” did not meet ICD-10 criteria for major depression, but >25% of those patients were prescribed an antidepressant.6 In a large retrospective study, Boland et al found that approximately 40% of patients referred to an inpatient psychiatric consultation service for depression did not meet criteria for a depressive illness, and primary medical services often confused organic syndromes such as delirium and dementia with depression.7 Similarly, Clarke et al found that 26% of medical and surgical inpatients referred to psychiatry with “depression” had another diagnosis—commonly delirium—that better accounted for their symptoms.8

What is the harm in overdiagnosing depression? Missing a serious or life-threatening diagnosis is a primary concern. For example, unrecognized delirium, which frequently was misdiagnosed as depression in the Berardi,6 Boland,7 and Clarke8 studies, is associated with myriad difficulties, including higher morbidity and mortality.9 Substance use disorders, which also commonly masquerade as depression, frequently are comorbid with medical illness. Delays in appropriate treatment of withdrawal syndromes—particularly of alcohol and sedative/hypnotic medications—are risk factors for increased mortality in these illnesses.10

Inappropriate, potentially harmful interventions are another concern. Many patients diagnosed with depression are prescribed antidepressants, but this is not always a benign intervention. Smith et al found that >10% of adult medical inpatients referred to a psychiatry consultation service who were started on an antidepressant had an adverse drug reaction severe enough to warrant discontinuing the medication.11 Antidepressant side effects relevant to medically ill patients include hyponatremia, serotonin syndrome, and exacerbation of delirium.12

Polypharmacy in medically ill patients increases the risk for serious drug-drug interactions. For example, serotonergic antidepressants can increase the risk for serotonin syndrome when combined with the analgesic tramadol, which has serotonergic activity,13 or the antibiotic linezolid, which is a reversible monoamine oxidase inhibitor.14 Many antidepressants—including paroxetine, fluoxetine, bupropion, sertraline, and duloxetine—are moderate to strong inhibitors of cytochrome P450 2D6 and therefore affect metabolism of many medications, including several beta blockers and antiarrhythmics, as well as the anti-estrogen tamoxifen. In the case of tamoxifen, which is a prodrug converted to active form by 2D6, concomitant use of a 2D6 inhibitor can substantially reduce the medication’s in vivo efficacy and lead to higher morbidity and mortality in breast cancer patients.15 As with any treatment, a decision to prescribe antidepressants needs to carefully be weighed in light of individual risks and benefits. This analysis starts by ensuring that an antidepressant is indicated.

 

 

Another concern is failing to recognize immediate human suffering for what it is. Hospitals and doctors’ offices are places of pain and loss as patients encounter morbidity and mortality in themselves and their loved ones. Rushing to pathologize the psychological or social manifestations of this pain can be invalidating to patients and may impair the doctor-patient relationship.

The 8 Ds

To determine what these “depression lookalike” syndromes could be, we identified 100 consecutive consultations to our adult inpatient psychiatry consultation-liaison team with a question of “depression.” We reviewed each patient’s chart, and recorded the diagnosis the psychiatrist gave to explain the patient’s depressed appearance. Data were recorded without patient identifiers, and the Mayo Clinic institutional review board (IRB) determined this study was exempt from IRB review.

Our sample included 45 men and 55 women with an average age of 48 (range: 18 to 91). On evaluation, 3 patients were given no psychiatric diagnosis, 29 were categorized as depressed, and 68 fell into one of 7 other “D” categories we describe below.

Depressed. These patients met criteria for a MDE in the context of major depressive disorder (MDD) or bipolar disorder, dysthymic disorder, mood disorder due to a general medical condition, substance-induced mood disorder, or depressive disorder not otherwise specified.

Demoralized. Patients who had difficulty adjusting to or coping with illness, and received a DSM-IV-TR diagnosis of adjustment disorder with the illness as the inciting stressor were placed in this category. Consistent with adjustment disorder criteria, these patients did not have depressive symptoms of sufficient intensity or duration to meet criteria for MDD or another primary mood disorder.

Difficult. For these patients, the primary issue was a breakdown in the therapeutic alliance with their treatment team. They received DSM-IV-TR diagnoses of personality disorder, noncompliance with treatment, or adult antisocial behavior.

Drugged. Patients in this category appeared depressed as a result of illicit substance use or misuse of alcohol or pharmaceuticals. DSM-IV-TR diagnoses included substance intoxication or withdrawal and substance abuse or dependence.

Delirious. This group consisted of patients with acute disruption in attention and level of consciousness that met DSM-IV-TR criteria for delirium. Patients whose delirious appearance was the result of illicit substance use or pharmaceutical misuse were categorized as “Drugged” rather than “Delirious.”

Disaffiliated. Patients in this category had dysphoria not commensurate with a full-blown mood disorder but attributable to grief from losing a major relationship to death, separation, or divorce. These patients received a DSM-IV-TR diagnosis of bereavement or a partner relational problem.

Delusional. These patients demonstrated amotivation and affective blunting as a result of a primary psychotic disorder such as schizophrenia. In preparation for emergent surgery, these patients had been prevented from taking anything orally, including antipsychotics, and their antipsychotics had not been restarted, which precipitated a gradual return of psychotic symptoms in the days after surgery.

Dulled. Two patients in our sample had irreversible cognitive deficits that explained their withdrawal and blunted affect; 1 had dementia and the other had mental retardation.

Managing the other Ds

In our sample, the most commonly misdiagnosed patients were those having difficulty adjusting to illness (Demoralized) or to other life events (Disaffiliated) (Table 1). In these cases, misdiagnosis has substantial treatment implications because these patients are better served by acute, illness-specific interventions that bolster coping skills, rather than pharmacotherapy or psychotherapy that targets entrenched depressive symptoms. For these patients, psychiatrists may “prescribe” interventions such as visits with a chaplain or other spiritual advisor, telephone calls or visits from family, friends, and other social supports, participation in physical or occupational therapy to improve adaptive functioning, or connecting with other patients in similar situations. Often, the key with these patients is to identify ways they have managed previous stressors and creatively use those resources to adapt to their new situation.

A second large group in our sample consisted of patients actively or passively fighting with their treatment team—the Difficult (Table 2). The treatment team or the patient’s caregivers and loved ones often are more distressed by the “difficult” patient’s symptoms than the patient, who may instead focus on his or her disappointment with caregivers who are unable to meet the patient’s unreasonable expectations. These challenges typically can be addressed by clarifying the salient issues for both the patient and team and establishing a liaison between patient and team to improve communication among all parties. Multidisciplinary care conferences can be an excellent way to ensure that the care team provides the patient with consistent communication and care.

 

 

A third group had potentially life-threatening conditions such as substance abuse/withdrawal or delirium as the cause of their “depressive” symptoms—the Drugged and the Delirious (Table 3). Recognizing an organic etiology of mood or behavioral symptoms is important because managing the underlying problem is the primary treatment strategy, not psychopharmacologic or psychotherapeutic intervention. Early identification and appropriate management of these patients could prevent further deterioration, improve medical outcomes, and shorten length of hospital stay.

A final group of patients was those whose chronic psychiatric and cognitive issues may go unrecognized or unappreciated until they interfere with the patient’s medical care—the Delusional and the Dulled (Table 2). In these cases, the correct diagnosis often hinges on obtaining a thorough history through collateral sources. The consulting psychiatrist can be crucial in co-managing these patients by establishing a liaison with outpatient providers, suggesting in-hospital management strategies such as alternate routes of administration of antipsychotics for patients with psychotic disorders, and connecting patients with outpatient supports after hospitalization. Continuity between inpatient and outpatient management is necessary to ensure a successful medical and psychiatric outcome.

Our 8 Ds are limited to the subset of patients referred by their medical teams with a question of depression. These referrals may have been motivated by a variety of patient, family, and team factors above and beyond the categories discussed in this article, and therefore may not accurately represent all patients who present with depressive symptoms in an inpatient setting. However, we hope that providing a mnemonic that suggests an extensive differential for a depressed phenotype may improve identification and management of these issues.

Table 1

Psychological crises that may look like depression

CategoryPercentage of our sampleDistinguishing featuresSuggested interventions
“Depressed” patients met DSM-IV-TR criteria for a depressive disorder29%Emotional symptoms: Depressed mood, anhedonia
Cognitive symptoms: concentration problems, indecisiveness, negative thoughts, irrational guilt
Physical symptoms: changes in sleep, appetite, energy
Initiate psychotherapy with or without antidepressants
“Demoralized” patients had difficulty coping with a medical illness23%Close temporal association with illness. Few neurovegetative symptoms. Able to maintain future orientation/hopeProvide compassion, recognition, and normalization. Connect patients with illness-specific supports (groups, social work, chaplaincy). Implement interventions to improve functioning (eg, PT/OT). Encourage patients to engage in activities that have helped them cope in the past
“Disaffiliated” patients had dysphoria attributable to grief from losing a major relationship3%Few neurovegetative symptoms. Able to maintain future orientation/hope. Improvement typical as time since loss increasesEncourage patients to connect with other supportive relationships. Refer patients to grief resources (eg, hospice, spiritual supports)
OT: occupational therapy; PT: physical therapy

Table 2

Differentiating patients with social challenges from those with depression

CategoryPercentage of our sampleDistinguishing featuresSuggested interventions
“Difficult” patients have a breakdown in the therapeutic alliance with their treatment team15%Mood changes often intense, immediate, and reactive to situation. Frequent breakdowns in communication with care team. Care team more distressed by patient’s symptoms than the patientEstablish frequent communication among care team members. Use multidisciplinary care conferences to clarify salient issues for patients and their team. Provide patients with consistent information and expectations
“Delusional” patients had affective blunting as a result of a psychotic disorder2%Suspicious about care team/procedures. Seems frightened or scans the room. On antipsychotics at admission. Slowly developing symptoms over several days after home medications are heldAcquire collateral history (an assigned community case manager or social worker can be an important source). Establish a plan for administering psychotropics in chronically mentally ill patients; consider IM or orally disintegrating formulations
“Dulled” patients had irreversible cognitive deficits2%Baseline impairments in memory and/or independent functioningAcquire collateral history. Perform a safety assessment of home environment with attention to need for additional supports
IM: intramuscular

Table 3

Substance abuse and delirium can mimic depression

CategoryPercentage of our sampleDistinguishing featuresSuggested interventions
“Drugged” patients appeared depressed as a result of substance use/ withdrawal12%Acute presentation closely mimicking mood, anxiety, or psychotic disorders. Emotional symptoms present when intoxicated or withdrawing and resolved during sobrietyImplement safety interventions to prevent self-harm or aggression during acute phase. Support and monitor withdrawal as indicated. Reassess mood state and symptoms once the patient is sober. Refer for chemical dependency evaluation
“Delirious” patients met DSM-IV-TR criteria for delirium11%Disoriented and inattentive. Onset over hours to days. Waxing and waning throughout the day. Possible hallucinations (often visual or tactile)Identify and correct underlying medical cause(s). Restore the patient’s sleep-wake cycle. Provide frequent reorientation and reassurance

Related Resources

  • Stern TA, Fricchione GL, Cassem NH, et al, eds. Massachusetts General Hospital handbook of general hospital psychiatry, 6th ed. Philadelphia, PA: Saunders Elsevier; 2010.
  • Levenson JL, ed. The American Psychiatric Publishing textbook of psychosomatic medicine. 2nd ed. Arlington, VA: American Psychiatric Publishing, Inc.; 2011.
  • Academy of Psychosomatic Medicine. www.apm.org.
  • Caplan JP, Stern TA. Mnemonics in a mnutshell: 32 aids to psychiatric diagnosis. Current Psychiatry. 2008;7(10):27-33.
 

 

Drug Brand Names

  • Bupropion • Wellbutrin, Zyban
  • Duloxetine • Cymbalta
  • Fluoxetine • Prozac
  • Linezolid • Zyvox
  • Paroxetine • Paxil
  • Sertraline • Zoloft
  • Tamoxifen • Nolvadex
  • Tramadol • Ultracet

Disclosures

Dr. Bostwick reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Rackley receives research/grant support from the Maternal and Child Health Bureau, Health Resources and Services Administration, U.S. Department of Health and Human Services, for a Collaborative Office Rounds program with primary care pediatricians.

References

1. Diagnostic and statistical manual of mental disorders 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.

2. Hansen MS, Fink P, Frydenberg M, et al. Use of health services, mental illness, and self-rated disability and health in medical inpatients. Psychosom Med. 2002;64(4):668-675.

3. Hosaka T, Aoki T, Watanabe T, et al. Comorbidity of depression among physically ill patients and its effect on the length of hospital stay. Psychiatry Clin Neurosci. 1999;53(4):491-495.

4. McCusker J, Cole M, Ciampi A, et al. Major depression in older medical inpatients predicts poor physical and mental health status over 12 months. Gen Hosp Psychiatry. 2007;29(4):340-348.

5. McCusker J, Cole M, Dufouil C, et al. The prevalence and correlates of major and minor depression in older medical inpatients. J Am Geriatr Soc. 2005;53(8):1344-1353.

6. Berardi D, Menchetti M, Cevenini N, et al. Increased recognition of depression in primary care. Comparison between primary-care physician and ICD-10 diagnosis of depression. Psychother Psychosom. 2005;74(4):225-230.

7. Boland RJ, Diaz S, Lamdan RM, et al. Overdiagnosis of depression in the general hospital. Gen Hosp Psychiatry. 1996;18(1):28-35.

8. Clarke DM, McKenzie DP, Smith GC. The recognition of depression in patients referred to a consultation-liaison service. J Psychosom Res. 1995;39(3):327-334.

9. Siddiqi N, House AO, Holmes JD. Occurrence and outcome of delirium in medical in-patients: a systematic literature review. Age Ageing. 2006;35(4):350-364.

10. Franklin JE, Levenson JL, McCance-Katz EF. Substance-related disorders. In: Levenson JL, ed. The American Psychiatric Publishing textbook of psychosomatic medicine. Washington, DC: American Psychiatric Publishing, Inc.; 2005:387–420.

11. Smith GC, Clarke DM, Handrinos D, et al. Consultation-liaison psychiatrists’ use of antidepressants in the physically ill. Psychosomatics. 2002;43(3):221-227.

12. Robinson MJ, Owen JA. Psychopharmacology. In: Levenson JL, ed. The American Psychiatric Publishing textbook of psychosomatic medicine. Washington, DC: American Psychiatric Publishing, Inc.; 2005:387–420.

13. Hersh EV, Pinto A, Moore PA. Adverse drug interactions involving common prescription and over-the-counter analgesic agents. Clin Ther. 2007;29(suppl):2477-2497.

14. Sola CL, Bostwick JM, Hart DA, et al. Anticipating potential linezolid-SSRI interactions in the general hospital setting: an MAOI in disguise. Mayo Clin Proc. 2006;81(3):330-334.

15. Stearns V, Johnson MD, Rae JM, et al. Active tamoxifen metabolite plasma concentrations after coadministration of tamoxifen and the selective serotonin reuptake inhibitor paroxetine. J Natl Cancer Inst. 2003;95(23):1758-1764.

References

1. Diagnostic and statistical manual of mental disorders 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.

2. Hansen MS, Fink P, Frydenberg M, et al. Use of health services, mental illness, and self-rated disability and health in medical inpatients. Psychosom Med. 2002;64(4):668-675.

3. Hosaka T, Aoki T, Watanabe T, et al. Comorbidity of depression among physically ill patients and its effect on the length of hospital stay. Psychiatry Clin Neurosci. 1999;53(4):491-495.

4. McCusker J, Cole M, Ciampi A, et al. Major depression in older medical inpatients predicts poor physical and mental health status over 12 months. Gen Hosp Psychiatry. 2007;29(4):340-348.

5. McCusker J, Cole M, Dufouil C, et al. The prevalence and correlates of major and minor depression in older medical inpatients. J Am Geriatr Soc. 2005;53(8):1344-1353.

6. Berardi D, Menchetti M, Cevenini N, et al. Increased recognition of depression in primary care. Comparison between primary-care physician and ICD-10 diagnosis of depression. Psychother Psychosom. 2005;74(4):225-230.

7. Boland RJ, Diaz S, Lamdan RM, et al. Overdiagnosis of depression in the general hospital. Gen Hosp Psychiatry. 1996;18(1):28-35.

8. Clarke DM, McKenzie DP, Smith GC. The recognition of depression in patients referred to a consultation-liaison service. J Psychosom Res. 1995;39(3):327-334.

9. Siddiqi N, House AO, Holmes JD. Occurrence and outcome of delirium in medical in-patients: a systematic literature review. Age Ageing. 2006;35(4):350-364.

10. Franklin JE, Levenson JL, McCance-Katz EF. Substance-related disorders. In: Levenson JL, ed. The American Psychiatric Publishing textbook of psychosomatic medicine. Washington, DC: American Psychiatric Publishing, Inc.; 2005:387–420.

11. Smith GC, Clarke DM, Handrinos D, et al. Consultation-liaison psychiatrists’ use of antidepressants in the physically ill. Psychosomatics. 2002;43(3):221-227.

12. Robinson MJ, Owen JA. Psychopharmacology. In: Levenson JL, ed. The American Psychiatric Publishing textbook of psychosomatic medicine. Washington, DC: American Psychiatric Publishing, Inc.; 2005:387–420.

13. Hersh EV, Pinto A, Moore PA. Adverse drug interactions involving common prescription and over-the-counter analgesic agents. Clin Ther. 2007;29(suppl):2477-2497.

14. Sola CL, Bostwick JM, Hart DA, et al. Anticipating potential linezolid-SSRI interactions in the general hospital setting: an MAOI in disguise. Mayo Clin Proc. 2006;81(3):330-334.

15. Stearns V, Johnson MD, Rae JM, et al. Active tamoxifen metabolite plasma concentrations after coadministration of tamoxifen and the selective serotonin reuptake inhibitor paroxetine. J Natl Cancer Inst. 2003;95(23):1758-1764.

Issue
Current Psychiatry - 11(06)
Issue
Current Psychiatry - 11(06)
Page Number
31-36
Page Number
31-36
Publications
Publications
Topics
Article Type
Display Headline
Recognizing mimics of depression: The ‘8 Ds’
Display Headline
Recognizing mimics of depression: The ‘8 Ds’
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

More on ‘antipsychiatry’

Article Type
Changed
Thu, 03/28/2019 - 16:19
Display Headline
More on ‘antipsychiatry’

In reference to the “antipsychiatry” editorial by Dr. Henry A. Nasrallah (“The antipsychiatry movement: Who and why,” From the Editor, Current Psychiatry, December 2011, p. 4-6, 53): many years ago, when I was working at Chestnut Lodge, the family of a hospitalized patient asked Dr. Thomas Szasz to evaluate—as a consultant and “antipsychiatrist”—what should be done for this patient. Contrary to the family’s expectations, Dr. Szasz’s opinion was that the patient needed psychiatric treatment and hospitalization.

As a resident at Yale University, I knew Dr. Theodore Lidz very well. It is true that at a time of limited biological knowledge, he emphasized family dynamics as contributing to severe psychopathology, but he did not—to my knowledge—object to electroconvulsive therapy for a particular catatonic patient. His being demonized as an “antipsychiatrist” offends me because it misrepresents him.

John S. Kafka, MD
Private Practice
Washington, DC

Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

In reference to the “antipsychiatry” editorial by Dr. Henry A. Nasrallah (“The antipsychiatry movement: Who and why,” From the Editor, Current Psychiatry, December 2011, p. 4-6, 53): many years ago, when I was working at Chestnut Lodge, the family of a hospitalized patient asked Dr. Thomas Szasz to evaluate—as a consultant and “antipsychiatrist”—what should be done for this patient. Contrary to the family’s expectations, Dr. Szasz’s opinion was that the patient needed psychiatric treatment and hospitalization.

As a resident at Yale University, I knew Dr. Theodore Lidz very well. It is true that at a time of limited biological knowledge, he emphasized family dynamics as contributing to severe psychopathology, but he did not—to my knowledge—object to electroconvulsive therapy for a particular catatonic patient. His being demonized as an “antipsychiatrist” offends me because it misrepresents him.

John S. Kafka, MD
Private Practice
Washington, DC

In reference to the “antipsychiatry” editorial by Dr. Henry A. Nasrallah (“The antipsychiatry movement: Who and why,” From the Editor, Current Psychiatry, December 2011, p. 4-6, 53): many years ago, when I was working at Chestnut Lodge, the family of a hospitalized patient asked Dr. Thomas Szasz to evaluate—as a consultant and “antipsychiatrist”—what should be done for this patient. Contrary to the family’s expectations, Dr. Szasz’s opinion was that the patient needed psychiatric treatment and hospitalization.

As a resident at Yale University, I knew Dr. Theodore Lidz very well. It is true that at a time of limited biological knowledge, he emphasized family dynamics as contributing to severe psychopathology, but he did not—to my knowledge—object to electroconvulsive therapy for a particular catatonic patient. His being demonized as an “antipsychiatrist” offends me because it misrepresents him.

John S. Kafka, MD
Private Practice
Washington, DC

Publications
Publications
Topics
Article Type
Display Headline
More on ‘antipsychiatry’
Display Headline
More on ‘antipsychiatry’
Sections
Article Source

PURLs Copyright

Inside the Article

Patients unaware of TMS?

Article Type
Changed
Mon, 04/16/2018 - 14:31
Display Headline
Patients unaware of TMS?

Recently, when The Dr. Oz Show’s Dr. Mehmet Oz turned to his television audience to ask if anyone had heard of transcranial magnetic stimulation (TMS) after a demonstration of its effectiveness in treatment-resistant depression (TRD), no one in the audience raised their hands.

I wonder why Drs. Desseilles, Fava, Mischoulon, and Freeman did not discuss TMS as an important choice for TRD in their article (“Personalizing depression treatment: 2 clinical tools,” Current Psychiatry, March 2012, p. 26-33). Harvard Medical School has done trials of TMS and McLean Hospital has created a positive video on TMS.

Andrew Krompier, MD
Private Practice
San Ramon, CA

Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

Recently, when The Dr. Oz Show’s Dr. Mehmet Oz turned to his television audience to ask if anyone had heard of transcranial magnetic stimulation (TMS) after a demonstration of its effectiveness in treatment-resistant depression (TRD), no one in the audience raised their hands.

I wonder why Drs. Desseilles, Fava, Mischoulon, and Freeman did not discuss TMS as an important choice for TRD in their article (“Personalizing depression treatment: 2 clinical tools,” Current Psychiatry, March 2012, p. 26-33). Harvard Medical School has done trials of TMS and McLean Hospital has created a positive video on TMS.

Andrew Krompier, MD
Private Practice
San Ramon, CA

Recently, when The Dr. Oz Show’s Dr. Mehmet Oz turned to his television audience to ask if anyone had heard of transcranial magnetic stimulation (TMS) after a demonstration of its effectiveness in treatment-resistant depression (TRD), no one in the audience raised their hands.

I wonder why Drs. Desseilles, Fava, Mischoulon, and Freeman did not discuss TMS as an important choice for TRD in their article (“Personalizing depression treatment: 2 clinical tools,” Current Psychiatry, March 2012, p. 26-33). Harvard Medical School has done trials of TMS and McLean Hospital has created a positive video on TMS.

Andrew Krompier, MD
Private Practice
San Ramon, CA

Publications
Publications
Topics
Article Type
Display Headline
Patients unaware of TMS?
Display Headline
Patients unaware of TMS?
Sections
Article Source

PURLs Copyright

Inside the Article

Masters of American Psychiatry: Glen O. Gabbard, MD

Article Type
Changed
Thu, 03/28/2019 - 16:19
Display Headline
Masters of American Psychiatry: Glen O. Gabbard, MD

Video file

 
Files
Issue
Current Psychiatry - 11(05)
Publications
Topics
Sections
Files
Files

Video file

 

Video file

 
Issue
Current Psychiatry - 11(05)
Issue
Current Psychiatry - 11(05)
Publications
Publications
Topics
Article Type
Display Headline
Masters of American Psychiatry: Glen O. Gabbard, MD
Display Headline
Masters of American Psychiatry: Glen O. Gabbard, MD
Sections
Disallow All Ads

Binge eating disorder: Identify patterns of dysregulated eating and binging triggers

Article Type
Changed
Wed, 09/26/2018 - 13:50
Display Headline
Binge eating disorder: Identify patterns of dysregulated eating and binging triggers

Issue
Current Psychiatry - 11(05)
Publications
Topics
Sections

Issue
Current Psychiatry - 11(05)
Issue
Current Psychiatry - 11(05)
Publications
Publications
Topics
Article Type
Display Headline
Binge eating disorder: Identify patterns of dysregulated eating and binging triggers
Display Headline
Binge eating disorder: Identify patterns of dysregulated eating and binging triggers
Sections
Disallow All Ads
Alternative CME
Use ProPublica

Epileptic and depressed

Article Type
Changed
Tue, 12/11/2018 - 15:30
Display Headline
Epileptic and depressed

CASE: New-onset seizures

Ms. R, age 33, is referred by her neurologist for treatment of depressive symptoms that have intensified after she was diagnosed with epilepsy 1 year ago. She has a history of bulimia and ongoing anxiety and depression. She also has long-standing neuropathic pain in her left lateral shin and ankle that started after her foot was amputated in a lawn mower accident at age 5. Ms. R says she didn’t take pain medication until age 24, when her pain specialist prescribed tramadol, 300 to 400 mg/d, which she continues to take.

Ms. R’s first seizure occurred 1 year ago. Despite trials of several antiepileptics, her seizures persist; she is taking lamotrigine, 200 mg/d, when she presents for treatment. She has no history of brain injuries or strokes to explain her epilepsy. An MRI and 3 electroencephalograms show no signs of focal, potentially epileptogenic lesions.

Ms. R reports worsening depressive symptoms—particularly impaired attention and concentration—over several months that interfere with her housekeeping and ability to finish simple tasks at work. She says she drinks alcohol occasionally, but denies substance abuse. We initiate venlafaxine, titrated to 300 mg/d, because Ms. R has a history of intolerable side effects with fluoxetine (gastrointestinal distress) and citalopram (weight gain).

The authors’ observations

Tramadol, a centrally acting synthetic analgesic, consists of 2 enantiomers that act as weak agonists at μ-opioid receptors while also inhibiting serotonin and norepinephrine reuptake.1 Euphoria associated with μ receptor activation often is considered a “high.” Most abused opioids are prototypical μ agonists. When opioids are injected or inhaled, drug levels in the brain rise rapidly, causing a “rush”—a brief, intense, pleasurable sensation—followed by a longer-lasting high. Tolerance and physical dependence occur when opioids are used chronically.

Despite tramadol’s μ-opioid activity, the FDA approved it as an unscheduled analgesic in 1994 based on several human studies.2 Experience with tramadol has confirmed it has low abuse potential, yet human laboratory data—and some epidemiologic data—show that repeated use can lead to physical dependence. Although tramadol is considered a relatively weak opioid, human studies suggest that it possesses μ-agonist activity. The Drug Abuse Warning Network reported >15,000 emergency department (ED) visits for nonmedical tramadol use in 2009, which was more than the number of ED visits for codeine products (7,958) or propoxyphene products (9,526), but much fewer than visits for hydrocodone (86,258) or oxycodone (148,449) products.3

The recommended tramadol dose is 50 to 100 mg every 4 to 6 hours (maximum 400 mg/d). Adverse effects range from dysphoria, constipation, and nausea to agitation, seizures, respiratory depression, and coma.4 Tramadol withdrawal is similar to opioid withdrawal, and is characterized by anxiety, restlessness, insomnia, yawning, rhinorrhea, lacrimation, diaphoresis, tremor, muscle spasms, vomiting, diarrhea, and tachycardia. Rarely, psychomotor agitation and confusion may occur.5

Tramadol and seizures

At clinically appropriate doses, tramadol slightly suppresses seizure severity,6 but higher doses can induce seizures.7-12 This paradox is explained by tramadol’s effect on γ-aminobutyric acid (GABA) receptors. Although at clinical doses tramadol does not affect GABA, which could precipitate seizures, at higher doses it has been shown to have an inhibitory effect on GABA receptors.13,14 No prospective studies have assessed how often tramadol-induced seizures occur. Case reports12,15 suggest that seizures are more likely with acute tramadol intoxication, in patients with a history of alcohol abuse, or with pharmacologic regimens that include other medications that may cause seizures. Tramadol-induced seizures are generalized tonic-clonic in nature, and typically occur within 24 hours of the last dose.16

HISTORY: Worsening seizures

Two months after she presents for psychiatric evaluation, Ms. R experiences 6 generalized convulsions lasting from 15 minutes to 1 hour with no identifiable precipitant. Because oxcarbazepine and lamotrigine have failed to suppress her seizures, her neurologist adds phenytoin, 200 mg/d, and increases lamotrigine from 200 to 300 mg/d. Her depression continues to worsen. She reports severe insomnia, anhedonia, restlessness, and hopelessness, so we add sertraline, 50 mg/d, to venlafaxine. Ms. R says the seizures are terrifying and she cannot work. She moves in with her parents because she is unable to care for herself.

During a psychiatric appointment, Ms. R confesses that for 2 years her pain has been so unbearable that she has been buying extra tramadol from Internet retailers and taking 600 to 800 mg/d in addition to the prescribed 400 mg/d.

The authors’ observations

Ms. R had a history of chronic pain Table 117 and developed seizures after escalating her tramadol use. After her first epilepsy attack, she did not tell her physicians she was taking additional tramadol nor did she stop taking it. Treatment with several antiepileptics was unsuccessful. Her seizures persisted as long as her tramadol addiction continued.

 

 

Table 1

DSM-IV-TR criteria for pain disorder

  1. Pain in ≥1 anatomical site is the predominant focus of the clinical presentation and is of sufficient severity to warrant clinical attention
  2. The pain causes clinically significant distress or impairment in social, occupational, or other important areas of functioning
  3. Psychological factors are judged to have an important role in the onset, severity, exacerbation, or maintenance of the pain
  4. The symptom or deficit is not intentionally produced or feigned (as in factitious disorder or malingering)
  5. The pain is not better accounted for by a mood, anxiety, or psychotic disorder and does not meet criteria for dyspareunia
Source: Reference 17

Spiller et al18 reported the lowest daily tramadol dose associated with seizures is 500 mg/d, although Talaie et al16 observed seizures at doses as low as 100 mg/d. Additionally, seizure risk may increase through tramadol’s interactions with several medications, including tricyclic antidepressants, selective serotonin reuptake inhibitors, phenothiazines, fluoroquinolone antibiotics, meperidine, clozapine, buspirone, bupropion, phenylephrine, guaifenesin, tripelennamine, thioridazine, theophylline, and acetaminophen, butalbital, and caffeine combination (Table 2).19 Transdermal selegiline is contraindicated with tramadol. For Ms. R, the sertraline and venlafaxine she was taking may have augmented tramadol’s seizure potential.

Table 2

Tramadol: Major drug-drug interactions

DrugSymptoms
SelegilineNausea, vomiting, cardiovascular collapse, respiratory depression, seizures, or serotonin syndrome (hypertension, hyperthermia, myoclonus, mental status changes); use of the transdermal formulation with tramadol is contraindicated
CarbamazepineDecreased tramadol efficacy and increased seizure risk
VenlafaxineIncreased risk of serotonin syndrome
LinezolidIncreased risk of serotonin syndrome
FluoxetineIncreased risk of seizures and serotonin syndrome; increased concentrations of tramadol and decreased concentrations of tramadol active metabolite, O-desmethyltramadol (M1)
OlanzapineIncreased risk of serotonin syndrome
MirtazapineIncreased risk of serotonin syndrome
HaloperidolIncreased risk of seizures
EscitalopramIncreased risk of seizures and serotonin syndrome
ClomipramineIncreased risk of seizures
RisperidoneIncreased risk of seizures
KetamineIncreased risk of respiratory depression and excessive CNS depression
ImipramineIncreased risk of seizures
DuloxetineIncreased risk of serotonin syndrome
NortriptylineIncreased risk of seizures
ClozapineIncreased risk of seizures
SertralineIncreased risk of seizures and serotonin syndrome
ParoxetineIncreased risk of seizures and serotonin syndrome; decrease in the analgesic effect of tramadol
AmitriptylineIncreased risk of seizures; increased concentrations of tramadol and decreased concentrations of tramadol active metabolite, M1
DesipramineIncreased risk of seizures
DoxepinIncreased risk of seizures
CitalopramIncreased risk of seizures and serotonin syndrome
FluvoxamineIncreased risk of seizures and serotonin syndrome
Source: Reference 19

It is important to avoid polypharmacy in patients taking tramadol.20 Most psychiatrists are aware of the risk of serotonin syndrome with antidepressants, but may be less likely to attribute serotonergic additive effects from other medication classes such as analgesics. Recognizing tramadol’s potential to contribute to serotonin syndrome—especially in light of concomitant usage with other serotonergic medications such as antidepressants—is essential.

Tramadol toxicity appears to be caused by monoamine uptake inhibition rather than its opioid effects.21 The most frequent pharmacokinetic drug-drug interactions that lead to side effects such as serotonin syndrome or seizures involve several isoenzymes of the hepatic cytochrome P450 (CYP). The isoenzymes CYP2D6 (substrates—eg, amitriptyline, tramadol, and venlafaxine; inhibitors—eg, fluoxetine and duloxetine) and CYP3A4 (substrates—eg, carbamazepine, oxycodone, and venlafaxine; inductors—eg, carbamazepine; inhibitors, eg—grapefruit juice) are most important clinically.22

Ms. R readily obtained tramadol from Internet retailers. In a 2004 report, a Google search yielded 2,150,000 sources for acquiring tramadol, most of which did not require a prescription.23 Chronic pain patients have a higher prevalence of substance abuse than the general population.24 Because Ms. R did not have a documented substance abuse history, none of her physicians screened her for drug abuse, although toxicology screening wouldn’t have helped because the tramadol had been prescribed. We didn’t think to directly ask Ms. R about medication misuse, but if we had, she might have revealed it sooner.

OUTCOME: Seizure free

With Ms. R’s permission, we speak to her neurologist, who agrees that excess tramadol likely induced her seizures. The seizures stop after Ms. R discontinues tramadol. After 3 months without seizures, phenytoin is discontinued and lamotrigine is tapered to 200 mg/d. Ms. R participates in a pain rehabilitation program and continues to take venlafaxine, 300 mg/d, and sertraline, 50 mg/d. Her mood improves and she returns to work. Her pain is managed by non-steroidal anti-inflammatory drugs because she decides to decrease her activity level. Ms. R also is trying alternative medicine modalities such as acupuncture and acupressure.

Related Resource

  • Clark MR, Treisman GJ. Chronic pain and addiction. Basel, Switzerland: Karger; 2011.

Drug Brand Names

  • Acetaminophen, butalbital, and caffeine • Fioricet
  • Amitriptyline • Elavil
  • Bupropion • Wellbutrin
  • Buspirone • Buspar
  • Carbamazepine • Tegretol, Carbatrol
  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Clozapine • Clozaril
  • Desipramine • Norpramin
  • Doxepin • Adapin, Silenor
  • Duloxetine • Cymbalta
  • Escitalopram • Lexapro
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Guaifenesin • Tenex
  • Haloperidol • Haldol
  • Imipramine • Tofranil
  • Ketamine • Ketalar
  • Lamotrigine • Lamictal
  • Linezolid • Zyvox
  • Meperidine • Demerol
  • Mirtazapine • Remeron
  • Nortriptyline • Aventyl
  • Olanzapine • Zyprexa
  • Oxcarbazepine • Trileptal
  • Oxycodone • Percolone, OxyContin
  • Paroxetine • Paxil
  • Phenylephrine • Lusonal
  • Phenytoin • Dilantin
  • Propoxyphene • Darvon
  • Risperidone • Risperdal
  • Selegiline • Eldepryl, EMSAM
  • Sertraline • Zoloft
  • Theophylline • Aerolate
  • Thioridazine • Mellaril
  • Tramadol • Ultram
  • Tripelennamine • Pyribenzamine
  • Venlafaxine • Effexor
 

 

Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufactur-ers of competing products.

References

1. Katz KD. Tramadol is an opioid. J Med Toxicol. 2008;4(2):145-

2. Preston KL, Jasinski DR, Testa M. Abuse potential and pharmacological comparison of tramadol and morphine. Drug Alcohol Depend. 1991;27(1):7-17.

3. U.S. Department of Health and Human Services. Substance Abuse and Mental Health Services Administration. Drug abuse warning network, 2009: national estimates of drug-related emergency department visits. Rockville, MD: Substance Abuse and Mental Health Services Administration; 2011. HHS publication (SMA) 11-4659, DAWN Series D-35.

4. Afshari R, Ghooshkhanehee H. Tramadol overdose induced seizure dramatic rise of CPK and acute renal failure. J Pak Med Assoc. 2009;59(3):178.-

5. Rodriguez Villamañan JC, Albaladejo Blanco C, Sanchez Sanchez A, et al. Withdrawal syndrome after long-term treatment with tramadol. Br J Gen Pract. 2000;50(454):406.-

6. Manocha A, Sharma KK, Mediratta PK. On the mechanism of anticonvulsant effect of tramadol in mice. Pharmacol Biochem Behav. 2005;82(1):74-81.

7. Boyd IW. Tramadol and seizures. Med J Aust. 2005;182(11):595-596.

8. Labate A, Newton MR, Vernon GM, et al. Tramadol and new-onset seizures. Med J Aust. 2005;182(1):42-43.

9. Gasse C, Derby L, Vasilakis-Scaramozza C, et al. Incidence of first-time idiopathic seizures in users of tramadol. Pharmacotherapy. 2000;20(6):629-634.

10. Kahn LH, Alderfer RJ, Graham DJ. Seizures reported with tramadol. JAMA. 1997;278(20):1661.-

11. Mazor SS, Feldman KW, Sugar NF, et al. Pediatric tramadol ingestion resulting in seizurelike activity: a case series. Pediatr Emerg Care. 2008;24(6):380-381.

12. Raffa RB, Stone DJ, Jr. Unexceptional seizure potential of tramadol or its enantiomers or metabolites in mice. J Pharmacol Exp Ther. 2008;325(2):500-506.

13. Rehni AK, Singh TG, Singh N, et al. Tramadol-induced seizurogenic effect: a possible role of opioid-dependent histamine H1 receptor activation-linked mechanism. Naunyn Schmiedebergs Arch Pharmacol. 2010;381(1):11-19.

14. Rehni AK, Singh I, Kumar M. Tramadol-induced seizurogenic effect: a possible role of opioid-dependent gamma-aminobutyric acid inhibitory pathway. Basic Clin Pharmacol Toxicol. 2008;103(3):262-266.

15. Jovanović-Cupić V, Martinović Z, Nesić N. Seizures associated with intoxication and abuse of tramadol. Clin Toxicol (Phila). 2006;44(2):143-146.

16. Talaie H, Panahandeh R, Fayaznouri M, et al. Dose-independent occurrence of seizure with tramadol. J Med Toxicol. 2009;5(2):63-67.

17. Diagnostic and statistical manual of mental disorders 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.

18. Spiller HA, Gorman SE, Villalobos D, et al. Prospective multicenter evaluation of tramadol exposure. J Toxicol Clin Toxicol. 1997;35(4):361-364.

19. Reus VI, Rawitscher L. Possible interaction of tramadol and antidepressants. Am J Psychiatry. 2000;157(5):839.-

20. Thundiyil JG, Kearney TE, Olson KR. Evolving epidemiology of drug-induced seizures reported to a Poison Control Center System. J Med Toxicol. 2007;3(1):15-19.

21. Looper KJ. Potential medical and surgical complications of serotonergic antidepressant medications. Psychosomatics. 2007;48(1):1-9.

22. Grond S, Sablotzki A. Clinical pharmacology of tramadol. Clin Pharmacokinet. 2004;43(13):879-923.

23. Lineberry TW, Bostwick JM. Taking the physician out of “physician shopping”: a case series of clinical problems associated with Internet purchases of medication. Mayo Clin Proc. 2004;79(8):1031-1034.

24. Savage SR. Assessment for addiction in pain-treatment settings. Clin J Pain. 2002;18(4 suppl):S28-S38.

Article PDF
Author and Disclosure Information

Magdalena Romanowicz, MD
Dr. Romanowicz is a Child and Adolescent Psychiatry Fellow, Stanford University, Stanford, CA
John Michael Bostwick, MD
Dr. Bostwick is Professor of Psychiatry, Mayo Clinic, Rochester, MN

Issue
Current Psychiatry - 11(05)
Publications
Topics
Page Number
45-49
Legacy Keywords
epilepsy; depression; bulimia; anxiety; chronic pain; seizures; tramadol; substance abuse
Sections
Author and Disclosure Information

Magdalena Romanowicz, MD
Dr. Romanowicz is a Child and Adolescent Psychiatry Fellow, Stanford University, Stanford, CA
John Michael Bostwick, MD
Dr. Bostwick is Professor of Psychiatry, Mayo Clinic, Rochester, MN

Author and Disclosure Information

Magdalena Romanowicz, MD
Dr. Romanowicz is a Child and Adolescent Psychiatry Fellow, Stanford University, Stanford, CA
John Michael Bostwick, MD
Dr. Bostwick is Professor of Psychiatry, Mayo Clinic, Rochester, MN

Article PDF
Article PDF

CASE: New-onset seizures

Ms. R, age 33, is referred by her neurologist for treatment of depressive symptoms that have intensified after she was diagnosed with epilepsy 1 year ago. She has a history of bulimia and ongoing anxiety and depression. She also has long-standing neuropathic pain in her left lateral shin and ankle that started after her foot was amputated in a lawn mower accident at age 5. Ms. R says she didn’t take pain medication until age 24, when her pain specialist prescribed tramadol, 300 to 400 mg/d, which she continues to take.

Ms. R’s first seizure occurred 1 year ago. Despite trials of several antiepileptics, her seizures persist; she is taking lamotrigine, 200 mg/d, when she presents for treatment. She has no history of brain injuries or strokes to explain her epilepsy. An MRI and 3 electroencephalograms show no signs of focal, potentially epileptogenic lesions.

Ms. R reports worsening depressive symptoms—particularly impaired attention and concentration—over several months that interfere with her housekeeping and ability to finish simple tasks at work. She says she drinks alcohol occasionally, but denies substance abuse. We initiate venlafaxine, titrated to 300 mg/d, because Ms. R has a history of intolerable side effects with fluoxetine (gastrointestinal distress) and citalopram (weight gain).

The authors’ observations

Tramadol, a centrally acting synthetic analgesic, consists of 2 enantiomers that act as weak agonists at μ-opioid receptors while also inhibiting serotonin and norepinephrine reuptake.1 Euphoria associated with μ receptor activation often is considered a “high.” Most abused opioids are prototypical μ agonists. When opioids are injected or inhaled, drug levels in the brain rise rapidly, causing a “rush”—a brief, intense, pleasurable sensation—followed by a longer-lasting high. Tolerance and physical dependence occur when opioids are used chronically.

Despite tramadol’s μ-opioid activity, the FDA approved it as an unscheduled analgesic in 1994 based on several human studies.2 Experience with tramadol has confirmed it has low abuse potential, yet human laboratory data—and some epidemiologic data—show that repeated use can lead to physical dependence. Although tramadol is considered a relatively weak opioid, human studies suggest that it possesses μ-agonist activity. The Drug Abuse Warning Network reported >15,000 emergency department (ED) visits for nonmedical tramadol use in 2009, which was more than the number of ED visits for codeine products (7,958) or propoxyphene products (9,526), but much fewer than visits for hydrocodone (86,258) or oxycodone (148,449) products.3

The recommended tramadol dose is 50 to 100 mg every 4 to 6 hours (maximum 400 mg/d). Adverse effects range from dysphoria, constipation, and nausea to agitation, seizures, respiratory depression, and coma.4 Tramadol withdrawal is similar to opioid withdrawal, and is characterized by anxiety, restlessness, insomnia, yawning, rhinorrhea, lacrimation, diaphoresis, tremor, muscle spasms, vomiting, diarrhea, and tachycardia. Rarely, psychomotor agitation and confusion may occur.5

Tramadol and seizures

At clinically appropriate doses, tramadol slightly suppresses seizure severity,6 but higher doses can induce seizures.7-12 This paradox is explained by tramadol’s effect on γ-aminobutyric acid (GABA) receptors. Although at clinical doses tramadol does not affect GABA, which could precipitate seizures, at higher doses it has been shown to have an inhibitory effect on GABA receptors.13,14 No prospective studies have assessed how often tramadol-induced seizures occur. Case reports12,15 suggest that seizures are more likely with acute tramadol intoxication, in patients with a history of alcohol abuse, or with pharmacologic regimens that include other medications that may cause seizures. Tramadol-induced seizures are generalized tonic-clonic in nature, and typically occur within 24 hours of the last dose.16

HISTORY: Worsening seizures

Two months after she presents for psychiatric evaluation, Ms. R experiences 6 generalized convulsions lasting from 15 minutes to 1 hour with no identifiable precipitant. Because oxcarbazepine and lamotrigine have failed to suppress her seizures, her neurologist adds phenytoin, 200 mg/d, and increases lamotrigine from 200 to 300 mg/d. Her depression continues to worsen. She reports severe insomnia, anhedonia, restlessness, and hopelessness, so we add sertraline, 50 mg/d, to venlafaxine. Ms. R says the seizures are terrifying and she cannot work. She moves in with her parents because she is unable to care for herself.

During a psychiatric appointment, Ms. R confesses that for 2 years her pain has been so unbearable that she has been buying extra tramadol from Internet retailers and taking 600 to 800 mg/d in addition to the prescribed 400 mg/d.

The authors’ observations

Ms. R had a history of chronic pain Table 117 and developed seizures after escalating her tramadol use. After her first epilepsy attack, she did not tell her physicians she was taking additional tramadol nor did she stop taking it. Treatment with several antiepileptics was unsuccessful. Her seizures persisted as long as her tramadol addiction continued.

 

 

Table 1

DSM-IV-TR criteria for pain disorder

  1. Pain in ≥1 anatomical site is the predominant focus of the clinical presentation and is of sufficient severity to warrant clinical attention
  2. The pain causes clinically significant distress or impairment in social, occupational, or other important areas of functioning
  3. Psychological factors are judged to have an important role in the onset, severity, exacerbation, or maintenance of the pain
  4. The symptom or deficit is not intentionally produced or feigned (as in factitious disorder or malingering)
  5. The pain is not better accounted for by a mood, anxiety, or psychotic disorder and does not meet criteria for dyspareunia
Source: Reference 17

Spiller et al18 reported the lowest daily tramadol dose associated with seizures is 500 mg/d, although Talaie et al16 observed seizures at doses as low as 100 mg/d. Additionally, seizure risk may increase through tramadol’s interactions with several medications, including tricyclic antidepressants, selective serotonin reuptake inhibitors, phenothiazines, fluoroquinolone antibiotics, meperidine, clozapine, buspirone, bupropion, phenylephrine, guaifenesin, tripelennamine, thioridazine, theophylline, and acetaminophen, butalbital, and caffeine combination (Table 2).19 Transdermal selegiline is contraindicated with tramadol. For Ms. R, the sertraline and venlafaxine she was taking may have augmented tramadol’s seizure potential.

Table 2

Tramadol: Major drug-drug interactions

DrugSymptoms
SelegilineNausea, vomiting, cardiovascular collapse, respiratory depression, seizures, or serotonin syndrome (hypertension, hyperthermia, myoclonus, mental status changes); use of the transdermal formulation with tramadol is contraindicated
CarbamazepineDecreased tramadol efficacy and increased seizure risk
VenlafaxineIncreased risk of serotonin syndrome
LinezolidIncreased risk of serotonin syndrome
FluoxetineIncreased risk of seizures and serotonin syndrome; increased concentrations of tramadol and decreased concentrations of tramadol active metabolite, O-desmethyltramadol (M1)
OlanzapineIncreased risk of serotonin syndrome
MirtazapineIncreased risk of serotonin syndrome
HaloperidolIncreased risk of seizures
EscitalopramIncreased risk of seizures and serotonin syndrome
ClomipramineIncreased risk of seizures
RisperidoneIncreased risk of seizures
KetamineIncreased risk of respiratory depression and excessive CNS depression
ImipramineIncreased risk of seizures
DuloxetineIncreased risk of serotonin syndrome
NortriptylineIncreased risk of seizures
ClozapineIncreased risk of seizures
SertralineIncreased risk of seizures and serotonin syndrome
ParoxetineIncreased risk of seizures and serotonin syndrome; decrease in the analgesic effect of tramadol
AmitriptylineIncreased risk of seizures; increased concentrations of tramadol and decreased concentrations of tramadol active metabolite, M1
DesipramineIncreased risk of seizures
DoxepinIncreased risk of seizures
CitalopramIncreased risk of seizures and serotonin syndrome
FluvoxamineIncreased risk of seizures and serotonin syndrome
Source: Reference 19

It is important to avoid polypharmacy in patients taking tramadol.20 Most psychiatrists are aware of the risk of serotonin syndrome with antidepressants, but may be less likely to attribute serotonergic additive effects from other medication classes such as analgesics. Recognizing tramadol’s potential to contribute to serotonin syndrome—especially in light of concomitant usage with other serotonergic medications such as antidepressants—is essential.

Tramadol toxicity appears to be caused by monoamine uptake inhibition rather than its opioid effects.21 The most frequent pharmacokinetic drug-drug interactions that lead to side effects such as serotonin syndrome or seizures involve several isoenzymes of the hepatic cytochrome P450 (CYP). The isoenzymes CYP2D6 (substrates—eg, amitriptyline, tramadol, and venlafaxine; inhibitors—eg, fluoxetine and duloxetine) and CYP3A4 (substrates—eg, carbamazepine, oxycodone, and venlafaxine; inductors—eg, carbamazepine; inhibitors, eg—grapefruit juice) are most important clinically.22

Ms. R readily obtained tramadol from Internet retailers. In a 2004 report, a Google search yielded 2,150,000 sources for acquiring tramadol, most of which did not require a prescription.23 Chronic pain patients have a higher prevalence of substance abuse than the general population.24 Because Ms. R did not have a documented substance abuse history, none of her physicians screened her for drug abuse, although toxicology screening wouldn’t have helped because the tramadol had been prescribed. We didn’t think to directly ask Ms. R about medication misuse, but if we had, she might have revealed it sooner.

OUTCOME: Seizure free

With Ms. R’s permission, we speak to her neurologist, who agrees that excess tramadol likely induced her seizures. The seizures stop after Ms. R discontinues tramadol. After 3 months without seizures, phenytoin is discontinued and lamotrigine is tapered to 200 mg/d. Ms. R participates in a pain rehabilitation program and continues to take venlafaxine, 300 mg/d, and sertraline, 50 mg/d. Her mood improves and she returns to work. Her pain is managed by non-steroidal anti-inflammatory drugs because she decides to decrease her activity level. Ms. R also is trying alternative medicine modalities such as acupuncture and acupressure.

Related Resource

  • Clark MR, Treisman GJ. Chronic pain and addiction. Basel, Switzerland: Karger; 2011.

Drug Brand Names

  • Acetaminophen, butalbital, and caffeine • Fioricet
  • Amitriptyline • Elavil
  • Bupropion • Wellbutrin
  • Buspirone • Buspar
  • Carbamazepine • Tegretol, Carbatrol
  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Clozapine • Clozaril
  • Desipramine • Norpramin
  • Doxepin • Adapin, Silenor
  • Duloxetine • Cymbalta
  • Escitalopram • Lexapro
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Guaifenesin • Tenex
  • Haloperidol • Haldol
  • Imipramine • Tofranil
  • Ketamine • Ketalar
  • Lamotrigine • Lamictal
  • Linezolid • Zyvox
  • Meperidine • Demerol
  • Mirtazapine • Remeron
  • Nortriptyline • Aventyl
  • Olanzapine • Zyprexa
  • Oxcarbazepine • Trileptal
  • Oxycodone • Percolone, OxyContin
  • Paroxetine • Paxil
  • Phenylephrine • Lusonal
  • Phenytoin • Dilantin
  • Propoxyphene • Darvon
  • Risperidone • Risperdal
  • Selegiline • Eldepryl, EMSAM
  • Sertraline • Zoloft
  • Theophylline • Aerolate
  • Thioridazine • Mellaril
  • Tramadol • Ultram
  • Tripelennamine • Pyribenzamine
  • Venlafaxine • Effexor
 

 

Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufactur-ers of competing products.

CASE: New-onset seizures

Ms. R, age 33, is referred by her neurologist for treatment of depressive symptoms that have intensified after she was diagnosed with epilepsy 1 year ago. She has a history of bulimia and ongoing anxiety and depression. She also has long-standing neuropathic pain in her left lateral shin and ankle that started after her foot was amputated in a lawn mower accident at age 5. Ms. R says she didn’t take pain medication until age 24, when her pain specialist prescribed tramadol, 300 to 400 mg/d, which she continues to take.

Ms. R’s first seizure occurred 1 year ago. Despite trials of several antiepileptics, her seizures persist; she is taking lamotrigine, 200 mg/d, when she presents for treatment. She has no history of brain injuries or strokes to explain her epilepsy. An MRI and 3 electroencephalograms show no signs of focal, potentially epileptogenic lesions.

Ms. R reports worsening depressive symptoms—particularly impaired attention and concentration—over several months that interfere with her housekeeping and ability to finish simple tasks at work. She says she drinks alcohol occasionally, but denies substance abuse. We initiate venlafaxine, titrated to 300 mg/d, because Ms. R has a history of intolerable side effects with fluoxetine (gastrointestinal distress) and citalopram (weight gain).

The authors’ observations

Tramadol, a centrally acting synthetic analgesic, consists of 2 enantiomers that act as weak agonists at μ-opioid receptors while also inhibiting serotonin and norepinephrine reuptake.1 Euphoria associated with μ receptor activation often is considered a “high.” Most abused opioids are prototypical μ agonists. When opioids are injected or inhaled, drug levels in the brain rise rapidly, causing a “rush”—a brief, intense, pleasurable sensation—followed by a longer-lasting high. Tolerance and physical dependence occur when opioids are used chronically.

Despite tramadol’s μ-opioid activity, the FDA approved it as an unscheduled analgesic in 1994 based on several human studies.2 Experience with tramadol has confirmed it has low abuse potential, yet human laboratory data—and some epidemiologic data—show that repeated use can lead to physical dependence. Although tramadol is considered a relatively weak opioid, human studies suggest that it possesses μ-agonist activity. The Drug Abuse Warning Network reported >15,000 emergency department (ED) visits for nonmedical tramadol use in 2009, which was more than the number of ED visits for codeine products (7,958) or propoxyphene products (9,526), but much fewer than visits for hydrocodone (86,258) or oxycodone (148,449) products.3

The recommended tramadol dose is 50 to 100 mg every 4 to 6 hours (maximum 400 mg/d). Adverse effects range from dysphoria, constipation, and nausea to agitation, seizures, respiratory depression, and coma.4 Tramadol withdrawal is similar to opioid withdrawal, and is characterized by anxiety, restlessness, insomnia, yawning, rhinorrhea, lacrimation, diaphoresis, tremor, muscle spasms, vomiting, diarrhea, and tachycardia. Rarely, psychomotor agitation and confusion may occur.5

Tramadol and seizures

At clinically appropriate doses, tramadol slightly suppresses seizure severity,6 but higher doses can induce seizures.7-12 This paradox is explained by tramadol’s effect on γ-aminobutyric acid (GABA) receptors. Although at clinical doses tramadol does not affect GABA, which could precipitate seizures, at higher doses it has been shown to have an inhibitory effect on GABA receptors.13,14 No prospective studies have assessed how often tramadol-induced seizures occur. Case reports12,15 suggest that seizures are more likely with acute tramadol intoxication, in patients with a history of alcohol abuse, or with pharmacologic regimens that include other medications that may cause seizures. Tramadol-induced seizures are generalized tonic-clonic in nature, and typically occur within 24 hours of the last dose.16

HISTORY: Worsening seizures

Two months after she presents for psychiatric evaluation, Ms. R experiences 6 generalized convulsions lasting from 15 minutes to 1 hour with no identifiable precipitant. Because oxcarbazepine and lamotrigine have failed to suppress her seizures, her neurologist adds phenytoin, 200 mg/d, and increases lamotrigine from 200 to 300 mg/d. Her depression continues to worsen. She reports severe insomnia, anhedonia, restlessness, and hopelessness, so we add sertraline, 50 mg/d, to venlafaxine. Ms. R says the seizures are terrifying and she cannot work. She moves in with her parents because she is unable to care for herself.

During a psychiatric appointment, Ms. R confesses that for 2 years her pain has been so unbearable that she has been buying extra tramadol from Internet retailers and taking 600 to 800 mg/d in addition to the prescribed 400 mg/d.

The authors’ observations

Ms. R had a history of chronic pain Table 117 and developed seizures after escalating her tramadol use. After her first epilepsy attack, she did not tell her physicians she was taking additional tramadol nor did she stop taking it. Treatment with several antiepileptics was unsuccessful. Her seizures persisted as long as her tramadol addiction continued.

 

 

Table 1

DSM-IV-TR criteria for pain disorder

  1. Pain in ≥1 anatomical site is the predominant focus of the clinical presentation and is of sufficient severity to warrant clinical attention
  2. The pain causes clinically significant distress or impairment in social, occupational, or other important areas of functioning
  3. Psychological factors are judged to have an important role in the onset, severity, exacerbation, or maintenance of the pain
  4. The symptom or deficit is not intentionally produced or feigned (as in factitious disorder or malingering)
  5. The pain is not better accounted for by a mood, anxiety, or psychotic disorder and does not meet criteria for dyspareunia
Source: Reference 17

Spiller et al18 reported the lowest daily tramadol dose associated with seizures is 500 mg/d, although Talaie et al16 observed seizures at doses as low as 100 mg/d. Additionally, seizure risk may increase through tramadol’s interactions with several medications, including tricyclic antidepressants, selective serotonin reuptake inhibitors, phenothiazines, fluoroquinolone antibiotics, meperidine, clozapine, buspirone, bupropion, phenylephrine, guaifenesin, tripelennamine, thioridazine, theophylline, and acetaminophen, butalbital, and caffeine combination (Table 2).19 Transdermal selegiline is contraindicated with tramadol. For Ms. R, the sertraline and venlafaxine she was taking may have augmented tramadol’s seizure potential.

Table 2

Tramadol: Major drug-drug interactions

DrugSymptoms
SelegilineNausea, vomiting, cardiovascular collapse, respiratory depression, seizures, or serotonin syndrome (hypertension, hyperthermia, myoclonus, mental status changes); use of the transdermal formulation with tramadol is contraindicated
CarbamazepineDecreased tramadol efficacy and increased seizure risk
VenlafaxineIncreased risk of serotonin syndrome
LinezolidIncreased risk of serotonin syndrome
FluoxetineIncreased risk of seizures and serotonin syndrome; increased concentrations of tramadol and decreased concentrations of tramadol active metabolite, O-desmethyltramadol (M1)
OlanzapineIncreased risk of serotonin syndrome
MirtazapineIncreased risk of serotonin syndrome
HaloperidolIncreased risk of seizures
EscitalopramIncreased risk of seizures and serotonin syndrome
ClomipramineIncreased risk of seizures
RisperidoneIncreased risk of seizures
KetamineIncreased risk of respiratory depression and excessive CNS depression
ImipramineIncreased risk of seizures
DuloxetineIncreased risk of serotonin syndrome
NortriptylineIncreased risk of seizures
ClozapineIncreased risk of seizures
SertralineIncreased risk of seizures and serotonin syndrome
ParoxetineIncreased risk of seizures and serotonin syndrome; decrease in the analgesic effect of tramadol
AmitriptylineIncreased risk of seizures; increased concentrations of tramadol and decreased concentrations of tramadol active metabolite, M1
DesipramineIncreased risk of seizures
DoxepinIncreased risk of seizures
CitalopramIncreased risk of seizures and serotonin syndrome
FluvoxamineIncreased risk of seizures and serotonin syndrome
Source: Reference 19

It is important to avoid polypharmacy in patients taking tramadol.20 Most psychiatrists are aware of the risk of serotonin syndrome with antidepressants, but may be less likely to attribute serotonergic additive effects from other medication classes such as analgesics. Recognizing tramadol’s potential to contribute to serotonin syndrome—especially in light of concomitant usage with other serotonergic medications such as antidepressants—is essential.

Tramadol toxicity appears to be caused by monoamine uptake inhibition rather than its opioid effects.21 The most frequent pharmacokinetic drug-drug interactions that lead to side effects such as serotonin syndrome or seizures involve several isoenzymes of the hepatic cytochrome P450 (CYP). The isoenzymes CYP2D6 (substrates—eg, amitriptyline, tramadol, and venlafaxine; inhibitors—eg, fluoxetine and duloxetine) and CYP3A4 (substrates—eg, carbamazepine, oxycodone, and venlafaxine; inductors—eg, carbamazepine; inhibitors, eg—grapefruit juice) are most important clinically.22

Ms. R readily obtained tramadol from Internet retailers. In a 2004 report, a Google search yielded 2,150,000 sources for acquiring tramadol, most of which did not require a prescription.23 Chronic pain patients have a higher prevalence of substance abuse than the general population.24 Because Ms. R did not have a documented substance abuse history, none of her physicians screened her for drug abuse, although toxicology screening wouldn’t have helped because the tramadol had been prescribed. We didn’t think to directly ask Ms. R about medication misuse, but if we had, she might have revealed it sooner.

OUTCOME: Seizure free

With Ms. R’s permission, we speak to her neurologist, who agrees that excess tramadol likely induced her seizures. The seizures stop after Ms. R discontinues tramadol. After 3 months without seizures, phenytoin is discontinued and lamotrigine is tapered to 200 mg/d. Ms. R participates in a pain rehabilitation program and continues to take venlafaxine, 300 mg/d, and sertraline, 50 mg/d. Her mood improves and she returns to work. Her pain is managed by non-steroidal anti-inflammatory drugs because she decides to decrease her activity level. Ms. R also is trying alternative medicine modalities such as acupuncture and acupressure.

Related Resource

  • Clark MR, Treisman GJ. Chronic pain and addiction. Basel, Switzerland: Karger; 2011.

Drug Brand Names

  • Acetaminophen, butalbital, and caffeine • Fioricet
  • Amitriptyline • Elavil
  • Bupropion • Wellbutrin
  • Buspirone • Buspar
  • Carbamazepine • Tegretol, Carbatrol
  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Clozapine • Clozaril
  • Desipramine • Norpramin
  • Doxepin • Adapin, Silenor
  • Duloxetine • Cymbalta
  • Escitalopram • Lexapro
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Guaifenesin • Tenex
  • Haloperidol • Haldol
  • Imipramine • Tofranil
  • Ketamine • Ketalar
  • Lamotrigine • Lamictal
  • Linezolid • Zyvox
  • Meperidine • Demerol
  • Mirtazapine • Remeron
  • Nortriptyline • Aventyl
  • Olanzapine • Zyprexa
  • Oxcarbazepine • Trileptal
  • Oxycodone • Percolone, OxyContin
  • Paroxetine • Paxil
  • Phenylephrine • Lusonal
  • Phenytoin • Dilantin
  • Propoxyphene • Darvon
  • Risperidone • Risperdal
  • Selegiline • Eldepryl, EMSAM
  • Sertraline • Zoloft
  • Theophylline • Aerolate
  • Thioridazine • Mellaril
  • Tramadol • Ultram
  • Tripelennamine • Pyribenzamine
  • Venlafaxine • Effexor
 

 

Disclosure

The authors report no financial relationship with any company whose products are mentioned in this article or with manufactur-ers of competing products.

References

1. Katz KD. Tramadol is an opioid. J Med Toxicol. 2008;4(2):145-

2. Preston KL, Jasinski DR, Testa M. Abuse potential and pharmacological comparison of tramadol and morphine. Drug Alcohol Depend. 1991;27(1):7-17.

3. U.S. Department of Health and Human Services. Substance Abuse and Mental Health Services Administration. Drug abuse warning network, 2009: national estimates of drug-related emergency department visits. Rockville, MD: Substance Abuse and Mental Health Services Administration; 2011. HHS publication (SMA) 11-4659, DAWN Series D-35.

4. Afshari R, Ghooshkhanehee H. Tramadol overdose induced seizure dramatic rise of CPK and acute renal failure. J Pak Med Assoc. 2009;59(3):178.-

5. Rodriguez Villamañan JC, Albaladejo Blanco C, Sanchez Sanchez A, et al. Withdrawal syndrome after long-term treatment with tramadol. Br J Gen Pract. 2000;50(454):406.-

6. Manocha A, Sharma KK, Mediratta PK. On the mechanism of anticonvulsant effect of tramadol in mice. Pharmacol Biochem Behav. 2005;82(1):74-81.

7. Boyd IW. Tramadol and seizures. Med J Aust. 2005;182(11):595-596.

8. Labate A, Newton MR, Vernon GM, et al. Tramadol and new-onset seizures. Med J Aust. 2005;182(1):42-43.

9. Gasse C, Derby L, Vasilakis-Scaramozza C, et al. Incidence of first-time idiopathic seizures in users of tramadol. Pharmacotherapy. 2000;20(6):629-634.

10. Kahn LH, Alderfer RJ, Graham DJ. Seizures reported with tramadol. JAMA. 1997;278(20):1661.-

11. Mazor SS, Feldman KW, Sugar NF, et al. Pediatric tramadol ingestion resulting in seizurelike activity: a case series. Pediatr Emerg Care. 2008;24(6):380-381.

12. Raffa RB, Stone DJ, Jr. Unexceptional seizure potential of tramadol or its enantiomers or metabolites in mice. J Pharmacol Exp Ther. 2008;325(2):500-506.

13. Rehni AK, Singh TG, Singh N, et al. Tramadol-induced seizurogenic effect: a possible role of opioid-dependent histamine H1 receptor activation-linked mechanism. Naunyn Schmiedebergs Arch Pharmacol. 2010;381(1):11-19.

14. Rehni AK, Singh I, Kumar M. Tramadol-induced seizurogenic effect: a possible role of opioid-dependent gamma-aminobutyric acid inhibitory pathway. Basic Clin Pharmacol Toxicol. 2008;103(3):262-266.

15. Jovanović-Cupić V, Martinović Z, Nesić N. Seizures associated with intoxication and abuse of tramadol. Clin Toxicol (Phila). 2006;44(2):143-146.

16. Talaie H, Panahandeh R, Fayaznouri M, et al. Dose-independent occurrence of seizure with tramadol. J Med Toxicol. 2009;5(2):63-67.

17. Diagnostic and statistical manual of mental disorders 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.

18. Spiller HA, Gorman SE, Villalobos D, et al. Prospective multicenter evaluation of tramadol exposure. J Toxicol Clin Toxicol. 1997;35(4):361-364.

19. Reus VI, Rawitscher L. Possible interaction of tramadol and antidepressants. Am J Psychiatry. 2000;157(5):839.-

20. Thundiyil JG, Kearney TE, Olson KR. Evolving epidemiology of drug-induced seizures reported to a Poison Control Center System. J Med Toxicol. 2007;3(1):15-19.

21. Looper KJ. Potential medical and surgical complications of serotonergic antidepressant medications. Psychosomatics. 2007;48(1):1-9.

22. Grond S, Sablotzki A. Clinical pharmacology of tramadol. Clin Pharmacokinet. 2004;43(13):879-923.

23. Lineberry TW, Bostwick JM. Taking the physician out of “physician shopping”: a case series of clinical problems associated with Internet purchases of medication. Mayo Clin Proc. 2004;79(8):1031-1034.

24. Savage SR. Assessment for addiction in pain-treatment settings. Clin J Pain. 2002;18(4 suppl):S28-S38.

References

1. Katz KD. Tramadol is an opioid. J Med Toxicol. 2008;4(2):145-

2. Preston KL, Jasinski DR, Testa M. Abuse potential and pharmacological comparison of tramadol and morphine. Drug Alcohol Depend. 1991;27(1):7-17.

3. U.S. Department of Health and Human Services. Substance Abuse and Mental Health Services Administration. Drug abuse warning network, 2009: national estimates of drug-related emergency department visits. Rockville, MD: Substance Abuse and Mental Health Services Administration; 2011. HHS publication (SMA) 11-4659, DAWN Series D-35.

4. Afshari R, Ghooshkhanehee H. Tramadol overdose induced seizure dramatic rise of CPK and acute renal failure. J Pak Med Assoc. 2009;59(3):178.-

5. Rodriguez Villamañan JC, Albaladejo Blanco C, Sanchez Sanchez A, et al. Withdrawal syndrome after long-term treatment with tramadol. Br J Gen Pract. 2000;50(454):406.-

6. Manocha A, Sharma KK, Mediratta PK. On the mechanism of anticonvulsant effect of tramadol in mice. Pharmacol Biochem Behav. 2005;82(1):74-81.

7. Boyd IW. Tramadol and seizures. Med J Aust. 2005;182(11):595-596.

8. Labate A, Newton MR, Vernon GM, et al. Tramadol and new-onset seizures. Med J Aust. 2005;182(1):42-43.

9. Gasse C, Derby L, Vasilakis-Scaramozza C, et al. Incidence of first-time idiopathic seizures in users of tramadol. Pharmacotherapy. 2000;20(6):629-634.

10. Kahn LH, Alderfer RJ, Graham DJ. Seizures reported with tramadol. JAMA. 1997;278(20):1661.-

11. Mazor SS, Feldman KW, Sugar NF, et al. Pediatric tramadol ingestion resulting in seizurelike activity: a case series. Pediatr Emerg Care. 2008;24(6):380-381.

12. Raffa RB, Stone DJ, Jr. Unexceptional seizure potential of tramadol or its enantiomers or metabolites in mice. J Pharmacol Exp Ther. 2008;325(2):500-506.

13. Rehni AK, Singh TG, Singh N, et al. Tramadol-induced seizurogenic effect: a possible role of opioid-dependent histamine H1 receptor activation-linked mechanism. Naunyn Schmiedebergs Arch Pharmacol. 2010;381(1):11-19.

14. Rehni AK, Singh I, Kumar M. Tramadol-induced seizurogenic effect: a possible role of opioid-dependent gamma-aminobutyric acid inhibitory pathway. Basic Clin Pharmacol Toxicol. 2008;103(3):262-266.

15. Jovanović-Cupić V, Martinović Z, Nesić N. Seizures associated with intoxication and abuse of tramadol. Clin Toxicol (Phila). 2006;44(2):143-146.

16. Talaie H, Panahandeh R, Fayaznouri M, et al. Dose-independent occurrence of seizure with tramadol. J Med Toxicol. 2009;5(2):63-67.

17. Diagnostic and statistical manual of mental disorders 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.

18. Spiller HA, Gorman SE, Villalobos D, et al. Prospective multicenter evaluation of tramadol exposure. J Toxicol Clin Toxicol. 1997;35(4):361-364.

19. Reus VI, Rawitscher L. Possible interaction of tramadol and antidepressants. Am J Psychiatry. 2000;157(5):839.-

20. Thundiyil JG, Kearney TE, Olson KR. Evolving epidemiology of drug-induced seizures reported to a Poison Control Center System. J Med Toxicol. 2007;3(1):15-19.

21. Looper KJ. Potential medical and surgical complications of serotonergic antidepressant medications. Psychosomatics. 2007;48(1):1-9.

22. Grond S, Sablotzki A. Clinical pharmacology of tramadol. Clin Pharmacokinet. 2004;43(13):879-923.

23. Lineberry TW, Bostwick JM. Taking the physician out of “physician shopping”: a case series of clinical problems associated with Internet purchases of medication. Mayo Clin Proc. 2004;79(8):1031-1034.

24. Savage SR. Assessment for addiction in pain-treatment settings. Clin J Pain. 2002;18(4 suppl):S28-S38.

Issue
Current Psychiatry - 11(05)
Issue
Current Psychiatry - 11(05)
Page Number
45-49
Page Number
45-49
Publications
Publications
Topics
Article Type
Display Headline
Epileptic and depressed
Display Headline
Epileptic and depressed
Legacy Keywords
epilepsy; depression; bulimia; anxiety; chronic pain; seizures; tramadol; substance abuse
Legacy Keywords
epilepsy; depression; bulimia; anxiety; chronic pain; seizures; tramadol; substance abuse
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media