Allowed Publications
Slot System
Featured Buckets
Featured Buckets Admin

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

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

How to lower suicide risk in depressed children and adolescents

Article Type
Changed
Tue, 12/11/2018 - 15:04
Display Headline
How to lower suicide risk in depressed children and adolescents

Discuss this article at www.facebook.com/CurrentPsychiatry

Although depression affects nearly 2% of children (age ≤12) and up to 10% of adolescents (age 13 to 18),1 the disorder often is underdiagnosed and undertreated in pediatric patients.2 Treating depression in young patients is challenging. Only 30% to 40% of depressed children and adolescents who receive evidence-based treatment achieve remission.3 In addition, 50% to 70% of those who initially achieve remission will experience recurrence within 5 years.4 Suicide is the third leading cause of death among children and adolescents, and depression greatly increases the likelihood of suicide.5,6

This article reviews assessing and treating depression in children and adolescents, and how to lower suicide risk in pediatric patients.

Symptoms vary with age

Depressive symptoms vary as a function of the child’s cognitive development and social functioning. Hopelessness and vegetative and motivational symptoms may be more frequent in adolescents than in children.7

In preschool-age children, depression manifests indirectly through somatic symptoms and behavioral disturbances. In this age group, sadness or irritability are sensitive and predominant symptoms of depression.8 In older children, sadness and loss of interest in social activities may indicate depression. In adolescents, feelings of mental and physical weariness, aloneness, disconnectedness, uncertainty, vulnerability, anger, irritability, and ambivalence toward friends suggest a depressive disorder.9

Genetic predisposition to depression, poor family support, dysfunctional parenting, and individual vulnerabilities such as poor self-esteem or emotional dysregulation may increase young patients’ risk for depression.10 Peer and family support may protect against depression. Personal competence stemming from social acceptance and body image satisfaction also may be protective factors. A sense of religious and existential well-being (finding meaning and purpose in life) are significantly associated with lower rates of depression among adolescents.11

A persistent illness

The mean duration of a depressive episode in children and adolescents is 7 to 8 months.12 However, subsyndromal depressive symptoms—as well as relapse and recurrence—are common. Long-term studies indicate that many depressed adolescents experience depressive episodes into adulthood.12 Factors that may predict recurrence in adulthood include:

  • severity of depressive episodes
  • concurrent psychotic symptoms
  • suicidal thoughts
  • history of recurrent depressive episodes
  • threshold residual symptoms
  • recent stressful life events
  • adverse family environment
  • family history of depression.12

Early symptom onset, greater depression severity, suicidality, presence of comorbid anxiety, disruptive disorders, and an adverse family environment also predict longer recovery time.12 A study of depressed adolescents found that a history of recurrent depression, family history of recurrent depression, personality disorder traits, and (for girls only) conflict with parents predicted recurrence of depression in young adulthood.4Table 1 summarizes factors that affect depression outcomes in children and adolescents.

Table 1

What affects depression outcomes in children and adolescents?

FactorOutcomes
AgePharmacotherapy and CBT are equally effective in younger and older adolescents.a Although age does not affect long-term treatment outcomes, older adolescents (age 18 to 19) with treatment-resistant depression may respond better to a combination of CBT and medicationb
SexFemales are more likely to experience relapse.c However, sex does not influence response to initial treatmentc
Socioeconomic statusAdolescents with high socioeconomic status are more likely to respond to CBT
Illness characteristicsSeverity of depression is the strongest predictor of poor outcome.d-f Patients with moderate depression are more likely to benefit from CBT added to medication.g However, adding CBT to medication did not affect outcomes in adolescents with self-injurious behavior.b,f Suicidal behaviors during treatment are less frequent when CBT is combined with medicationh,i
Substance abusePatients with substance use disorders are less likely to respond to depression treatmentf and those who continued to abuse substances during treatment are less likely to achieve remission than those who abstainb
Cognitive measuresHigher levels of hopelessness are associated with poor outcomes. For adolescents with treatment-resistant depression who experience hopelessness, adding CBT to pharmacotherapy did not provide additional benefit. Some studies have noted that adolescents with cognitive distortions are more likely to benefit from CBT plus pharmacotherapyb
Family characteristics/environmentHigh family stress is associated with poor treatment outcomes.f Experiencing loss and physically dangerous events does not affect depression outcomes. Trauma and history of abuse adversely effect depression treatment outcomes
CBT: cognitive-behavioral therapy
  1. Birmaher B, Brent D; AACAP Work Group on Quality Issues, et al. Practice parameter for the assessment and treatment of children and adolescents with depressive disorders. J Am Acad Child Adolesc Psychiatry. 2007;46(11):1503-1526.
  2. Emslie GJ, Mayes T, Porta G, et al. Treatment of Resistant Depression in Adolescents (TORDIA): week 24 outcomes. Am J Psychiatry. 2010;167(7):782-791.
  3. Curry J, Silva S, Rohde P, et al. Recovery and recurrence following treatment for adolescent major depression. Arch Gen Psychiatry. 2011;68(3):263-269.
  4. Wilkinson P, Dubicka B, Kelvin R, et al. Treated depression in adolescents: predictors of outcome at 28 weeks. Br J Psychiatry. 2009;194(4):334-341.
  5. Curry J, Rohde P, Simons A, et al. Predictors and moderators of acute outcome in the Treatment for Adolescents with Depression Study (TADS). J Am Acad Child Adolesc Psychiatry. 2006;45(12):1427-1439.
  6. Asarnow JR, Emslie G, Clarke G, et al. Treatment of selective serotonin reuptake inhibitor-resistant depression in adolescents: predictors and moderators of treatment response. J Am Acad Child Adolesc Psychiatry. 2009;48(3):330-339.
  7. Emslie GJ, Kennard BD, Mayes TL. Predictors of treatment response in adolescent depression. Pediatr Ann. 2011;40(6):300-306.
  8. Emslie G, Kratochvil C, Vitiello B, et al. Treatment for Adolescents with Depression Study (TADS): safety results. J Am Acad Child Adolesc Psychiatry. 2006;45(12):1440-1455.
  9. Vitiello B. Combined cognitive-behavioural therapy and pharmacotherapy for adolescent depression: does it improve outcomes compared with monotherapy? CNS Drugs. 2009;23(4):271-280.
 

 

Assessment strategies

Semi-structured interviews such as the Child and Adolescent Psychiatric Assessment, the Diagnostic Interview for Children and Adolescents, and the Kiddie Schedule for Affective Disorder and Schizophrenia are useful for assessing depression in pediatric patients (Table 2).13-16 These tools can be used to assess depression criteria based on information gathered from several sources. Many instruments can be used to assess and monitor pediatric depression, including the Children’s Depression Inventory, the Reynolds Child Depression Scales and Adolescent Depression Scales, and the Child Depression Rating Scale.

To assess suicide risk in depressed younger patients:

  • ask about emotional difficulties
  • identify lack of developmental progress
  • estimate their level of distress
  • detect impairment in functioning
  • estimate the level of danger to themselves and others.17

The best way to assess for suicidal ideation is to ask about it directly while interviewing the patient and his or her parents. Simple questions such as “Have you ever thought about killing yourself or wish you were dead?” and “Have you ever done anything on purpose to hurt or kill yourself?” can be effective.10 These questions are best placed in the middle or toward the end of a list of questions about depressive symptoms.

Adolescents may be more likely than adults to disclose information about suicidality on self-reports.6 However, self-assessment suicide scales are not a substitute for clinical assessment because they tend to be oversensitive and non-specific and lack predictive value. A positive response to either of these questions should prompt a more detailed clinical investigation. There is no evidence that asking about suicide risk increases suicidal behavior, even in high-risk youths.

Table 2

Assessing children and adolescents: 3 semi-structured interviews

InterviewFeatures
Child and Adolescent Psychiatric Assessment13For patients age 9 to 17. Assesses symptoms from the past 3 months. Administration time: 1 to 2 hours. Requires minimal interviewer experience. Assesses impairment in multiple areas (family, peers, school, leisure activities)
Diagnostic Interview for Children and Adolescents14Separate versions for children (age 6 to 12) and adolescents (age 13 to 17). Assesses lifetime psychopathology. Administration time: 1 to 2 hours. Interrater reliability varies (poor to good)
Kiddie Schedule for Affective Disorders and Schizophrenia15Assesses lifetime and current psychopathology. Administration time: 35 minutes to 2.5 hours. Interrater reliability: fair to excellent16

Treatment options

Psychotherapy. Several controlled studies and meta-analyses support the efficacy of cognitive-behavioral therapy (CBT) for mild depression in pediatric patients.18-20 Two recent meta-analyses of CBT studies in depressed adolescents found the mean effect size of CBT was 0.34 to 0.35.19,21 However, a separate analysis found CBT did not have long-term benefits for depressed adolescents, particularly patients with a history of abuse.22

Interpersonal therapy also can be effective in adolescent outpatients with mild to moderate depression. One study found the effect size of psychotherapy was modest (0.36).19

Pharmacotherapy. Two meta-analyses support selective serotonin reuptake inhibitors (SSRIs) for treating mild to moderate depression in children and adolescents. One found 61% of depressed patients age <19 who received an SSRI were “much improved” or “very much improved.”23 Another meta-analysis that compared SSRIs and placebo found fluoxetine was more effective than sertraline or citalopram for depressed adolescents.24 Other studies have shown that for severe depression, the effect size of antidepressants (0.69) is higher than that of placebo (0.39).25 Antidepressants are more effective in adolescents than in children.25

Fluoxetine is the only FDA-approved medication for treating depression in children age ≥8. In 2007 the FDA extended to all antidepressants its “black-box” warning about increased risk of suicidality in patients up to age 24. The results of studies that analyzed data about the safety of antidepressants in children and adolescents have been mixed—some found evidence of increased suicidality with antidepressant use,26,27 whereas others showed no increased risk.28,29Table 3 summarizes steps to minimize the risk of antidepressant-induced suicidality.17

Psychotherapy plus pharmacotherapy. Researchers who compared fluoxetine to CBT and to a combination of the 2 in adolescents with moderate to severe depression found that fluoxetine was most effective in the first 12 weeks of treatment.30 Surprisingly, CBT’s effectiveness was not different from placebo.30 However, studies have shown that combining psychotherapy and medication results in greater symptom improvement,30 faster clinical response,31 improvement of global functioning and quality of life,32 and reduced suicidality.33 At 6 months, the difference in response between medication and psychotherapy was small.25 The Treatment of Resistant Depression in Adolescents study found that for chronic adolescent depression, pharmacotherapy (fluoxetine and venlafaxine) combined with CBT produced a higher response rate than pharmacotherapy alone (54% vs 41%).34

 

 

Table 3

Protecting against antidepressant-induced suicidality

Before initiating antidepressant treatment
  Review the patient’s psychiatric history
  Assess for past suicidal behavior
  Assess for a family history of mental illness or mood disorders and suicide attempts
  Screen for unrecognized bipolar spectrum disorders
  Educate patients and their families to watch for signs of worsening depression or suicidality, and to report such symptoms immediately
During antidepressant treatment
  Pay attention to abrupt changes in symptoms, particularly symptoms that were not part of the patient’s initial presentation
  Watch for deterioration of symptoms
  Monitor for emergence of ‘activating’ symptoms (ie, irritability, impulsivity, anxiety, insomnia, agitation, hostility, akathisia, hypomania, or mania)
  Evaluate the patient’s suicide risk factors, including having a specific plan and/or access to lethal means
  Consider hospitalization if the patient is at high risk for suicide
Source: Reference 17

Lowering suicide risk

Up to 60% of adolescents who commit suicide had a depressive disorder. Risk factors for child and adolescent suicide attempts include:

  • self-harm behaviors
  • psychiatric disorders
  • family disturbances
  • substance abuse
  • physical/sexual abuse.17

How to best manage suicidal youths depends on an adequate assessment of the severity of the patient’s current problems and conflicts and the degree of suicidal intent. Assessment of coping resources, access to support systems, and the attitude of the patient and family toward intervention and follow-up also is important.

Children and adolescents at high risk for suicide—those with a plan or recent suicide attempt with high probability of lethality, stated current intent to kill themselves, or recent suicidal ideation or behavior—may need inpatient psychiatric admission. Although no studies have shown that admitting high-risk suicidal patients prevents suicide, hospitalization often is the safest course of action. Develop ing a comprehensive outpatient treatment plan before discharge is essential. Patients with fewer risk factors, especially those who want help and have social support, hope for the future, and a desire to resolve conflicts, may require only a brief crisis-oriented intervention.

The following recommendations for managing suicidality in children and adolescents are based on clinical experience and have not been empirically validated.

Develop a safety plan to direct the patient’s behavior under various situations. For example, the patient would agree in writing that “If I feel depressed, I will do X, Y, and Z to address it,” or “If I find myself having suicidal thoughts, I will contact ABC.” Having a safety plan lowers the risk of a suicide attempt more than having a suicide contract, which does not give the patient any tools.35

Create a ‘hope box.’ This is a box in which the patient collects mementos and other objects that remind him or her of hope and reasons to live. The patient should be able to access it at all times, so he or she can tap into it during crisis periods to avert suicidal acts.35

Counteract alienation. A sense of social isolation and burdensomeness may be “tipping factors” for suicidal acts when adolescents feel depressed.35 Clinicians should try to help connect patients to meaningful social activities, even in small doses.

Manage overarousal. Overarousal in depressed children and adolescents is manifested as agitation. Insomnia is a clinically modifiable risk factor. Insomnia initially responds well to behavioral interventions such as sleep hygiene, sleep restriction, and stimulus control techniques.35

Related Resources

Drug Brand Names

  • Citalopram • Celexa
  • Fluoxetine • Prozac
  • Sertraline • Zoloft
  • Venlafaxine • Effexor

Disclosures

Drs. Shailesh Jain and Islam report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Rakesh Jain is a consultant to or has received research/grant support from Eli Lilly and Company, Merck, Pfizer Inc., Shionogi Pharmaceuticals, and Shire.

References

1. Birmaher B, Brent D. AACAP Work Group on Quality Issues, et al. Practice parameter for the assessment and treatment of children and adolescents with depressive disorders. J Am Acad Child Adolesc Psychiatry. 2007;46(11):1503-1526.

2. Lewinsohn PM, Clarke GN, Seeley JR, et al. Major depression in community adolescents: age at onset, episode duration, and time to recurrence. J Am Acad Child Adolesc Psychiatry. 1994;33(6):809-818.

3. Emslie GJ, Kennard BD, Mayes TL. Predictors of treatment response in adolescent depression. Pediatr Ann. 2011;40(6):300-306.

4. Lewinsohn PM, Rohde P, Seeley JR, et al. Natural course of adolescent major depressive disorder in a community sample: predictors of recurrence in young adults. Am J Psychiatry. 2000;157(10):1584-1591.

5. Foley DL, Goldston DB, Costello EJ, et al. Proximal psychiatric risk factors for suicidality in youth: the Great Smoky Mountains Study. Arch Gen Psychiatry. 2006;63(9):1017-1024.

6. Gould MS, Greenberg T, Velting DM, et al. Youth suicide risk and preventive interventions: a review of the past 10 years. J Am Acad Child Adolesc Psychiatry. 2003;42(4):386-405.

7. Weiss B, Garber J. Developmental differences in the phenomenology of depression. Dev Psychopathol. 2003;15(2):403-430.

8. Calles JL, Jr. Depression in children and adolescents. Prim Care. 2007;34(2):243-258abstract vi.

9. Farmer TJ. The experience of major depression: adolescents’ perspectives. Issues Ment Health Nurs. 2002;23(6):567-585.

10. Zalsman G, Brent DA, Weersing VR. Depressive disorders in childhood and adolescence: an overview: epidemiology clinical manifestation and risk factors. Child Adolesc Psychiatr Clin N Am. 2006;15(4):827-841, vii.

11. Cotton S, Larkin E, Hoopes A, et al. The impact of adolescent spirituality on depressive symptoms and health risk behaviors. J Adolesc Health. 2005;36(6):529.-

12. Birmaher B, Arbelaez C, Brent D. Course and outcome of child and adolescent major depressive disorder. Child Adolesc Psychiatr Clin N Am. 2002;11(3):619-637, x.

13. Angold A, Costello EJ. A test-retest reliability study of child-reported psychiatric symptoms and diagnoses using the Child and Adolescent Psychiatric Assessment (CAPA-C). Psychol Med. 1995;25(4):755-762.

14. Reich W. Diagnostic interview for children and adolescents (DICA). J Am Acad Child Adolesc Psychiatry. 2000;39(1):59-66.

15. Puig-Antich J, Lukens E, Brent D. Psychosocial schedule for school age children - revised. Pittsburgh PA: Western Psychiatric Institute and Clinic; 1986.

16. Ambrosini PJ. Historical development and present status of the schedule for affective disorders and schizophrenia for school-age children (K-SADS). J Am Acad Child Adolesc Psychiatry. 2000;39(1):49-58.

17. Dodig-Curković K, Curković M, Radić J, et al. Suicidal behavior and suicide among children and adolescents-risk factors and epidemiological characteristics. Coll Antropol. 2010;34(2):771-777.

18. Harrington R, Whittaker J, Shoebridge P, et al. Systematic review of efficacy of cognitive behaviour therapies in childhood and adolescent depressive disorder. BMJ. 1998;316(7144):1559-1563.

19. Weisz JR, McCarty CA, Valeri SM. Effects of psychotherapy for depression in children and adolescents: a meta-analysis. Psychol Bull. 2006;132(1):132-149.

20. Mufson L, Dorta KP, Wickramaratne P, et al. A randomized effectiveness trial of interpersonal psychotherapy for depressed adolescents. Arch Gen Psychiatry. 2004;61(6):577-584.

21. Klein JB, Jacobs RH, Reinecke MA. Cognitive-behavioral therapy for adolescent depression: a meta-analytic investigation of changes in effect-size estimates. J Am Acad Child Adolesc Psychiatry. 2007;46(11):1403-1413.

22. Vitiello B, Emslie G, Clarke G, et al. Long-term outcome of adolescent depression initially resistant to selective serotonin reuptake inhibitor treatment: a follow-up study of the TORDIA sample. J Clin Psychiatry. 2011;72(3):388-396.

23. Bridge JA, Iyengar S, Salary CB, et al. Clinical response and risk for reported suicidal ideation and suicide attempts in pediatric antidepressant treatment: a meta-analysis of randomized controlled trials. JAMA. 2007;297(15):1683-1696.

24. Usala T, Clavenna A, Zuddas A, et al. Randomised controlled trials of selective serotonin reuptake inhibitors in treating depression in children and adolescents: a systematic review and meta-analysis. Eur Neuropsychopharmacol. 2008;18(1):62-73.

25. March JS, Silva S, Petrycki S, et al. The Treatment for Adolescents With Depression Study (TADS): long-term effectiveness and safety outcomes. Arch Gen Psychiatry. 2007;64(10):1132-1143.

26. Bridge JA, Iyengar S, Salary CB, et al. Clinical response and risk for reported suicidal ideation and suicide attempts in pediatric antidepressant treatment: a meta-analysis of randomized controlled trials. JAMA. 2007;297(15):1683-1696.

27. Stone M, Laughren T, Jones ML, et al. Risk of suicidality in clinical trials of antidepressants in adults: analysis of proprietary data submitted to US Food and Drug Administration. BMJ. 2009;339:b2880.-

28. Khan A, Khan S, Kolts R, et al. Suicide rates in clinical trials of SSRIs, other antidepressants, and placebo: analysis of FDA reports. Am J Psychiatry. 2003;160(4):790-792.

29. Simon GE, Savarino J. Suicide attempts among patients starting depression treatment with medications or psychotherapy. Am J Psychiatry. 2007;164(7):1029-1034.

30. March J, Silva S, Petrycki S, et al. Fluoxetine, cognitive-behavioral therapy, and their combination for adolescents with depression: Treatment for Adolescents With Depression Study (TADS) randomized controlled trial. JAMA. 2004;292(7):807-820.

31. Kratochvil C, Emslie G, Silva S, et al. Acute time to response in the Treatment for Adolescents with Depression Study (TADS). J Am Acad Child Adolesc Psychiatry. 2006;45(12):1412-1418.

32. Vitiello B, Rohde P, Silva S, et al. Functioning and quality of life in the Treatment for Adolescents with Depression Study (TADS). J Am Acad Child Adolesc Psychiatry. 2006;45(12):1419-1426.

33. Emslie G, Kratochvil C, Vitiello B, et al. Treatment for Adolescents with Depression Study (TADS): safety results. J Am Acad Child Adolesc Psychiatry. 2006;45(12):1440-1455.

34. Brent D, Emslie G, Clarke G, et al. Switching to another SSRI or to venlafaxine with or without cognitive behavioral therapy for adolescents with SSRI-resistant depression: the TORDIA randomized controlled trial. JAMA. 2008;299(8):901-913.

35. Joiner TE, Ribeiro JD. Assessment and management of suicidal behavior in teens. Psychiatr Ann. 2011;41(4):220-225.

Article PDF
Author and Disclosure Information

Shailesh Jain, MD, MPH, ABDA
Regional Chair, Department of Psychiatry, Associate Professor, Texas Tech University Health Sciences Center, Permian Basin, Odessa, TX
Rakesh Jain, MD, MPH
Director, Psychiatric Drug Research, R/D Research Clinic, Inc., Lake Jackson, TX, Assistant Clinical Professor, Department of Psychiatry, Texas Tech University Health Sciences Center, Permian Basin, Odessa, TX
Jamal Islam, MD, MS
Associate Professor, Regional Vice Chairman, Research Director, Department of Family and Community Medicine, Texas Tech University Health Sciences Center, Permian Basin, Odessa, TX

Issue
Current Psychiatry - 11(05)
Publications
Topics
Page Number
21-32
Legacy Keywords
suicide; suicide risk; depression; children; adolescents; symptoms; interviews; psychotherapy; pharmacotherapy; treatments
Sections
Author and Disclosure Information

Shailesh Jain, MD, MPH, ABDA
Regional Chair, Department of Psychiatry, Associate Professor, Texas Tech University Health Sciences Center, Permian Basin, Odessa, TX
Rakesh Jain, MD, MPH
Director, Psychiatric Drug Research, R/D Research Clinic, Inc., Lake Jackson, TX, Assistant Clinical Professor, Department of Psychiatry, Texas Tech University Health Sciences Center, Permian Basin, Odessa, TX
Jamal Islam, MD, MS
Associate Professor, Regional Vice Chairman, Research Director, Department of Family and Community Medicine, Texas Tech University Health Sciences Center, Permian Basin, Odessa, TX

Author and Disclosure Information

Shailesh Jain, MD, MPH, ABDA
Regional Chair, Department of Psychiatry, Associate Professor, Texas Tech University Health Sciences Center, Permian Basin, Odessa, TX
Rakesh Jain, MD, MPH
Director, Psychiatric Drug Research, R/D Research Clinic, Inc., Lake Jackson, TX, Assistant Clinical Professor, Department of Psychiatry, Texas Tech University Health Sciences Center, Permian Basin, Odessa, TX
Jamal Islam, MD, MS
Associate Professor, Regional Vice Chairman, Research Director, Department of Family and Community Medicine, Texas Tech University Health Sciences Center, Permian Basin, Odessa, TX

Article PDF
Article PDF

Discuss this article at www.facebook.com/CurrentPsychiatry

Although depression affects nearly 2% of children (age ≤12) and up to 10% of adolescents (age 13 to 18),1 the disorder often is underdiagnosed and undertreated in pediatric patients.2 Treating depression in young patients is challenging. Only 30% to 40% of depressed children and adolescents who receive evidence-based treatment achieve remission.3 In addition, 50% to 70% of those who initially achieve remission will experience recurrence within 5 years.4 Suicide is the third leading cause of death among children and adolescents, and depression greatly increases the likelihood of suicide.5,6

This article reviews assessing and treating depression in children and adolescents, and how to lower suicide risk in pediatric patients.

Symptoms vary with age

Depressive symptoms vary as a function of the child’s cognitive development and social functioning. Hopelessness and vegetative and motivational symptoms may be more frequent in adolescents than in children.7

In preschool-age children, depression manifests indirectly through somatic symptoms and behavioral disturbances. In this age group, sadness or irritability are sensitive and predominant symptoms of depression.8 In older children, sadness and loss of interest in social activities may indicate depression. In adolescents, feelings of mental and physical weariness, aloneness, disconnectedness, uncertainty, vulnerability, anger, irritability, and ambivalence toward friends suggest a depressive disorder.9

Genetic predisposition to depression, poor family support, dysfunctional parenting, and individual vulnerabilities such as poor self-esteem or emotional dysregulation may increase young patients’ risk for depression.10 Peer and family support may protect against depression. Personal competence stemming from social acceptance and body image satisfaction also may be protective factors. A sense of religious and existential well-being (finding meaning and purpose in life) are significantly associated with lower rates of depression among adolescents.11

A persistent illness

The mean duration of a depressive episode in children and adolescents is 7 to 8 months.12 However, subsyndromal depressive symptoms—as well as relapse and recurrence—are common. Long-term studies indicate that many depressed adolescents experience depressive episodes into adulthood.12 Factors that may predict recurrence in adulthood include:

  • severity of depressive episodes
  • concurrent psychotic symptoms
  • suicidal thoughts
  • history of recurrent depressive episodes
  • threshold residual symptoms
  • recent stressful life events
  • adverse family environment
  • family history of depression.12

Early symptom onset, greater depression severity, suicidality, presence of comorbid anxiety, disruptive disorders, and an adverse family environment also predict longer recovery time.12 A study of depressed adolescents found that a history of recurrent depression, family history of recurrent depression, personality disorder traits, and (for girls only) conflict with parents predicted recurrence of depression in young adulthood.4Table 1 summarizes factors that affect depression outcomes in children and adolescents.

Table 1

What affects depression outcomes in children and adolescents?

FactorOutcomes
AgePharmacotherapy and CBT are equally effective in younger and older adolescents.a Although age does not affect long-term treatment outcomes, older adolescents (age 18 to 19) with treatment-resistant depression may respond better to a combination of CBT and medicationb
SexFemales are more likely to experience relapse.c However, sex does not influence response to initial treatmentc
Socioeconomic statusAdolescents with high socioeconomic status are more likely to respond to CBT
Illness characteristicsSeverity of depression is the strongest predictor of poor outcome.d-f Patients with moderate depression are more likely to benefit from CBT added to medication.g However, adding CBT to medication did not affect outcomes in adolescents with self-injurious behavior.b,f Suicidal behaviors during treatment are less frequent when CBT is combined with medicationh,i
Substance abusePatients with substance use disorders are less likely to respond to depression treatmentf and those who continued to abuse substances during treatment are less likely to achieve remission than those who abstainb
Cognitive measuresHigher levels of hopelessness are associated with poor outcomes. For adolescents with treatment-resistant depression who experience hopelessness, adding CBT to pharmacotherapy did not provide additional benefit. Some studies have noted that adolescents with cognitive distortions are more likely to benefit from CBT plus pharmacotherapyb
Family characteristics/environmentHigh family stress is associated with poor treatment outcomes.f Experiencing loss and physically dangerous events does not affect depression outcomes. Trauma and history of abuse adversely effect depression treatment outcomes
CBT: cognitive-behavioral therapy
  1. Birmaher B, Brent D; AACAP Work Group on Quality Issues, et al. Practice parameter for the assessment and treatment of children and adolescents with depressive disorders. J Am Acad Child Adolesc Psychiatry. 2007;46(11):1503-1526.
  2. Emslie GJ, Mayes T, Porta G, et al. Treatment of Resistant Depression in Adolescents (TORDIA): week 24 outcomes. Am J Psychiatry. 2010;167(7):782-791.
  3. Curry J, Silva S, Rohde P, et al. Recovery and recurrence following treatment for adolescent major depression. Arch Gen Psychiatry. 2011;68(3):263-269.
  4. Wilkinson P, Dubicka B, Kelvin R, et al. Treated depression in adolescents: predictors of outcome at 28 weeks. Br J Psychiatry. 2009;194(4):334-341.
  5. Curry J, Rohde P, Simons A, et al. Predictors and moderators of acute outcome in the Treatment for Adolescents with Depression Study (TADS). J Am Acad Child Adolesc Psychiatry. 2006;45(12):1427-1439.
  6. Asarnow JR, Emslie G, Clarke G, et al. Treatment of selective serotonin reuptake inhibitor-resistant depression in adolescents: predictors and moderators of treatment response. J Am Acad Child Adolesc Psychiatry. 2009;48(3):330-339.
  7. Emslie GJ, Kennard BD, Mayes TL. Predictors of treatment response in adolescent depression. Pediatr Ann. 2011;40(6):300-306.
  8. Emslie G, Kratochvil C, Vitiello B, et al. Treatment for Adolescents with Depression Study (TADS): safety results. J Am Acad Child Adolesc Psychiatry. 2006;45(12):1440-1455.
  9. Vitiello B. Combined cognitive-behavioural therapy and pharmacotherapy for adolescent depression: does it improve outcomes compared with monotherapy? CNS Drugs. 2009;23(4):271-280.
 

 

Assessment strategies

Semi-structured interviews such as the Child and Adolescent Psychiatric Assessment, the Diagnostic Interview for Children and Adolescents, and the Kiddie Schedule for Affective Disorder and Schizophrenia are useful for assessing depression in pediatric patients (Table 2).13-16 These tools can be used to assess depression criteria based on information gathered from several sources. Many instruments can be used to assess and monitor pediatric depression, including the Children’s Depression Inventory, the Reynolds Child Depression Scales and Adolescent Depression Scales, and the Child Depression Rating Scale.

To assess suicide risk in depressed younger patients:

  • ask about emotional difficulties
  • identify lack of developmental progress
  • estimate their level of distress
  • detect impairment in functioning
  • estimate the level of danger to themselves and others.17

The best way to assess for suicidal ideation is to ask about it directly while interviewing the patient and his or her parents. Simple questions such as “Have you ever thought about killing yourself or wish you were dead?” and “Have you ever done anything on purpose to hurt or kill yourself?” can be effective.10 These questions are best placed in the middle or toward the end of a list of questions about depressive symptoms.

Adolescents may be more likely than adults to disclose information about suicidality on self-reports.6 However, self-assessment suicide scales are not a substitute for clinical assessment because they tend to be oversensitive and non-specific and lack predictive value. A positive response to either of these questions should prompt a more detailed clinical investigation. There is no evidence that asking about suicide risk increases suicidal behavior, even in high-risk youths.

Table 2

Assessing children and adolescents: 3 semi-structured interviews

InterviewFeatures
Child and Adolescent Psychiatric Assessment13For patients age 9 to 17. Assesses symptoms from the past 3 months. Administration time: 1 to 2 hours. Requires minimal interviewer experience. Assesses impairment in multiple areas (family, peers, school, leisure activities)
Diagnostic Interview for Children and Adolescents14Separate versions for children (age 6 to 12) and adolescents (age 13 to 17). Assesses lifetime psychopathology. Administration time: 1 to 2 hours. Interrater reliability varies (poor to good)
Kiddie Schedule for Affective Disorders and Schizophrenia15Assesses lifetime and current psychopathology. Administration time: 35 minutes to 2.5 hours. Interrater reliability: fair to excellent16

Treatment options

Psychotherapy. Several controlled studies and meta-analyses support the efficacy of cognitive-behavioral therapy (CBT) for mild depression in pediatric patients.18-20 Two recent meta-analyses of CBT studies in depressed adolescents found the mean effect size of CBT was 0.34 to 0.35.19,21 However, a separate analysis found CBT did not have long-term benefits for depressed adolescents, particularly patients with a history of abuse.22

Interpersonal therapy also can be effective in adolescent outpatients with mild to moderate depression. One study found the effect size of psychotherapy was modest (0.36).19

Pharmacotherapy. Two meta-analyses support selective serotonin reuptake inhibitors (SSRIs) for treating mild to moderate depression in children and adolescents. One found 61% of depressed patients age <19 who received an SSRI were “much improved” or “very much improved.”23 Another meta-analysis that compared SSRIs and placebo found fluoxetine was more effective than sertraline or citalopram for depressed adolescents.24 Other studies have shown that for severe depression, the effect size of antidepressants (0.69) is higher than that of placebo (0.39).25 Antidepressants are more effective in adolescents than in children.25

Fluoxetine is the only FDA-approved medication for treating depression in children age ≥8. In 2007 the FDA extended to all antidepressants its “black-box” warning about increased risk of suicidality in patients up to age 24. The results of studies that analyzed data about the safety of antidepressants in children and adolescents have been mixed—some found evidence of increased suicidality with antidepressant use,26,27 whereas others showed no increased risk.28,29Table 3 summarizes steps to minimize the risk of antidepressant-induced suicidality.17

Psychotherapy plus pharmacotherapy. Researchers who compared fluoxetine to CBT and to a combination of the 2 in adolescents with moderate to severe depression found that fluoxetine was most effective in the first 12 weeks of treatment.30 Surprisingly, CBT’s effectiveness was not different from placebo.30 However, studies have shown that combining psychotherapy and medication results in greater symptom improvement,30 faster clinical response,31 improvement of global functioning and quality of life,32 and reduced suicidality.33 At 6 months, the difference in response between medication and psychotherapy was small.25 The Treatment of Resistant Depression in Adolescents study found that for chronic adolescent depression, pharmacotherapy (fluoxetine and venlafaxine) combined with CBT produced a higher response rate than pharmacotherapy alone (54% vs 41%).34

 

 

Table 3

Protecting against antidepressant-induced suicidality

Before initiating antidepressant treatment
  Review the patient’s psychiatric history
  Assess for past suicidal behavior
  Assess for a family history of mental illness or mood disorders and suicide attempts
  Screen for unrecognized bipolar spectrum disorders
  Educate patients and their families to watch for signs of worsening depression or suicidality, and to report such symptoms immediately
During antidepressant treatment
  Pay attention to abrupt changes in symptoms, particularly symptoms that were not part of the patient’s initial presentation
  Watch for deterioration of symptoms
  Monitor for emergence of ‘activating’ symptoms (ie, irritability, impulsivity, anxiety, insomnia, agitation, hostility, akathisia, hypomania, or mania)
  Evaluate the patient’s suicide risk factors, including having a specific plan and/or access to lethal means
  Consider hospitalization if the patient is at high risk for suicide
Source: Reference 17

Lowering suicide risk

Up to 60% of adolescents who commit suicide had a depressive disorder. Risk factors for child and adolescent suicide attempts include:

  • self-harm behaviors
  • psychiatric disorders
  • family disturbances
  • substance abuse
  • physical/sexual abuse.17

How to best manage suicidal youths depends on an adequate assessment of the severity of the patient’s current problems and conflicts and the degree of suicidal intent. Assessment of coping resources, access to support systems, and the attitude of the patient and family toward intervention and follow-up also is important.

Children and adolescents at high risk for suicide—those with a plan or recent suicide attempt with high probability of lethality, stated current intent to kill themselves, or recent suicidal ideation or behavior—may need inpatient psychiatric admission. Although no studies have shown that admitting high-risk suicidal patients prevents suicide, hospitalization often is the safest course of action. Develop ing a comprehensive outpatient treatment plan before discharge is essential. Patients with fewer risk factors, especially those who want help and have social support, hope for the future, and a desire to resolve conflicts, may require only a brief crisis-oriented intervention.

The following recommendations for managing suicidality in children and adolescents are based on clinical experience and have not been empirically validated.

Develop a safety plan to direct the patient’s behavior under various situations. For example, the patient would agree in writing that “If I feel depressed, I will do X, Y, and Z to address it,” or “If I find myself having suicidal thoughts, I will contact ABC.” Having a safety plan lowers the risk of a suicide attempt more than having a suicide contract, which does not give the patient any tools.35

Create a ‘hope box.’ This is a box in which the patient collects mementos and other objects that remind him or her of hope and reasons to live. The patient should be able to access it at all times, so he or she can tap into it during crisis periods to avert suicidal acts.35

Counteract alienation. A sense of social isolation and burdensomeness may be “tipping factors” for suicidal acts when adolescents feel depressed.35 Clinicians should try to help connect patients to meaningful social activities, even in small doses.

Manage overarousal. Overarousal in depressed children and adolescents is manifested as agitation. Insomnia is a clinically modifiable risk factor. Insomnia initially responds well to behavioral interventions such as sleep hygiene, sleep restriction, and stimulus control techniques.35

Related Resources

Drug Brand Names

  • Citalopram • Celexa
  • Fluoxetine • Prozac
  • Sertraline • Zoloft
  • Venlafaxine • Effexor

Disclosures

Drs. Shailesh Jain and Islam report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Rakesh Jain is a consultant to or has received research/grant support from Eli Lilly and Company, Merck, Pfizer Inc., Shionogi Pharmaceuticals, and Shire.

Discuss this article at www.facebook.com/CurrentPsychiatry

Although depression affects nearly 2% of children (age ≤12) and up to 10% of adolescents (age 13 to 18),1 the disorder often is underdiagnosed and undertreated in pediatric patients.2 Treating depression in young patients is challenging. Only 30% to 40% of depressed children and adolescents who receive evidence-based treatment achieve remission.3 In addition, 50% to 70% of those who initially achieve remission will experience recurrence within 5 years.4 Suicide is the third leading cause of death among children and adolescents, and depression greatly increases the likelihood of suicide.5,6

This article reviews assessing and treating depression in children and adolescents, and how to lower suicide risk in pediatric patients.

Symptoms vary with age

Depressive symptoms vary as a function of the child’s cognitive development and social functioning. Hopelessness and vegetative and motivational symptoms may be more frequent in adolescents than in children.7

In preschool-age children, depression manifests indirectly through somatic symptoms and behavioral disturbances. In this age group, sadness or irritability are sensitive and predominant symptoms of depression.8 In older children, sadness and loss of interest in social activities may indicate depression. In adolescents, feelings of mental and physical weariness, aloneness, disconnectedness, uncertainty, vulnerability, anger, irritability, and ambivalence toward friends suggest a depressive disorder.9

Genetic predisposition to depression, poor family support, dysfunctional parenting, and individual vulnerabilities such as poor self-esteem or emotional dysregulation may increase young patients’ risk for depression.10 Peer and family support may protect against depression. Personal competence stemming from social acceptance and body image satisfaction also may be protective factors. A sense of religious and existential well-being (finding meaning and purpose in life) are significantly associated with lower rates of depression among adolescents.11

A persistent illness

The mean duration of a depressive episode in children and adolescents is 7 to 8 months.12 However, subsyndromal depressive symptoms—as well as relapse and recurrence—are common. Long-term studies indicate that many depressed adolescents experience depressive episodes into adulthood.12 Factors that may predict recurrence in adulthood include:

  • severity of depressive episodes
  • concurrent psychotic symptoms
  • suicidal thoughts
  • history of recurrent depressive episodes
  • threshold residual symptoms
  • recent stressful life events
  • adverse family environment
  • family history of depression.12

Early symptom onset, greater depression severity, suicidality, presence of comorbid anxiety, disruptive disorders, and an adverse family environment also predict longer recovery time.12 A study of depressed adolescents found that a history of recurrent depression, family history of recurrent depression, personality disorder traits, and (for girls only) conflict with parents predicted recurrence of depression in young adulthood.4Table 1 summarizes factors that affect depression outcomes in children and adolescents.

Table 1

What affects depression outcomes in children and adolescents?

FactorOutcomes
AgePharmacotherapy and CBT are equally effective in younger and older adolescents.a Although age does not affect long-term treatment outcomes, older adolescents (age 18 to 19) with treatment-resistant depression may respond better to a combination of CBT and medicationb
SexFemales are more likely to experience relapse.c However, sex does not influence response to initial treatmentc
Socioeconomic statusAdolescents with high socioeconomic status are more likely to respond to CBT
Illness characteristicsSeverity of depression is the strongest predictor of poor outcome.d-f Patients with moderate depression are more likely to benefit from CBT added to medication.g However, adding CBT to medication did not affect outcomes in adolescents with self-injurious behavior.b,f Suicidal behaviors during treatment are less frequent when CBT is combined with medicationh,i
Substance abusePatients with substance use disorders are less likely to respond to depression treatmentf and those who continued to abuse substances during treatment are less likely to achieve remission than those who abstainb
Cognitive measuresHigher levels of hopelessness are associated with poor outcomes. For adolescents with treatment-resistant depression who experience hopelessness, adding CBT to pharmacotherapy did not provide additional benefit. Some studies have noted that adolescents with cognitive distortions are more likely to benefit from CBT plus pharmacotherapyb
Family characteristics/environmentHigh family stress is associated with poor treatment outcomes.f Experiencing loss and physically dangerous events does not affect depression outcomes. Trauma and history of abuse adversely effect depression treatment outcomes
CBT: cognitive-behavioral therapy
  1. Birmaher B, Brent D; AACAP Work Group on Quality Issues, et al. Practice parameter for the assessment and treatment of children and adolescents with depressive disorders. J Am Acad Child Adolesc Psychiatry. 2007;46(11):1503-1526.
  2. Emslie GJ, Mayes T, Porta G, et al. Treatment of Resistant Depression in Adolescents (TORDIA): week 24 outcomes. Am J Psychiatry. 2010;167(7):782-791.
  3. Curry J, Silva S, Rohde P, et al. Recovery and recurrence following treatment for adolescent major depression. Arch Gen Psychiatry. 2011;68(3):263-269.
  4. Wilkinson P, Dubicka B, Kelvin R, et al. Treated depression in adolescents: predictors of outcome at 28 weeks. Br J Psychiatry. 2009;194(4):334-341.
  5. Curry J, Rohde P, Simons A, et al. Predictors and moderators of acute outcome in the Treatment for Adolescents with Depression Study (TADS). J Am Acad Child Adolesc Psychiatry. 2006;45(12):1427-1439.
  6. Asarnow JR, Emslie G, Clarke G, et al. Treatment of selective serotonin reuptake inhibitor-resistant depression in adolescents: predictors and moderators of treatment response. J Am Acad Child Adolesc Psychiatry. 2009;48(3):330-339.
  7. Emslie GJ, Kennard BD, Mayes TL. Predictors of treatment response in adolescent depression. Pediatr Ann. 2011;40(6):300-306.
  8. Emslie G, Kratochvil C, Vitiello B, et al. Treatment for Adolescents with Depression Study (TADS): safety results. J Am Acad Child Adolesc Psychiatry. 2006;45(12):1440-1455.
  9. Vitiello B. Combined cognitive-behavioural therapy and pharmacotherapy for adolescent depression: does it improve outcomes compared with monotherapy? CNS Drugs. 2009;23(4):271-280.
 

 

Assessment strategies

Semi-structured interviews such as the Child and Adolescent Psychiatric Assessment, the Diagnostic Interview for Children and Adolescents, and the Kiddie Schedule for Affective Disorder and Schizophrenia are useful for assessing depression in pediatric patients (Table 2).13-16 These tools can be used to assess depression criteria based on information gathered from several sources. Many instruments can be used to assess and monitor pediatric depression, including the Children’s Depression Inventory, the Reynolds Child Depression Scales and Adolescent Depression Scales, and the Child Depression Rating Scale.

To assess suicide risk in depressed younger patients:

  • ask about emotional difficulties
  • identify lack of developmental progress
  • estimate their level of distress
  • detect impairment in functioning
  • estimate the level of danger to themselves and others.17

The best way to assess for suicidal ideation is to ask about it directly while interviewing the patient and his or her parents. Simple questions such as “Have you ever thought about killing yourself or wish you were dead?” and “Have you ever done anything on purpose to hurt or kill yourself?” can be effective.10 These questions are best placed in the middle or toward the end of a list of questions about depressive symptoms.

Adolescents may be more likely than adults to disclose information about suicidality on self-reports.6 However, self-assessment suicide scales are not a substitute for clinical assessment because they tend to be oversensitive and non-specific and lack predictive value. A positive response to either of these questions should prompt a more detailed clinical investigation. There is no evidence that asking about suicide risk increases suicidal behavior, even in high-risk youths.

Table 2

Assessing children and adolescents: 3 semi-structured interviews

InterviewFeatures
Child and Adolescent Psychiatric Assessment13For patients age 9 to 17. Assesses symptoms from the past 3 months. Administration time: 1 to 2 hours. Requires minimal interviewer experience. Assesses impairment in multiple areas (family, peers, school, leisure activities)
Diagnostic Interview for Children and Adolescents14Separate versions for children (age 6 to 12) and adolescents (age 13 to 17). Assesses lifetime psychopathology. Administration time: 1 to 2 hours. Interrater reliability varies (poor to good)
Kiddie Schedule for Affective Disorders and Schizophrenia15Assesses lifetime and current psychopathology. Administration time: 35 minutes to 2.5 hours. Interrater reliability: fair to excellent16

Treatment options

Psychotherapy. Several controlled studies and meta-analyses support the efficacy of cognitive-behavioral therapy (CBT) for mild depression in pediatric patients.18-20 Two recent meta-analyses of CBT studies in depressed adolescents found the mean effect size of CBT was 0.34 to 0.35.19,21 However, a separate analysis found CBT did not have long-term benefits for depressed adolescents, particularly patients with a history of abuse.22

Interpersonal therapy also can be effective in adolescent outpatients with mild to moderate depression. One study found the effect size of psychotherapy was modest (0.36).19

Pharmacotherapy. Two meta-analyses support selective serotonin reuptake inhibitors (SSRIs) for treating mild to moderate depression in children and adolescents. One found 61% of depressed patients age <19 who received an SSRI were “much improved” or “very much improved.”23 Another meta-analysis that compared SSRIs and placebo found fluoxetine was more effective than sertraline or citalopram for depressed adolescents.24 Other studies have shown that for severe depression, the effect size of antidepressants (0.69) is higher than that of placebo (0.39).25 Antidepressants are more effective in adolescents than in children.25

Fluoxetine is the only FDA-approved medication for treating depression in children age ≥8. In 2007 the FDA extended to all antidepressants its “black-box” warning about increased risk of suicidality in patients up to age 24. The results of studies that analyzed data about the safety of antidepressants in children and adolescents have been mixed—some found evidence of increased suicidality with antidepressant use,26,27 whereas others showed no increased risk.28,29Table 3 summarizes steps to minimize the risk of antidepressant-induced suicidality.17

Psychotherapy plus pharmacotherapy. Researchers who compared fluoxetine to CBT and to a combination of the 2 in adolescents with moderate to severe depression found that fluoxetine was most effective in the first 12 weeks of treatment.30 Surprisingly, CBT’s effectiveness was not different from placebo.30 However, studies have shown that combining psychotherapy and medication results in greater symptom improvement,30 faster clinical response,31 improvement of global functioning and quality of life,32 and reduced suicidality.33 At 6 months, the difference in response between medication and psychotherapy was small.25 The Treatment of Resistant Depression in Adolescents study found that for chronic adolescent depression, pharmacotherapy (fluoxetine and venlafaxine) combined with CBT produced a higher response rate than pharmacotherapy alone (54% vs 41%).34

 

 

Table 3

Protecting against antidepressant-induced suicidality

Before initiating antidepressant treatment
  Review the patient’s psychiatric history
  Assess for past suicidal behavior
  Assess for a family history of mental illness or mood disorders and suicide attempts
  Screen for unrecognized bipolar spectrum disorders
  Educate patients and their families to watch for signs of worsening depression or suicidality, and to report such symptoms immediately
During antidepressant treatment
  Pay attention to abrupt changes in symptoms, particularly symptoms that were not part of the patient’s initial presentation
  Watch for deterioration of symptoms
  Monitor for emergence of ‘activating’ symptoms (ie, irritability, impulsivity, anxiety, insomnia, agitation, hostility, akathisia, hypomania, or mania)
  Evaluate the patient’s suicide risk factors, including having a specific plan and/or access to lethal means
  Consider hospitalization if the patient is at high risk for suicide
Source: Reference 17

Lowering suicide risk

Up to 60% of adolescents who commit suicide had a depressive disorder. Risk factors for child and adolescent suicide attempts include:

  • self-harm behaviors
  • psychiatric disorders
  • family disturbances
  • substance abuse
  • physical/sexual abuse.17

How to best manage suicidal youths depends on an adequate assessment of the severity of the patient’s current problems and conflicts and the degree of suicidal intent. Assessment of coping resources, access to support systems, and the attitude of the patient and family toward intervention and follow-up also is important.

Children and adolescents at high risk for suicide—those with a plan or recent suicide attempt with high probability of lethality, stated current intent to kill themselves, or recent suicidal ideation or behavior—may need inpatient psychiatric admission. Although no studies have shown that admitting high-risk suicidal patients prevents suicide, hospitalization often is the safest course of action. Develop ing a comprehensive outpatient treatment plan before discharge is essential. Patients with fewer risk factors, especially those who want help and have social support, hope for the future, and a desire to resolve conflicts, may require only a brief crisis-oriented intervention.

The following recommendations for managing suicidality in children and adolescents are based on clinical experience and have not been empirically validated.

Develop a safety plan to direct the patient’s behavior under various situations. For example, the patient would agree in writing that “If I feel depressed, I will do X, Y, and Z to address it,” or “If I find myself having suicidal thoughts, I will contact ABC.” Having a safety plan lowers the risk of a suicide attempt more than having a suicide contract, which does not give the patient any tools.35

Create a ‘hope box.’ This is a box in which the patient collects mementos and other objects that remind him or her of hope and reasons to live. The patient should be able to access it at all times, so he or she can tap into it during crisis periods to avert suicidal acts.35

Counteract alienation. A sense of social isolation and burdensomeness may be “tipping factors” for suicidal acts when adolescents feel depressed.35 Clinicians should try to help connect patients to meaningful social activities, even in small doses.

Manage overarousal. Overarousal in depressed children and adolescents is manifested as agitation. Insomnia is a clinically modifiable risk factor. Insomnia initially responds well to behavioral interventions such as sleep hygiene, sleep restriction, and stimulus control techniques.35

Related Resources

Drug Brand Names

  • Citalopram • Celexa
  • Fluoxetine • Prozac
  • Sertraline • Zoloft
  • Venlafaxine • Effexor

Disclosures

Drs. Shailesh Jain and Islam report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Rakesh Jain is a consultant to or has received research/grant support from Eli Lilly and Company, Merck, Pfizer Inc., Shionogi Pharmaceuticals, and Shire.

References

1. Birmaher B, Brent D. AACAP Work Group on Quality Issues, et al. Practice parameter for the assessment and treatment of children and adolescents with depressive disorders. J Am Acad Child Adolesc Psychiatry. 2007;46(11):1503-1526.

2. Lewinsohn PM, Clarke GN, Seeley JR, et al. Major depression in community adolescents: age at onset, episode duration, and time to recurrence. J Am Acad Child Adolesc Psychiatry. 1994;33(6):809-818.

3. Emslie GJ, Kennard BD, Mayes TL. Predictors of treatment response in adolescent depression. Pediatr Ann. 2011;40(6):300-306.

4. Lewinsohn PM, Rohde P, Seeley JR, et al. Natural course of adolescent major depressive disorder in a community sample: predictors of recurrence in young adults. Am J Psychiatry. 2000;157(10):1584-1591.

5. Foley DL, Goldston DB, Costello EJ, et al. Proximal psychiatric risk factors for suicidality in youth: the Great Smoky Mountains Study. Arch Gen Psychiatry. 2006;63(9):1017-1024.

6. Gould MS, Greenberg T, Velting DM, et al. Youth suicide risk and preventive interventions: a review of the past 10 years. J Am Acad Child Adolesc Psychiatry. 2003;42(4):386-405.

7. Weiss B, Garber J. Developmental differences in the phenomenology of depression. Dev Psychopathol. 2003;15(2):403-430.

8. Calles JL, Jr. Depression in children and adolescents. Prim Care. 2007;34(2):243-258abstract vi.

9. Farmer TJ. The experience of major depression: adolescents’ perspectives. Issues Ment Health Nurs. 2002;23(6):567-585.

10. Zalsman G, Brent DA, Weersing VR. Depressive disorders in childhood and adolescence: an overview: epidemiology clinical manifestation and risk factors. Child Adolesc Psychiatr Clin N Am. 2006;15(4):827-841, vii.

11. Cotton S, Larkin E, Hoopes A, et al. The impact of adolescent spirituality on depressive symptoms and health risk behaviors. J Adolesc Health. 2005;36(6):529.-

12. Birmaher B, Arbelaez C, Brent D. Course and outcome of child and adolescent major depressive disorder. Child Adolesc Psychiatr Clin N Am. 2002;11(3):619-637, x.

13. Angold A, Costello EJ. A test-retest reliability study of child-reported psychiatric symptoms and diagnoses using the Child and Adolescent Psychiatric Assessment (CAPA-C). Psychol Med. 1995;25(4):755-762.

14. Reich W. Diagnostic interview for children and adolescents (DICA). J Am Acad Child Adolesc Psychiatry. 2000;39(1):59-66.

15. Puig-Antich J, Lukens E, Brent D. Psychosocial schedule for school age children - revised. Pittsburgh PA: Western Psychiatric Institute and Clinic; 1986.

16. Ambrosini PJ. Historical development and present status of the schedule for affective disorders and schizophrenia for school-age children (K-SADS). J Am Acad Child Adolesc Psychiatry. 2000;39(1):49-58.

17. Dodig-Curković K, Curković M, Radić J, et al. Suicidal behavior and suicide among children and adolescents-risk factors and epidemiological characteristics. Coll Antropol. 2010;34(2):771-777.

18. Harrington R, Whittaker J, Shoebridge P, et al. Systematic review of efficacy of cognitive behaviour therapies in childhood and adolescent depressive disorder. BMJ. 1998;316(7144):1559-1563.

19. Weisz JR, McCarty CA, Valeri SM. Effects of psychotherapy for depression in children and adolescents: a meta-analysis. Psychol Bull. 2006;132(1):132-149.

20. Mufson L, Dorta KP, Wickramaratne P, et al. A randomized effectiveness trial of interpersonal psychotherapy for depressed adolescents. Arch Gen Psychiatry. 2004;61(6):577-584.

21. Klein JB, Jacobs RH, Reinecke MA. Cognitive-behavioral therapy for adolescent depression: a meta-analytic investigation of changes in effect-size estimates. J Am Acad Child Adolesc Psychiatry. 2007;46(11):1403-1413.

22. Vitiello B, Emslie G, Clarke G, et al. Long-term outcome of adolescent depression initially resistant to selective serotonin reuptake inhibitor treatment: a follow-up study of the TORDIA sample. J Clin Psychiatry. 2011;72(3):388-396.

23. Bridge JA, Iyengar S, Salary CB, et al. Clinical response and risk for reported suicidal ideation and suicide attempts in pediatric antidepressant treatment: a meta-analysis of randomized controlled trials. JAMA. 2007;297(15):1683-1696.

24. Usala T, Clavenna A, Zuddas A, et al. Randomised controlled trials of selective serotonin reuptake inhibitors in treating depression in children and adolescents: a systematic review and meta-analysis. Eur Neuropsychopharmacol. 2008;18(1):62-73.

25. March JS, Silva S, Petrycki S, et al. The Treatment for Adolescents With Depression Study (TADS): long-term effectiveness and safety outcomes. Arch Gen Psychiatry. 2007;64(10):1132-1143.

26. Bridge JA, Iyengar S, Salary CB, et al. Clinical response and risk for reported suicidal ideation and suicide attempts in pediatric antidepressant treatment: a meta-analysis of randomized controlled trials. JAMA. 2007;297(15):1683-1696.

27. Stone M, Laughren T, Jones ML, et al. Risk of suicidality in clinical trials of antidepressants in adults: analysis of proprietary data submitted to US Food and Drug Administration. BMJ. 2009;339:b2880.-

28. Khan A, Khan S, Kolts R, et al. Suicide rates in clinical trials of SSRIs, other antidepressants, and placebo: analysis of FDA reports. Am J Psychiatry. 2003;160(4):790-792.

29. Simon GE, Savarino J. Suicide attempts among patients starting depression treatment with medications or psychotherapy. Am J Psychiatry. 2007;164(7):1029-1034.

30. March J, Silva S, Petrycki S, et al. Fluoxetine, cognitive-behavioral therapy, and their combination for adolescents with depression: Treatment for Adolescents With Depression Study (TADS) randomized controlled trial. JAMA. 2004;292(7):807-820.

31. Kratochvil C, Emslie G, Silva S, et al. Acute time to response in the Treatment for Adolescents with Depression Study (TADS). J Am Acad Child Adolesc Psychiatry. 2006;45(12):1412-1418.

32. Vitiello B, Rohde P, Silva S, et al. Functioning and quality of life in the Treatment for Adolescents with Depression Study (TADS). J Am Acad Child Adolesc Psychiatry. 2006;45(12):1419-1426.

33. Emslie G, Kratochvil C, Vitiello B, et al. Treatment for Adolescents with Depression Study (TADS): safety results. J Am Acad Child Adolesc Psychiatry. 2006;45(12):1440-1455.

34. Brent D, Emslie G, Clarke G, et al. Switching to another SSRI or to venlafaxine with or without cognitive behavioral therapy for adolescents with SSRI-resistant depression: the TORDIA randomized controlled trial. JAMA. 2008;299(8):901-913.

35. Joiner TE, Ribeiro JD. Assessment and management of suicidal behavior in teens. Psychiatr Ann. 2011;41(4):220-225.

References

1. Birmaher B, Brent D. AACAP Work Group on Quality Issues, et al. Practice parameter for the assessment and treatment of children and adolescents with depressive disorders. J Am Acad Child Adolesc Psychiatry. 2007;46(11):1503-1526.

2. Lewinsohn PM, Clarke GN, Seeley JR, et al. Major depression in community adolescents: age at onset, episode duration, and time to recurrence. J Am Acad Child Adolesc Psychiatry. 1994;33(6):809-818.

3. Emslie GJ, Kennard BD, Mayes TL. Predictors of treatment response in adolescent depression. Pediatr Ann. 2011;40(6):300-306.

4. Lewinsohn PM, Rohde P, Seeley JR, et al. Natural course of adolescent major depressive disorder in a community sample: predictors of recurrence in young adults. Am J Psychiatry. 2000;157(10):1584-1591.

5. Foley DL, Goldston DB, Costello EJ, et al. Proximal psychiatric risk factors for suicidality in youth: the Great Smoky Mountains Study. Arch Gen Psychiatry. 2006;63(9):1017-1024.

6. Gould MS, Greenberg T, Velting DM, et al. Youth suicide risk and preventive interventions: a review of the past 10 years. J Am Acad Child Adolesc Psychiatry. 2003;42(4):386-405.

7. Weiss B, Garber J. Developmental differences in the phenomenology of depression. Dev Psychopathol. 2003;15(2):403-430.

8. Calles JL, Jr. Depression in children and adolescents. Prim Care. 2007;34(2):243-258abstract vi.

9. Farmer TJ. The experience of major depression: adolescents’ perspectives. Issues Ment Health Nurs. 2002;23(6):567-585.

10. Zalsman G, Brent DA, Weersing VR. Depressive disorders in childhood and adolescence: an overview: epidemiology clinical manifestation and risk factors. Child Adolesc Psychiatr Clin N Am. 2006;15(4):827-841, vii.

11. Cotton S, Larkin E, Hoopes A, et al. The impact of adolescent spirituality on depressive symptoms and health risk behaviors. J Adolesc Health. 2005;36(6):529.-

12. Birmaher B, Arbelaez C, Brent D. Course and outcome of child and adolescent major depressive disorder. Child Adolesc Psychiatr Clin N Am. 2002;11(3):619-637, x.

13. Angold A, Costello EJ. A test-retest reliability study of child-reported psychiatric symptoms and diagnoses using the Child and Adolescent Psychiatric Assessment (CAPA-C). Psychol Med. 1995;25(4):755-762.

14. Reich W. Diagnostic interview for children and adolescents (DICA). J Am Acad Child Adolesc Psychiatry. 2000;39(1):59-66.

15. Puig-Antich J, Lukens E, Brent D. Psychosocial schedule for school age children - revised. Pittsburgh PA: Western Psychiatric Institute and Clinic; 1986.

16. Ambrosini PJ. Historical development and present status of the schedule for affective disorders and schizophrenia for school-age children (K-SADS). J Am Acad Child Adolesc Psychiatry. 2000;39(1):49-58.

17. Dodig-Curković K, Curković M, Radić J, et al. Suicidal behavior and suicide among children and adolescents-risk factors and epidemiological characteristics. Coll Antropol. 2010;34(2):771-777.

18. Harrington R, Whittaker J, Shoebridge P, et al. Systematic review of efficacy of cognitive behaviour therapies in childhood and adolescent depressive disorder. BMJ. 1998;316(7144):1559-1563.

19. Weisz JR, McCarty CA, Valeri SM. Effects of psychotherapy for depression in children and adolescents: a meta-analysis. Psychol Bull. 2006;132(1):132-149.

20. Mufson L, Dorta KP, Wickramaratne P, et al. A randomized effectiveness trial of interpersonal psychotherapy for depressed adolescents. Arch Gen Psychiatry. 2004;61(6):577-584.

21. Klein JB, Jacobs RH, Reinecke MA. Cognitive-behavioral therapy for adolescent depression: a meta-analytic investigation of changes in effect-size estimates. J Am Acad Child Adolesc Psychiatry. 2007;46(11):1403-1413.

22. Vitiello B, Emslie G, Clarke G, et al. Long-term outcome of adolescent depression initially resistant to selective serotonin reuptake inhibitor treatment: a follow-up study of the TORDIA sample. J Clin Psychiatry. 2011;72(3):388-396.

23. Bridge JA, Iyengar S, Salary CB, et al. Clinical response and risk for reported suicidal ideation and suicide attempts in pediatric antidepressant treatment: a meta-analysis of randomized controlled trials. JAMA. 2007;297(15):1683-1696.

24. Usala T, Clavenna A, Zuddas A, et al. Randomised controlled trials of selective serotonin reuptake inhibitors in treating depression in children and adolescents: a systematic review and meta-analysis. Eur Neuropsychopharmacol. 2008;18(1):62-73.

25. March JS, Silva S, Petrycki S, et al. The Treatment for Adolescents With Depression Study (TADS): long-term effectiveness and safety outcomes. Arch Gen Psychiatry. 2007;64(10):1132-1143.

26. Bridge JA, Iyengar S, Salary CB, et al. Clinical response and risk for reported suicidal ideation and suicide attempts in pediatric antidepressant treatment: a meta-analysis of randomized controlled trials. JAMA. 2007;297(15):1683-1696.

27. Stone M, Laughren T, Jones ML, et al. Risk of suicidality in clinical trials of antidepressants in adults: analysis of proprietary data submitted to US Food and Drug Administration. BMJ. 2009;339:b2880.-

28. Khan A, Khan S, Kolts R, et al. Suicide rates in clinical trials of SSRIs, other antidepressants, and placebo: analysis of FDA reports. Am J Psychiatry. 2003;160(4):790-792.

29. Simon GE, Savarino J. Suicide attempts among patients starting depression treatment with medications or psychotherapy. Am J Psychiatry. 2007;164(7):1029-1034.

30. March J, Silva S, Petrycki S, et al. Fluoxetine, cognitive-behavioral therapy, and their combination for adolescents with depression: Treatment for Adolescents With Depression Study (TADS) randomized controlled trial. JAMA. 2004;292(7):807-820.

31. Kratochvil C, Emslie G, Silva S, et al. Acute time to response in the Treatment for Adolescents with Depression Study (TADS). J Am Acad Child Adolesc Psychiatry. 2006;45(12):1412-1418.

32. Vitiello B, Rohde P, Silva S, et al. Functioning and quality of life in the Treatment for Adolescents with Depression Study (TADS). J Am Acad Child Adolesc Psychiatry. 2006;45(12):1419-1426.

33. Emslie G, Kratochvil C, Vitiello B, et al. Treatment for Adolescents with Depression Study (TADS): safety results. J Am Acad Child Adolesc Psychiatry. 2006;45(12):1440-1455.

34. Brent D, Emslie G, Clarke G, et al. Switching to another SSRI or to venlafaxine with or without cognitive behavioral therapy for adolescents with SSRI-resistant depression: the TORDIA randomized controlled trial. JAMA. 2008;299(8):901-913.

35. Joiner TE, Ribeiro JD. Assessment and management of suicidal behavior in teens. Psychiatr Ann. 2011;41(4):220-225.

Issue
Current Psychiatry - 11(05)
Issue
Current Psychiatry - 11(05)
Page Number
21-32
Page Number
21-32
Publications
Publications
Topics
Article Type
Display Headline
How to lower suicide risk in depressed children and adolescents
Display Headline
How to lower suicide risk in depressed children and adolescents
Legacy Keywords
suicide; suicide risk; depression; children; adolescents; symptoms; interviews; psychotherapy; pharmacotherapy; treatments
Legacy Keywords
suicide; suicide risk; depression; children; adolescents; symptoms; interviews; psychotherapy; pharmacotherapy; treatments
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Binge eating disorder: Evidence-based treatments

Article Type
Changed
Tue, 12/11/2018 - 15:04
Display Headline
Binge eating disorder: Evidence-based treatments

Binge eating is consumption of an unusually large amount of food coupled with a feeling of loss of control over eating. Binge eating disorder (BED) is characterized by recurrent episodes of binge eating without inappropriate compensatory behaviors (eg, self-induced vomiting, misuse of laxatives, diuretics, or other agents, excessive exercise).1 It is the most common eating disorder in the United States, with a lifetime prevalence of approximately 3.5% in women and 2% in men.2 The diagnosis falls within the DSM-IV-TR category of eating disorders not otherwise specified,1 but clinicians often view it as a distinct clinical phenomenon. In DSM-IV-TR, an individual would meet criteria for BED if he or she engages in regular binge eating behavior in the absence of recurrent compensatory behaviors ≥2 days per week over 6 months.1 Proposed changes for DSM-5 recognize a distinct BED diagnosis, reduce the frequency criterion to once per week and the duration criterion to the past 3 months, and shift the focus from binge days to binge episodes (Table 1).3

Table 1

Proposed DSM-5 criteria for binge eating disorder

  1. Recurrent episodes of binge eating. An episode of binge eating is characterized by both of the following:
  2. The binge-eating episodes are associated with ≥3 of the following:
  3. Marked distress regarding binge eating is present
  4. The binge eating occurs, on average, at least once a week for 3 months
  5. The binge eating is not associated with the recurrent use of inappropriate compensatory behavior (for example, purging) and does not occur exclusively during the course anorexia nervosa, bulimia nervosa, or avoidant/restrictive food intake disorder
Source:Reference 3

BED can occur in individuals of all body mass indices (BMI), but is common among individuals who are overweight or obese as well as those with depression or type 2 diabetes; BED can complicate treatment of these conditions.2,4,5 Primary treatment goals are:

  • abstinence from binge eating
  • improved psychological functioning
  • appropriate weight regulation in overweight patients.

We report on 3 approaches to BED treatment: medication only, behavioral intervention only, and medication plus behavioral intervention. This article provides insights about emerging changes in diagnostic criteria for BED as well as evidence-informed treatment options and recommendations.

The evidence base

We conducted a review of 23 BED studies: 7 medication only, 5 medication plus behavioral, and 11 behavioral only. We focused on studies conducted since September 2005 that included binge frequency, weight, and depression as primary outcomes (see Berkman et al6 for a review of BED treatment studies before 2005). The studies included 2,527 participants (2,216 women and 311 men). Although the sex distribution of BED in the general population tends to slightly favor women,2 the proportion of women presenting for treatment generally is considerably higher than that of men. In studies that reported on race and/or ethnicity, 1,639 participants were identified as white, 191 as African American, 25 as Hispanic, 2 as Asian, 1 as Native American, and 25 as “other.” Ages ranged from 18 to 77.

Several medications are effective

In placebo-controlled studies, a high-dose selective serotonin reuptake inhibitor (escitalopram7), 2 anticonvulsants (zonisamide8 and topiramate9), a selective norepinephrine reuptake inhibitor (atomoxetine10), and an appetite suppressant (sibutramine11) were associated with significant decreases in binge eating frequency, weight, and BMI in overweight/obese patients diagnosed with BED (Table 2). In an open-label trial, memantine—a N-methyl-D-aspartate receptor antagonist often used to treat symptoms of Alzheimer’s disease—was associated with a significant reduction in binge eating but no change in weight.12 Lamotrigine was not significantly different from placebo in reducing binge eating or weight, but showed promise in reducing metabolic parameters such as glucose and triglyceride levels commonly associated with obesity and type 2 diabetes.13 Because BED often is comorbid with obesity and type 2 diabetes, lamotrigine augmentation when treating obese individuals with BED warrants further investigation. As with any pharmacologic agent, carefully consider potential side effects and interactions with other drugs before prescribing medications for BED. Informing patients of potential side effects is crucial for patient safety and accuracy of the data collected in well-controlled treatment studies.

Table 2

Pharmacotherapy for binge eating disorder

StudyDrug/dosageComments
Guerdjikova et al, 20087Escitalopram, 10 to 30 mg/d, vs placebo for 12 weeksEscitalopram was significantly better than placebo in reducing weight, BMI, and illness severity
McElroy et al, 20068Zonisamide, 100 to 600 mg/d, vs placebo for 16 weeksZonisamide was significantly better than placebo in reducing BE, weight, BMI, and various aspects of unhealthy eating behavior
McElroy et al, 20079Topiramate, 25 to 400 mg/d, vs placebo for 16 weeksTopiramate was significantly better than placebo in reducing BE, weight, BMI, and related psychological features of BE
McElroy et al, 200710Atomoxetine, 40 to 120 mg/d, vs placebo for 10 weeksAtomoxetine was significantly better than placebo in reducing BE, weight, BMI, and obsessive-compulsive features of BE, and in achieving remission
Wilfley et al, 200811Sibutramine, 15 mg/d, vs placebo for 24 weeksSibutramine was significantly better than placebo in reducing BE, weight, BMI, and related psychological features of BE
Brennan et al, 200812Open-label memantine, 5 to 20 mg/d, for 12 weeksMemantine was associated with decreased binge frequency and related psychological features of BE
Guerdjikova et al, 200913Lamotrigine, 50 to 400 mg/d, vs placebo for 16 weeksLamotrigine was not significantly different from placebo
BE: binge eating; BMI: body mass index
 

 

CBT vs other behavioral approaches

Cognitive-behavioral therapy (CBT), which focuses on identifying and modifying unhealthy thoughts that maintain disordered eating behaviors, is the most widely studied behavioral intervention for BED. Other studied treatments include interpersonal psychotherapy (IPT), motivational interviewing (MI), and structured behavioral weight loss (BWL) (Table 3).14-24 IPT is a psychodynamically based, time-limited treatment that focuses on the interpersonal context of the disorder and on building interpersonal skills. MI emphasizes exploring and resolving ambivalence about treatment, and works to facilitate change through motivational processes. BWL is centered on making dietary and physical activity changes to achieve weight loss. Behavioral treatments have been delivered in various formats, such as an individual or group setting, by electronic interface, and via self-help approaches. Most studies compared active treatment to a control group, but some compared active treatments head-to-head.

Table 3

CBT and other behavioral interventions for BED

StudyInterventionComments
Annunziato et al, 2009142 groups received CBT and hypocaloric diet for 8 weeks followed by 14 weeks of enhanced nutritional program (ie, reduced consumption of high energy density foods and once-daily liquid meal replacement) or control (normal diet)Enhanced nutritional program was not significantly different from the control in reducing weight, BE, or psychological features of BE; variability in adherence to the enhanced nutritional program was identified as a significant effect modifier
Ashton et al, 2009154 sessions of group CBT in an open trialCBT was associated with significant reductions in BE and psychological features of BE in post-bariatric surgery patients
Dingemans et al, 200716CBT vs wait-list controlCBT significantly better than the wait-list control in reducing BE and psychological features of BE, and in achieving abstinence from BE
Friederich et al, 20071715-session CBT blended with elements of interpersonal therapy (IPT), nutritional counseling, and supervised walking program; no control groupTreatment significantly reduced weight, BE, and related psychological features of BE in patients meeting sub-threshold and full criteria for BED
Grilo et al, 200518Guided self-help CBT (CBTgsh) vs guided self-help behavioral weight loss (BWLgsh) vs non-specific attention control for 12 weeksCBTgsh significantly better than BWLgsh and control in BE remission; CBTgsh significantly better than BWLgsh, which was significantly better than control in reducing cognitive restraint; CBTgsh significantly better than control in reducing depression and eating-related psychopathology; no differences between groups in BMI change
Ricca et al, 201019Individual (I-CBT) vs group CBT (G-CBT) for 24 weeks in patients meeting subthreshold and full criteria for BEDBE and BMI were significantly reduced in both groups at 24 weeks and 3-year follow-up. I-CBT was not better than G-CBT in reducing BE or weight at 24 weeks or 3-year follow-up; I-CBT was significantly better than G-CBT in reducing eating-related psychopathology at 24 weeks and 3-year follow-up; I-CBT was significantly better than G-CBT in recovery (ie, no longer meeting full BED criteria) at 24 weeks but not at 3-year follow-up
Schlup et al, 2009208 weekly sessions of group CBT vs wait-list controlCBT was significantly better than wait-list control in reducing BE and eating concerns and in achieving abstinence at end of treatment; CBT was not different than control in reducing BMI; treatment-related reductions in BE and eating concerns were maintained at 12-month follow-up
Shapiro et al, 20072110 weekly sessions of group CBT (G-CBT) vs CD-ROM delivered CBT (CD-CBT) vs wait-list controlG-CBT and CD-CBT were not different from each other but both were significantly better than wait-list control in reducing BE
Tasca et al, 200622Group CBT (G-CBT) vs group psychodynamic interpersonal therapy (G-IPT) vs wait-list control for 16 weeksG-CBT and G-IPT were not different from each other; G-CBT and G-IPT were significantly better than wait list in reducing BE and interpersonal problems (but not BMI) and increasing cognitive restraint post-treatment; depression was reduced in both groups at 6 months but only in G-IPT at 12 months; reductions in BE maintained at 12 months
Wilson et al, 20102310 sessions of guided self-help CBT (CBTgsh) vs 19 sessions of IPT vs 20 sessions of behavioral weight loss (BWL) over 6 monthsBWL was significantly better than IPT and CBTgsh in reducing BMI and in the number of patients achieving 5% weight loss at post-treatment but effects were not sustained over time; BWL was significantly better than CBTgsh in increasing dietary restraint
Cassin et al, 200824Self-help book + motivational interviewing (SH-MI) vs self-help book alone (SH) for 16 weeksSH-MI was significantly better than SH in reducing BE and depression
BE: binge eating; BED: binge eating disorder; BMI: body mass index; CBT: cognitive-behavioral therapy

Studies found that CBT and IPT are effective in reducing the frequency of binge eating, whether measured by the number of binge eating episodes or days a patient reports having engaged in binge eating.14-23 However, some studies suggested that CBT can help a substantial number of patients achieve abstinence from binge eating.16,20 Adding MI to a self-help approach may improve binge eating outcomes,24 and binge eating can be successfully reduced using individual, group, and CD-ROM delivery formats.21 In direct comparisons, individual CBT outperformed group CBT in helping patients recover from BED (ie, no longer meeting diagnostic criteria),19 and CBT delivered via guided self-help outperformed BWL in helping patients achieve remission.18

 

 

Psychological features of BED typically include low levels of cognitive restraint and high levels of disinhibition, hunger, and shape and weight concerns. Improvements in these psychological measures were observed with CBT,15-20,22 IPT,22 and MI.24 In direct comparisons, self-help CBT demonstrated greater reductions in perceived hunger and disinhibition than self-help BWL,18 and individual CBT outperformed group CBT in reducing shape and weight concerns.19 Isolated studies reported improvements in depression after self-help CBT18 and MI,24 and sustained improvements22 after group CBT (6 months) and group IPT (12 months). Additional research is needed to determine whether CBT crafted specifically for BED improves self-rated depression or if enhancements targeting depressive symptoms are required.

The impact of behavioral interventions on weight in overweight patients has been mixed. Although some CBT studies reported a substantial decrease in weight,17,19 others suggested that weight loss among patients treated with CBT is not superior to those in a wait-list control group16 or is not significant over the course of treatment.20,21 The impact of BWL on weight outcomes in BED also has been unimpressive: after 12 weeks, self-help BWL was no better than self-help CBT in reducing BMI18; after 16 weeks, BWL was better than CBT and IPT in achieving clinically significant (≥5%) weight loss, but this advantage was not sustained at 1- and 2-year follow-up.23 It is difficult to determine why successfully treated BED patients fail to lose weight because one would expect decreases in binge eating to lead to weight loss. It is possible that calories previously consumed during binge eating episodes are distributed over non-binge meals or that patients label binges and non-binge meals differently as a result of treatment.

Combining treatments

BED patients often are treated with a combination of psychotherapy and pharmacotherapy (Table 4).25-29 When added to CBT, topiramate was associated with improvements in weight and some psychological outcomes,25,26 but fluoxetine was not.27,28 Direct comparisons also showed that CBT, alone or combined with fluoxetine, was better than fluoxetine alone in reducing binge eating.27 When combined with an individualized hypocaloric diet, the anti-obesity medication orlistat reduced weight in obese BED patients but had no appreciable effect on binge eating.29 Collectively, the studies we reviewed suggested that combining medication and CBT may improve binge eating and weight loss outcomes; however, additional trials are necessary to determine more definitively which medications combined with CBT are best at producing sustained weight loss while reducing binge eating frequency.

Table 4

Combining medication with behavioral interventions for BED

StudyDrug/dosageComments
Brambilla et al, 2009253 groups treated for 6 months:
Group 1: CBT + setraline (50 to 150 mg/d) + topiramate (25 to 150 mg/d) + reduced calorie diet
Group 2: CBT + sertraline (50 to 150 mg/d) + reduced calorie diet
Group 3: CBT + nutritional counseling
Weight, BMI, and psychological features of BE reduced significantly only in group 1
Claudino et al, 200726Group 1: CBT + topiramate (25 to 300 mg/d)
Group 2: CBT + placebo
19 sessions over 21 weeks
Significant reductions in BE and depression in both groups; topiramate significantly better than placebo in reducing weight and in achieving BE remission
Devlin et al, 2005274 groups, all received behavioral weight control intervention for 5 months (20 weeks) plus either:
Group 1: CBT + fluoxetine
Group 2: CBT + placebo
Group 3: fluoxetineGroup 4 placebo
(fluoxetine dose, 20 to 60 mg/d)
CBT groups (1 and 2) significantly better than non-CBT groups (3 and 4) in reducing BE and achieving abstinence from BE; fluoxetine significantly better than placebo in reducing depression
Molinari et al, 2005283 groups, all received nutritional and diet counseling for 54 weeks (4 were inpatient) plus:
Group 1: CBT
Group 2: fluoxetine (20 to 60 mg/d)
Group 3: CBT + fluoxetine
At 12 months, CBT (groups 1 and 3) associated with lower BE frequency and greater percentage of weight loss than fluoxetine
Golay et al, 200529Hypocaloric diet + orlistat (120 mg/d) vs hypocaloric diet + placebo for 24 weeksOrlistat not different from placebo in reducing the number of patients classified with BED; orlistat significantly better than placebo in reducing weight and body fat
BE: binge eating; BED: binge eating disorder; BMI: body mass index; CBT: cognitive-behavioral therapy

Recommendations

Evidence suggests that pharmacotherapy and CBT—alone or in combination—are effective in reducing binge eating, and pharmacotherapy is effective in reducing weight in overweight individuals with BED. More research is needed for IPT and MI. It is unclear which medications provide the greatest benefit in terms of binge eating remission; however, pharmacotherapy has a clear advantage in facilitating short-term weight loss. Also, all BED patients should receive medical management to address possible complications such as hypertension or type 2 diabetes. In addition to reducing binge eating, CBT can improve related psychological comorbidities (eg, eating-related psychopathology and depression) and may have additional benefit when combined with pharmacotherapy.

 

 

In light of these findings, we recommend augmenting psychotherapeutic care with pharmacotherapy and medical management to address all relevant psychological and medical domains. Future investigations should address the benefits of coordinated psychological and medical care and evaluate how to maintain treatment gains.

Related Resources

  • Binge Eating Disorder Association. www.bedaonline.com.
  • Brownley KA, Berkman ND, Sedway JA, et al. Binge eating disorder treatment: a systematic review of randomized controlled trials. Int J Eat Disord. 2007;40(4):337-348.

Drug Brand Names

  • Atomoxetine • Strattera
  • Escitalopram • Lexapro
  • Fluoxetine • Prozac
  • Lamotrigine • Lamictal
  • Memantine • Namenda
  • Orlistat • Alli, Xenical
  • Sertraline • Zoloft
  • Sibutramine • Meridia
  • Topiramate • Topamax, Topiragen
  • Zonisamide • Zonegram

Disclosures

Dr. Peat receives a post-doctoral trainee grant from the National Institutes of Health.

Drs. Brownley, Berkman, and Bulik report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

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

2. Hudson JI, Hiripi E, Pope HG, et al. The prevalence and correlates of eating disorders in the National Comorbidity Survey Replication. Biol Psychiatry. 2007;61(3):348-358.

3. American Psychiatric Association. Proposed revision to DSM-5: K-05 Feeding and eating disorders. http://www.dsm5.org/ProposedRevision/Pages/proposedrevision.aspx?rid=372. Updated January 31 2011. Accessed March 26, 2012.

4. Grucza RA, Pryzbeck TR, Cloninger CR. Prevalence and correlates of binge eating disorder in a community sample. Compr Psychiatry. 2007;48(2):124-131.

5. Meneghini LF, Spadola J, Florez H. Prevalence and associations of binge eating disorder in a multiethnic population with type 2 diabetes. Diabetes Care. 2006;29(12):2760.-

6. Berkman ND, Bulik CM, Brownley KA, et al. Management of eating disorders. Evidence report/technology assessment No. 135. Rockville, MD: Agency for Healthcare Research and Quality; 2006. AHRQ Publication No. 06-E010.

7. Guerdjikova AI, McElroy SL, Kotwal R, et al. High-dose escitalopram in the treatment of binge-eating disorder with obesity: a placebo-controlled monotherapy trial. Hum Psychopharmacol. 2008;23(1):1-11.

8. McElroy SL, Kotwal R, Guerdjikova AL, et al. Zonisamide in the treatment of binge eating disorder with obesity: a randomized controlled trial. J Clin Psychiatry. 2006;67(12):1897-1906.

9. McElroy SL, Hudson JI, Capece JA, et al. Topiramate for the treatment of binge eating disorder associated with obesity: a placebo-controlled study. Biol Psychiatry. 2007;61(9):1039-1048.

10. McElroy SL, Guerdjikova A, Kotwal R, et al. Atomoxetine in the treatment of binge-eating disorder: a randomized placebo-controlled trial. J Clin Psychiatry. 2007;68(3):390-398.

11. Wilfley DE, Crow SJ, Hudson JI, et al. Efficacy of sibutramine for the treatment of binge eating disorder: a randomized multicenter placebo-controlled double-blind study. Am J Psychiatry. 2008;165(1):51-58.

12. Brennan BP, Roberts JL, Fogarty KV, et al. Memantine in the treatment of binge eating disorder: an open-label, prospective trial. Int J Eat Disord. 2008;41(6):520-526.

13. Guerdjikova AI, McElroy SL, Welge JA, et al. Lamotrigine in the treatment of binge-eating disorder with obesity: a randomized, placebo-controlled monotherapy trial. Int Clin Psychopharmacol. 2009;24(3):150-158.

14. Annunziato RA, Timko CA, Crerand CE, et al. A randomized trial examining differential meal replacement adherence in a weight loss maintenance program after one-year follow-up. Eat Behav. 2009;10(3):176-183.

15. Ashton K, Drerup M, Windover A, et al. Brief, four-session group CBT reduces binge eating behaviors among bariatric surgery candidates. Surg Obes Relat Dis. 2009;5(2):257-262.

16. Dingemans AE, Spinhoven P, van Furth EF. Predictors and mediators of treatment outcome in patients with binge eating disorder. Behav Res Ther. 2007;45(11):2551-2562.

17. Friederich HC, Schild S, Wild B, et al. Treatment outcome in people with subthreshold compared with full-syndrome binge eating disorder. Obesity. 2007;15(2):283-287.

18. Grilo CM, Masheb RM. A randomized controlled comparison of guided self-help cognitive behavioral therapy and behavioral weight loss for binge eating disorder. Behav Res Ther. 2005;43(11):1509-1525.

19. Ricca V, Castellini G, Mannucci E, et al. Comparison of individual and group cognitive behavioral therapy for binge eating disorder. A randomized, three-year follow-up study. Appetite. 2010;55(3):656-665.

20. Schlup B, Munsch S, Meyer AH, et al. The efficacy of a short version of a cognitive-behavioral treatment followed by booster sessions for binge eating disorder. Behav Res Ther. 2009;47(7):628-635.

21. Shapiro JR, Reba-Harrelson L, Dymek-Valentine M, et al. Feasibility and acceptability of CD-ROM-based cognitive-behavioral treatment for binge-eating disorder. Eur Eat Disord Rev. 2007;15(3):175-184.

22. Tasca GA, Ritchie K, Conrad G, et al. Attachment scales predict outcome in a randomized controlled trial of two group therapies for binge eating disorder: an aptitude by treatment interaction. Psychother Res. 2006;16(1):106-121.

23. Wilson GT, Wilfley DE, Agras WS, et al. Psychological treatments of binge eating disorder. Arch Gen Psychiatry. 2010;67(1):94-101.

24. Cassin SE, von Ranson KM, Heng K, et al. Adapted motivational interviewing for women with binge eating disorder: a randomized controlled trial. Psychol Addict Behav. 2008;22(3):417-425.

25. Brambilla F, Samek L, Company M, et al. Multivariate therapeutic approach to binge-eating disorder: combined nutritional, psychological and pharmacological treatment. Int Clin Psychopharmacol. 2009;24(6):312-317.

26. Claudino AM, de Oliveira IR, Appolinario JC, et al. Double-blind, randomized, placebo-controlled trial of topiramate plus cognitive-behavior therapy in binge-eating disorder. J Clin Psychiatry. 2007;68(9):1324-1332.

27. Devlin MJ, Goldfein JA, Petkova E, et al. Cognitive behavioral therapy and fluoxetine as adjuncts to group behavioral therapy for binge eating disorder. Obes Res. 2005;13(6):1077-1088.

28. Molinari E, Baruffi M, Croci M, et al. Binge eating disorder in obesity: comparison of different therapeutic strategies. Eat Weight Disord. 2005;10(3):154-161.

29. Golay A, Laurent-Jaccard A, Habicht F, et al. Effect of orlistat in obese patients with binge eating disorder. Obes Res. 2005;13(10):1701-1708.

Article PDF
Author and Disclosure Information

Christine M. Peat, PhD
Postdoctoral Fellow, Eating Disorders Program, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC
Kimberly A. Brownley, PhD
Assistant Professor, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC
Nancy D. Berkman, PhD
Senior Health Policy Research Analyst, Health Care Quality and Outcomes Program, RTI International, Research Triangle Park, NC
Cynthia M. Bulik, PhD, FAED
Distinguished Professor of Eating Disorders, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC

Audio / Podcast
Issue
Current Psychiatry - 11(05)
Publications
Topics
Page Number
32-39
Legacy Keywords
binge eating disorder; BED; treatments; pharmacotherapy; cognitive behavioral therapy; weight; treatments
Sections
Audio / Podcast
Audio / Podcast
Author and Disclosure Information

Christine M. Peat, PhD
Postdoctoral Fellow, Eating Disorders Program, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC
Kimberly A. Brownley, PhD
Assistant Professor, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC
Nancy D. Berkman, PhD
Senior Health Policy Research Analyst, Health Care Quality and Outcomes Program, RTI International, Research Triangle Park, NC
Cynthia M. Bulik, PhD, FAED
Distinguished Professor of Eating Disorders, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC

Author and Disclosure Information

Christine M. Peat, PhD
Postdoctoral Fellow, Eating Disorders Program, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC
Kimberly A. Brownley, PhD
Assistant Professor, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC
Nancy D. Berkman, PhD
Senior Health Policy Research Analyst, Health Care Quality and Outcomes Program, RTI International, Research Triangle Park, NC
Cynthia M. Bulik, PhD, FAED
Distinguished Professor of Eating Disorders, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC

Article PDF
Article PDF

Binge eating is consumption of an unusually large amount of food coupled with a feeling of loss of control over eating. Binge eating disorder (BED) is characterized by recurrent episodes of binge eating without inappropriate compensatory behaviors (eg, self-induced vomiting, misuse of laxatives, diuretics, or other agents, excessive exercise).1 It is the most common eating disorder in the United States, with a lifetime prevalence of approximately 3.5% in women and 2% in men.2 The diagnosis falls within the DSM-IV-TR category of eating disorders not otherwise specified,1 but clinicians often view it as a distinct clinical phenomenon. In DSM-IV-TR, an individual would meet criteria for BED if he or she engages in regular binge eating behavior in the absence of recurrent compensatory behaviors ≥2 days per week over 6 months.1 Proposed changes for DSM-5 recognize a distinct BED diagnosis, reduce the frequency criterion to once per week and the duration criterion to the past 3 months, and shift the focus from binge days to binge episodes (Table 1).3

Table 1

Proposed DSM-5 criteria for binge eating disorder

  1. Recurrent episodes of binge eating. An episode of binge eating is characterized by both of the following:
  2. The binge-eating episodes are associated with ≥3 of the following:
  3. Marked distress regarding binge eating is present
  4. The binge eating occurs, on average, at least once a week for 3 months
  5. The binge eating is not associated with the recurrent use of inappropriate compensatory behavior (for example, purging) and does not occur exclusively during the course anorexia nervosa, bulimia nervosa, or avoidant/restrictive food intake disorder
Source:Reference 3

BED can occur in individuals of all body mass indices (BMI), but is common among individuals who are overweight or obese as well as those with depression or type 2 diabetes; BED can complicate treatment of these conditions.2,4,5 Primary treatment goals are:

  • abstinence from binge eating
  • improved psychological functioning
  • appropriate weight regulation in overweight patients.

We report on 3 approaches to BED treatment: medication only, behavioral intervention only, and medication plus behavioral intervention. This article provides insights about emerging changes in diagnostic criteria for BED as well as evidence-informed treatment options and recommendations.

The evidence base

We conducted a review of 23 BED studies: 7 medication only, 5 medication plus behavioral, and 11 behavioral only. We focused on studies conducted since September 2005 that included binge frequency, weight, and depression as primary outcomes (see Berkman et al6 for a review of BED treatment studies before 2005). The studies included 2,527 participants (2,216 women and 311 men). Although the sex distribution of BED in the general population tends to slightly favor women,2 the proportion of women presenting for treatment generally is considerably higher than that of men. In studies that reported on race and/or ethnicity, 1,639 participants were identified as white, 191 as African American, 25 as Hispanic, 2 as Asian, 1 as Native American, and 25 as “other.” Ages ranged from 18 to 77.

Several medications are effective

In placebo-controlled studies, a high-dose selective serotonin reuptake inhibitor (escitalopram7), 2 anticonvulsants (zonisamide8 and topiramate9), a selective norepinephrine reuptake inhibitor (atomoxetine10), and an appetite suppressant (sibutramine11) were associated with significant decreases in binge eating frequency, weight, and BMI in overweight/obese patients diagnosed with BED (Table 2). In an open-label trial, memantine—a N-methyl-D-aspartate receptor antagonist often used to treat symptoms of Alzheimer’s disease—was associated with a significant reduction in binge eating but no change in weight.12 Lamotrigine was not significantly different from placebo in reducing binge eating or weight, but showed promise in reducing metabolic parameters such as glucose and triglyceride levels commonly associated with obesity and type 2 diabetes.13 Because BED often is comorbid with obesity and type 2 diabetes, lamotrigine augmentation when treating obese individuals with BED warrants further investigation. As with any pharmacologic agent, carefully consider potential side effects and interactions with other drugs before prescribing medications for BED. Informing patients of potential side effects is crucial for patient safety and accuracy of the data collected in well-controlled treatment studies.

Table 2

Pharmacotherapy for binge eating disorder

StudyDrug/dosageComments
Guerdjikova et al, 20087Escitalopram, 10 to 30 mg/d, vs placebo for 12 weeksEscitalopram was significantly better than placebo in reducing weight, BMI, and illness severity
McElroy et al, 20068Zonisamide, 100 to 600 mg/d, vs placebo for 16 weeksZonisamide was significantly better than placebo in reducing BE, weight, BMI, and various aspects of unhealthy eating behavior
McElroy et al, 20079Topiramate, 25 to 400 mg/d, vs placebo for 16 weeksTopiramate was significantly better than placebo in reducing BE, weight, BMI, and related psychological features of BE
McElroy et al, 200710Atomoxetine, 40 to 120 mg/d, vs placebo for 10 weeksAtomoxetine was significantly better than placebo in reducing BE, weight, BMI, and obsessive-compulsive features of BE, and in achieving remission
Wilfley et al, 200811Sibutramine, 15 mg/d, vs placebo for 24 weeksSibutramine was significantly better than placebo in reducing BE, weight, BMI, and related psychological features of BE
Brennan et al, 200812Open-label memantine, 5 to 20 mg/d, for 12 weeksMemantine was associated with decreased binge frequency and related psychological features of BE
Guerdjikova et al, 200913Lamotrigine, 50 to 400 mg/d, vs placebo for 16 weeksLamotrigine was not significantly different from placebo
BE: binge eating; BMI: body mass index
 

 

CBT vs other behavioral approaches

Cognitive-behavioral therapy (CBT), which focuses on identifying and modifying unhealthy thoughts that maintain disordered eating behaviors, is the most widely studied behavioral intervention for BED. Other studied treatments include interpersonal psychotherapy (IPT), motivational interviewing (MI), and structured behavioral weight loss (BWL) (Table 3).14-24 IPT is a psychodynamically based, time-limited treatment that focuses on the interpersonal context of the disorder and on building interpersonal skills. MI emphasizes exploring and resolving ambivalence about treatment, and works to facilitate change through motivational processes. BWL is centered on making dietary and physical activity changes to achieve weight loss. Behavioral treatments have been delivered in various formats, such as an individual or group setting, by electronic interface, and via self-help approaches. Most studies compared active treatment to a control group, but some compared active treatments head-to-head.

Table 3

CBT and other behavioral interventions for BED

StudyInterventionComments
Annunziato et al, 2009142 groups received CBT and hypocaloric diet for 8 weeks followed by 14 weeks of enhanced nutritional program (ie, reduced consumption of high energy density foods and once-daily liquid meal replacement) or control (normal diet)Enhanced nutritional program was not significantly different from the control in reducing weight, BE, or psychological features of BE; variability in adherence to the enhanced nutritional program was identified as a significant effect modifier
Ashton et al, 2009154 sessions of group CBT in an open trialCBT was associated with significant reductions in BE and psychological features of BE in post-bariatric surgery patients
Dingemans et al, 200716CBT vs wait-list controlCBT significantly better than the wait-list control in reducing BE and psychological features of BE, and in achieving abstinence from BE
Friederich et al, 20071715-session CBT blended with elements of interpersonal therapy (IPT), nutritional counseling, and supervised walking program; no control groupTreatment significantly reduced weight, BE, and related psychological features of BE in patients meeting sub-threshold and full criteria for BED
Grilo et al, 200518Guided self-help CBT (CBTgsh) vs guided self-help behavioral weight loss (BWLgsh) vs non-specific attention control for 12 weeksCBTgsh significantly better than BWLgsh and control in BE remission; CBTgsh significantly better than BWLgsh, which was significantly better than control in reducing cognitive restraint; CBTgsh significantly better than control in reducing depression and eating-related psychopathology; no differences between groups in BMI change
Ricca et al, 201019Individual (I-CBT) vs group CBT (G-CBT) for 24 weeks in patients meeting subthreshold and full criteria for BEDBE and BMI were significantly reduced in both groups at 24 weeks and 3-year follow-up. I-CBT was not better than G-CBT in reducing BE or weight at 24 weeks or 3-year follow-up; I-CBT was significantly better than G-CBT in reducing eating-related psychopathology at 24 weeks and 3-year follow-up; I-CBT was significantly better than G-CBT in recovery (ie, no longer meeting full BED criteria) at 24 weeks but not at 3-year follow-up
Schlup et al, 2009208 weekly sessions of group CBT vs wait-list controlCBT was significantly better than wait-list control in reducing BE and eating concerns and in achieving abstinence at end of treatment; CBT was not different than control in reducing BMI; treatment-related reductions in BE and eating concerns were maintained at 12-month follow-up
Shapiro et al, 20072110 weekly sessions of group CBT (G-CBT) vs CD-ROM delivered CBT (CD-CBT) vs wait-list controlG-CBT and CD-CBT were not different from each other but both were significantly better than wait-list control in reducing BE
Tasca et al, 200622Group CBT (G-CBT) vs group psychodynamic interpersonal therapy (G-IPT) vs wait-list control for 16 weeksG-CBT and G-IPT were not different from each other; G-CBT and G-IPT were significantly better than wait list in reducing BE and interpersonal problems (but not BMI) and increasing cognitive restraint post-treatment; depression was reduced in both groups at 6 months but only in G-IPT at 12 months; reductions in BE maintained at 12 months
Wilson et al, 20102310 sessions of guided self-help CBT (CBTgsh) vs 19 sessions of IPT vs 20 sessions of behavioral weight loss (BWL) over 6 monthsBWL was significantly better than IPT and CBTgsh in reducing BMI and in the number of patients achieving 5% weight loss at post-treatment but effects were not sustained over time; BWL was significantly better than CBTgsh in increasing dietary restraint
Cassin et al, 200824Self-help book + motivational interviewing (SH-MI) vs self-help book alone (SH) for 16 weeksSH-MI was significantly better than SH in reducing BE and depression
BE: binge eating; BED: binge eating disorder; BMI: body mass index; CBT: cognitive-behavioral therapy

Studies found that CBT and IPT are effective in reducing the frequency of binge eating, whether measured by the number of binge eating episodes or days a patient reports having engaged in binge eating.14-23 However, some studies suggested that CBT can help a substantial number of patients achieve abstinence from binge eating.16,20 Adding MI to a self-help approach may improve binge eating outcomes,24 and binge eating can be successfully reduced using individual, group, and CD-ROM delivery formats.21 In direct comparisons, individual CBT outperformed group CBT in helping patients recover from BED (ie, no longer meeting diagnostic criteria),19 and CBT delivered via guided self-help outperformed BWL in helping patients achieve remission.18

 

 

Psychological features of BED typically include low levels of cognitive restraint and high levels of disinhibition, hunger, and shape and weight concerns. Improvements in these psychological measures were observed with CBT,15-20,22 IPT,22 and MI.24 In direct comparisons, self-help CBT demonstrated greater reductions in perceived hunger and disinhibition than self-help BWL,18 and individual CBT outperformed group CBT in reducing shape and weight concerns.19 Isolated studies reported improvements in depression after self-help CBT18 and MI,24 and sustained improvements22 after group CBT (6 months) and group IPT (12 months). Additional research is needed to determine whether CBT crafted specifically for BED improves self-rated depression or if enhancements targeting depressive symptoms are required.

The impact of behavioral interventions on weight in overweight patients has been mixed. Although some CBT studies reported a substantial decrease in weight,17,19 others suggested that weight loss among patients treated with CBT is not superior to those in a wait-list control group16 or is not significant over the course of treatment.20,21 The impact of BWL on weight outcomes in BED also has been unimpressive: after 12 weeks, self-help BWL was no better than self-help CBT in reducing BMI18; after 16 weeks, BWL was better than CBT and IPT in achieving clinically significant (≥5%) weight loss, but this advantage was not sustained at 1- and 2-year follow-up.23 It is difficult to determine why successfully treated BED patients fail to lose weight because one would expect decreases in binge eating to lead to weight loss. It is possible that calories previously consumed during binge eating episodes are distributed over non-binge meals or that patients label binges and non-binge meals differently as a result of treatment.

Combining treatments

BED patients often are treated with a combination of psychotherapy and pharmacotherapy (Table 4).25-29 When added to CBT, topiramate was associated with improvements in weight and some psychological outcomes,25,26 but fluoxetine was not.27,28 Direct comparisons also showed that CBT, alone or combined with fluoxetine, was better than fluoxetine alone in reducing binge eating.27 When combined with an individualized hypocaloric diet, the anti-obesity medication orlistat reduced weight in obese BED patients but had no appreciable effect on binge eating.29 Collectively, the studies we reviewed suggested that combining medication and CBT may improve binge eating and weight loss outcomes; however, additional trials are necessary to determine more definitively which medications combined with CBT are best at producing sustained weight loss while reducing binge eating frequency.

Table 4

Combining medication with behavioral interventions for BED

StudyDrug/dosageComments
Brambilla et al, 2009253 groups treated for 6 months:
Group 1: CBT + setraline (50 to 150 mg/d) + topiramate (25 to 150 mg/d) + reduced calorie diet
Group 2: CBT + sertraline (50 to 150 mg/d) + reduced calorie diet
Group 3: CBT + nutritional counseling
Weight, BMI, and psychological features of BE reduced significantly only in group 1
Claudino et al, 200726Group 1: CBT + topiramate (25 to 300 mg/d)
Group 2: CBT + placebo
19 sessions over 21 weeks
Significant reductions in BE and depression in both groups; topiramate significantly better than placebo in reducing weight and in achieving BE remission
Devlin et al, 2005274 groups, all received behavioral weight control intervention for 5 months (20 weeks) plus either:
Group 1: CBT + fluoxetine
Group 2: CBT + placebo
Group 3: fluoxetineGroup 4 placebo
(fluoxetine dose, 20 to 60 mg/d)
CBT groups (1 and 2) significantly better than non-CBT groups (3 and 4) in reducing BE and achieving abstinence from BE; fluoxetine significantly better than placebo in reducing depression
Molinari et al, 2005283 groups, all received nutritional and diet counseling for 54 weeks (4 were inpatient) plus:
Group 1: CBT
Group 2: fluoxetine (20 to 60 mg/d)
Group 3: CBT + fluoxetine
At 12 months, CBT (groups 1 and 3) associated with lower BE frequency and greater percentage of weight loss than fluoxetine
Golay et al, 200529Hypocaloric diet + orlistat (120 mg/d) vs hypocaloric diet + placebo for 24 weeksOrlistat not different from placebo in reducing the number of patients classified with BED; orlistat significantly better than placebo in reducing weight and body fat
BE: binge eating; BED: binge eating disorder; BMI: body mass index; CBT: cognitive-behavioral therapy

Recommendations

Evidence suggests that pharmacotherapy and CBT—alone or in combination—are effective in reducing binge eating, and pharmacotherapy is effective in reducing weight in overweight individuals with BED. More research is needed for IPT and MI. It is unclear which medications provide the greatest benefit in terms of binge eating remission; however, pharmacotherapy has a clear advantage in facilitating short-term weight loss. Also, all BED patients should receive medical management to address possible complications such as hypertension or type 2 diabetes. In addition to reducing binge eating, CBT can improve related psychological comorbidities (eg, eating-related psychopathology and depression) and may have additional benefit when combined with pharmacotherapy.

 

 

In light of these findings, we recommend augmenting psychotherapeutic care with pharmacotherapy and medical management to address all relevant psychological and medical domains. Future investigations should address the benefits of coordinated psychological and medical care and evaluate how to maintain treatment gains.

Related Resources

  • Binge Eating Disorder Association. www.bedaonline.com.
  • Brownley KA, Berkman ND, Sedway JA, et al. Binge eating disorder treatment: a systematic review of randomized controlled trials. Int J Eat Disord. 2007;40(4):337-348.

Drug Brand Names

  • Atomoxetine • Strattera
  • Escitalopram • Lexapro
  • Fluoxetine • Prozac
  • Lamotrigine • Lamictal
  • Memantine • Namenda
  • Orlistat • Alli, Xenical
  • Sertraline • Zoloft
  • Sibutramine • Meridia
  • Topiramate • Topamax, Topiragen
  • Zonisamide • Zonegram

Disclosures

Dr. Peat receives a post-doctoral trainee grant from the National Institutes of Health.

Drs. Brownley, Berkman, and Bulik report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Binge eating is consumption of an unusually large amount of food coupled with a feeling of loss of control over eating. Binge eating disorder (BED) is characterized by recurrent episodes of binge eating without inappropriate compensatory behaviors (eg, self-induced vomiting, misuse of laxatives, diuretics, or other agents, excessive exercise).1 It is the most common eating disorder in the United States, with a lifetime prevalence of approximately 3.5% in women and 2% in men.2 The diagnosis falls within the DSM-IV-TR category of eating disorders not otherwise specified,1 but clinicians often view it as a distinct clinical phenomenon. In DSM-IV-TR, an individual would meet criteria for BED if he or she engages in regular binge eating behavior in the absence of recurrent compensatory behaviors ≥2 days per week over 6 months.1 Proposed changes for DSM-5 recognize a distinct BED diagnosis, reduce the frequency criterion to once per week and the duration criterion to the past 3 months, and shift the focus from binge days to binge episodes (Table 1).3

Table 1

Proposed DSM-5 criteria for binge eating disorder

  1. Recurrent episodes of binge eating. An episode of binge eating is characterized by both of the following:
  2. The binge-eating episodes are associated with ≥3 of the following:
  3. Marked distress regarding binge eating is present
  4. The binge eating occurs, on average, at least once a week for 3 months
  5. The binge eating is not associated with the recurrent use of inappropriate compensatory behavior (for example, purging) and does not occur exclusively during the course anorexia nervosa, bulimia nervosa, or avoidant/restrictive food intake disorder
Source:Reference 3

BED can occur in individuals of all body mass indices (BMI), but is common among individuals who are overweight or obese as well as those with depression or type 2 diabetes; BED can complicate treatment of these conditions.2,4,5 Primary treatment goals are:

  • abstinence from binge eating
  • improved psychological functioning
  • appropriate weight regulation in overweight patients.

We report on 3 approaches to BED treatment: medication only, behavioral intervention only, and medication plus behavioral intervention. This article provides insights about emerging changes in diagnostic criteria for BED as well as evidence-informed treatment options and recommendations.

The evidence base

We conducted a review of 23 BED studies: 7 medication only, 5 medication plus behavioral, and 11 behavioral only. We focused on studies conducted since September 2005 that included binge frequency, weight, and depression as primary outcomes (see Berkman et al6 for a review of BED treatment studies before 2005). The studies included 2,527 participants (2,216 women and 311 men). Although the sex distribution of BED in the general population tends to slightly favor women,2 the proportion of women presenting for treatment generally is considerably higher than that of men. In studies that reported on race and/or ethnicity, 1,639 participants were identified as white, 191 as African American, 25 as Hispanic, 2 as Asian, 1 as Native American, and 25 as “other.” Ages ranged from 18 to 77.

Several medications are effective

In placebo-controlled studies, a high-dose selective serotonin reuptake inhibitor (escitalopram7), 2 anticonvulsants (zonisamide8 and topiramate9), a selective norepinephrine reuptake inhibitor (atomoxetine10), and an appetite suppressant (sibutramine11) were associated with significant decreases in binge eating frequency, weight, and BMI in overweight/obese patients diagnosed with BED (Table 2). In an open-label trial, memantine—a N-methyl-D-aspartate receptor antagonist often used to treat symptoms of Alzheimer’s disease—was associated with a significant reduction in binge eating but no change in weight.12 Lamotrigine was not significantly different from placebo in reducing binge eating or weight, but showed promise in reducing metabolic parameters such as glucose and triglyceride levels commonly associated with obesity and type 2 diabetes.13 Because BED often is comorbid with obesity and type 2 diabetes, lamotrigine augmentation when treating obese individuals with BED warrants further investigation. As with any pharmacologic agent, carefully consider potential side effects and interactions with other drugs before prescribing medications for BED. Informing patients of potential side effects is crucial for patient safety and accuracy of the data collected in well-controlled treatment studies.

Table 2

Pharmacotherapy for binge eating disorder

StudyDrug/dosageComments
Guerdjikova et al, 20087Escitalopram, 10 to 30 mg/d, vs placebo for 12 weeksEscitalopram was significantly better than placebo in reducing weight, BMI, and illness severity
McElroy et al, 20068Zonisamide, 100 to 600 mg/d, vs placebo for 16 weeksZonisamide was significantly better than placebo in reducing BE, weight, BMI, and various aspects of unhealthy eating behavior
McElroy et al, 20079Topiramate, 25 to 400 mg/d, vs placebo for 16 weeksTopiramate was significantly better than placebo in reducing BE, weight, BMI, and related psychological features of BE
McElroy et al, 200710Atomoxetine, 40 to 120 mg/d, vs placebo for 10 weeksAtomoxetine was significantly better than placebo in reducing BE, weight, BMI, and obsessive-compulsive features of BE, and in achieving remission
Wilfley et al, 200811Sibutramine, 15 mg/d, vs placebo for 24 weeksSibutramine was significantly better than placebo in reducing BE, weight, BMI, and related psychological features of BE
Brennan et al, 200812Open-label memantine, 5 to 20 mg/d, for 12 weeksMemantine was associated with decreased binge frequency and related psychological features of BE
Guerdjikova et al, 200913Lamotrigine, 50 to 400 mg/d, vs placebo for 16 weeksLamotrigine was not significantly different from placebo
BE: binge eating; BMI: body mass index
 

 

CBT vs other behavioral approaches

Cognitive-behavioral therapy (CBT), which focuses on identifying and modifying unhealthy thoughts that maintain disordered eating behaviors, is the most widely studied behavioral intervention for BED. Other studied treatments include interpersonal psychotherapy (IPT), motivational interviewing (MI), and structured behavioral weight loss (BWL) (Table 3).14-24 IPT is a psychodynamically based, time-limited treatment that focuses on the interpersonal context of the disorder and on building interpersonal skills. MI emphasizes exploring and resolving ambivalence about treatment, and works to facilitate change through motivational processes. BWL is centered on making dietary and physical activity changes to achieve weight loss. Behavioral treatments have been delivered in various formats, such as an individual or group setting, by electronic interface, and via self-help approaches. Most studies compared active treatment to a control group, but some compared active treatments head-to-head.

Table 3

CBT and other behavioral interventions for BED

StudyInterventionComments
Annunziato et al, 2009142 groups received CBT and hypocaloric diet for 8 weeks followed by 14 weeks of enhanced nutritional program (ie, reduced consumption of high energy density foods and once-daily liquid meal replacement) or control (normal diet)Enhanced nutritional program was not significantly different from the control in reducing weight, BE, or psychological features of BE; variability in adherence to the enhanced nutritional program was identified as a significant effect modifier
Ashton et al, 2009154 sessions of group CBT in an open trialCBT was associated with significant reductions in BE and psychological features of BE in post-bariatric surgery patients
Dingemans et al, 200716CBT vs wait-list controlCBT significantly better than the wait-list control in reducing BE and psychological features of BE, and in achieving abstinence from BE
Friederich et al, 20071715-session CBT blended with elements of interpersonal therapy (IPT), nutritional counseling, and supervised walking program; no control groupTreatment significantly reduced weight, BE, and related psychological features of BE in patients meeting sub-threshold and full criteria for BED
Grilo et al, 200518Guided self-help CBT (CBTgsh) vs guided self-help behavioral weight loss (BWLgsh) vs non-specific attention control for 12 weeksCBTgsh significantly better than BWLgsh and control in BE remission; CBTgsh significantly better than BWLgsh, which was significantly better than control in reducing cognitive restraint; CBTgsh significantly better than control in reducing depression and eating-related psychopathology; no differences between groups in BMI change
Ricca et al, 201019Individual (I-CBT) vs group CBT (G-CBT) for 24 weeks in patients meeting subthreshold and full criteria for BEDBE and BMI were significantly reduced in both groups at 24 weeks and 3-year follow-up. I-CBT was not better than G-CBT in reducing BE or weight at 24 weeks or 3-year follow-up; I-CBT was significantly better than G-CBT in reducing eating-related psychopathology at 24 weeks and 3-year follow-up; I-CBT was significantly better than G-CBT in recovery (ie, no longer meeting full BED criteria) at 24 weeks but not at 3-year follow-up
Schlup et al, 2009208 weekly sessions of group CBT vs wait-list controlCBT was significantly better than wait-list control in reducing BE and eating concerns and in achieving abstinence at end of treatment; CBT was not different than control in reducing BMI; treatment-related reductions in BE and eating concerns were maintained at 12-month follow-up
Shapiro et al, 20072110 weekly sessions of group CBT (G-CBT) vs CD-ROM delivered CBT (CD-CBT) vs wait-list controlG-CBT and CD-CBT were not different from each other but both were significantly better than wait-list control in reducing BE
Tasca et al, 200622Group CBT (G-CBT) vs group psychodynamic interpersonal therapy (G-IPT) vs wait-list control for 16 weeksG-CBT and G-IPT were not different from each other; G-CBT and G-IPT were significantly better than wait list in reducing BE and interpersonal problems (but not BMI) and increasing cognitive restraint post-treatment; depression was reduced in both groups at 6 months but only in G-IPT at 12 months; reductions in BE maintained at 12 months
Wilson et al, 20102310 sessions of guided self-help CBT (CBTgsh) vs 19 sessions of IPT vs 20 sessions of behavioral weight loss (BWL) over 6 monthsBWL was significantly better than IPT and CBTgsh in reducing BMI and in the number of patients achieving 5% weight loss at post-treatment but effects were not sustained over time; BWL was significantly better than CBTgsh in increasing dietary restraint
Cassin et al, 200824Self-help book + motivational interviewing (SH-MI) vs self-help book alone (SH) for 16 weeksSH-MI was significantly better than SH in reducing BE and depression
BE: binge eating; BED: binge eating disorder; BMI: body mass index; CBT: cognitive-behavioral therapy

Studies found that CBT and IPT are effective in reducing the frequency of binge eating, whether measured by the number of binge eating episodes or days a patient reports having engaged in binge eating.14-23 However, some studies suggested that CBT can help a substantial number of patients achieve abstinence from binge eating.16,20 Adding MI to a self-help approach may improve binge eating outcomes,24 and binge eating can be successfully reduced using individual, group, and CD-ROM delivery formats.21 In direct comparisons, individual CBT outperformed group CBT in helping patients recover from BED (ie, no longer meeting diagnostic criteria),19 and CBT delivered via guided self-help outperformed BWL in helping patients achieve remission.18

 

 

Psychological features of BED typically include low levels of cognitive restraint and high levels of disinhibition, hunger, and shape and weight concerns. Improvements in these psychological measures were observed with CBT,15-20,22 IPT,22 and MI.24 In direct comparisons, self-help CBT demonstrated greater reductions in perceived hunger and disinhibition than self-help BWL,18 and individual CBT outperformed group CBT in reducing shape and weight concerns.19 Isolated studies reported improvements in depression after self-help CBT18 and MI,24 and sustained improvements22 after group CBT (6 months) and group IPT (12 months). Additional research is needed to determine whether CBT crafted specifically for BED improves self-rated depression or if enhancements targeting depressive symptoms are required.

The impact of behavioral interventions on weight in overweight patients has been mixed. Although some CBT studies reported a substantial decrease in weight,17,19 others suggested that weight loss among patients treated with CBT is not superior to those in a wait-list control group16 or is not significant over the course of treatment.20,21 The impact of BWL on weight outcomes in BED also has been unimpressive: after 12 weeks, self-help BWL was no better than self-help CBT in reducing BMI18; after 16 weeks, BWL was better than CBT and IPT in achieving clinically significant (≥5%) weight loss, but this advantage was not sustained at 1- and 2-year follow-up.23 It is difficult to determine why successfully treated BED patients fail to lose weight because one would expect decreases in binge eating to lead to weight loss. It is possible that calories previously consumed during binge eating episodes are distributed over non-binge meals or that patients label binges and non-binge meals differently as a result of treatment.

Combining treatments

BED patients often are treated with a combination of psychotherapy and pharmacotherapy (Table 4).25-29 When added to CBT, topiramate was associated with improvements in weight and some psychological outcomes,25,26 but fluoxetine was not.27,28 Direct comparisons also showed that CBT, alone or combined with fluoxetine, was better than fluoxetine alone in reducing binge eating.27 When combined with an individualized hypocaloric diet, the anti-obesity medication orlistat reduced weight in obese BED patients but had no appreciable effect on binge eating.29 Collectively, the studies we reviewed suggested that combining medication and CBT may improve binge eating and weight loss outcomes; however, additional trials are necessary to determine more definitively which medications combined with CBT are best at producing sustained weight loss while reducing binge eating frequency.

Table 4

Combining medication with behavioral interventions for BED

StudyDrug/dosageComments
Brambilla et al, 2009253 groups treated for 6 months:
Group 1: CBT + setraline (50 to 150 mg/d) + topiramate (25 to 150 mg/d) + reduced calorie diet
Group 2: CBT + sertraline (50 to 150 mg/d) + reduced calorie diet
Group 3: CBT + nutritional counseling
Weight, BMI, and psychological features of BE reduced significantly only in group 1
Claudino et al, 200726Group 1: CBT + topiramate (25 to 300 mg/d)
Group 2: CBT + placebo
19 sessions over 21 weeks
Significant reductions in BE and depression in both groups; topiramate significantly better than placebo in reducing weight and in achieving BE remission
Devlin et al, 2005274 groups, all received behavioral weight control intervention for 5 months (20 weeks) plus either:
Group 1: CBT + fluoxetine
Group 2: CBT + placebo
Group 3: fluoxetineGroup 4 placebo
(fluoxetine dose, 20 to 60 mg/d)
CBT groups (1 and 2) significantly better than non-CBT groups (3 and 4) in reducing BE and achieving abstinence from BE; fluoxetine significantly better than placebo in reducing depression
Molinari et al, 2005283 groups, all received nutritional and diet counseling for 54 weeks (4 were inpatient) plus:
Group 1: CBT
Group 2: fluoxetine (20 to 60 mg/d)
Group 3: CBT + fluoxetine
At 12 months, CBT (groups 1 and 3) associated with lower BE frequency and greater percentage of weight loss than fluoxetine
Golay et al, 200529Hypocaloric diet + orlistat (120 mg/d) vs hypocaloric diet + placebo for 24 weeksOrlistat not different from placebo in reducing the number of patients classified with BED; orlistat significantly better than placebo in reducing weight and body fat
BE: binge eating; BED: binge eating disorder; BMI: body mass index; CBT: cognitive-behavioral therapy

Recommendations

Evidence suggests that pharmacotherapy and CBT—alone or in combination—are effective in reducing binge eating, and pharmacotherapy is effective in reducing weight in overweight individuals with BED. More research is needed for IPT and MI. It is unclear which medications provide the greatest benefit in terms of binge eating remission; however, pharmacotherapy has a clear advantage in facilitating short-term weight loss. Also, all BED patients should receive medical management to address possible complications such as hypertension or type 2 diabetes. In addition to reducing binge eating, CBT can improve related psychological comorbidities (eg, eating-related psychopathology and depression) and may have additional benefit when combined with pharmacotherapy.

 

 

In light of these findings, we recommend augmenting psychotherapeutic care with pharmacotherapy and medical management to address all relevant psychological and medical domains. Future investigations should address the benefits of coordinated psychological and medical care and evaluate how to maintain treatment gains.

Related Resources

  • Binge Eating Disorder Association. www.bedaonline.com.
  • Brownley KA, Berkman ND, Sedway JA, et al. Binge eating disorder treatment: a systematic review of randomized controlled trials. Int J Eat Disord. 2007;40(4):337-348.

Drug Brand Names

  • Atomoxetine • Strattera
  • Escitalopram • Lexapro
  • Fluoxetine • Prozac
  • Lamotrigine • Lamictal
  • Memantine • Namenda
  • Orlistat • Alli, Xenical
  • Sertraline • Zoloft
  • Sibutramine • Meridia
  • Topiramate • Topamax, Topiragen
  • Zonisamide • Zonegram

Disclosures

Dr. Peat receives a post-doctoral trainee grant from the National Institutes of Health.

Drs. Brownley, Berkman, and Bulik report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

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

2. Hudson JI, Hiripi E, Pope HG, et al. The prevalence and correlates of eating disorders in the National Comorbidity Survey Replication. Biol Psychiatry. 2007;61(3):348-358.

3. American Psychiatric Association. Proposed revision to DSM-5: K-05 Feeding and eating disorders. http://www.dsm5.org/ProposedRevision/Pages/proposedrevision.aspx?rid=372. Updated January 31 2011. Accessed March 26, 2012.

4. Grucza RA, Pryzbeck TR, Cloninger CR. Prevalence and correlates of binge eating disorder in a community sample. Compr Psychiatry. 2007;48(2):124-131.

5. Meneghini LF, Spadola J, Florez H. Prevalence and associations of binge eating disorder in a multiethnic population with type 2 diabetes. Diabetes Care. 2006;29(12):2760.-

6. Berkman ND, Bulik CM, Brownley KA, et al. Management of eating disorders. Evidence report/technology assessment No. 135. Rockville, MD: Agency for Healthcare Research and Quality; 2006. AHRQ Publication No. 06-E010.

7. Guerdjikova AI, McElroy SL, Kotwal R, et al. High-dose escitalopram in the treatment of binge-eating disorder with obesity: a placebo-controlled monotherapy trial. Hum Psychopharmacol. 2008;23(1):1-11.

8. McElroy SL, Kotwal R, Guerdjikova AL, et al. Zonisamide in the treatment of binge eating disorder with obesity: a randomized controlled trial. J Clin Psychiatry. 2006;67(12):1897-1906.

9. McElroy SL, Hudson JI, Capece JA, et al. Topiramate for the treatment of binge eating disorder associated with obesity: a placebo-controlled study. Biol Psychiatry. 2007;61(9):1039-1048.

10. McElroy SL, Guerdjikova A, Kotwal R, et al. Atomoxetine in the treatment of binge-eating disorder: a randomized placebo-controlled trial. J Clin Psychiatry. 2007;68(3):390-398.

11. Wilfley DE, Crow SJ, Hudson JI, et al. Efficacy of sibutramine for the treatment of binge eating disorder: a randomized multicenter placebo-controlled double-blind study. Am J Psychiatry. 2008;165(1):51-58.

12. Brennan BP, Roberts JL, Fogarty KV, et al. Memantine in the treatment of binge eating disorder: an open-label, prospective trial. Int J Eat Disord. 2008;41(6):520-526.

13. Guerdjikova AI, McElroy SL, Welge JA, et al. Lamotrigine in the treatment of binge-eating disorder with obesity: a randomized, placebo-controlled monotherapy trial. Int Clin Psychopharmacol. 2009;24(3):150-158.

14. Annunziato RA, Timko CA, Crerand CE, et al. A randomized trial examining differential meal replacement adherence in a weight loss maintenance program after one-year follow-up. Eat Behav. 2009;10(3):176-183.

15. Ashton K, Drerup M, Windover A, et al. Brief, four-session group CBT reduces binge eating behaviors among bariatric surgery candidates. Surg Obes Relat Dis. 2009;5(2):257-262.

16. Dingemans AE, Spinhoven P, van Furth EF. Predictors and mediators of treatment outcome in patients with binge eating disorder. Behav Res Ther. 2007;45(11):2551-2562.

17. Friederich HC, Schild S, Wild B, et al. Treatment outcome in people with subthreshold compared with full-syndrome binge eating disorder. Obesity. 2007;15(2):283-287.

18. Grilo CM, Masheb RM. A randomized controlled comparison of guided self-help cognitive behavioral therapy and behavioral weight loss for binge eating disorder. Behav Res Ther. 2005;43(11):1509-1525.

19. Ricca V, Castellini G, Mannucci E, et al. Comparison of individual and group cognitive behavioral therapy for binge eating disorder. A randomized, three-year follow-up study. Appetite. 2010;55(3):656-665.

20. Schlup B, Munsch S, Meyer AH, et al. The efficacy of a short version of a cognitive-behavioral treatment followed by booster sessions for binge eating disorder. Behav Res Ther. 2009;47(7):628-635.

21. Shapiro JR, Reba-Harrelson L, Dymek-Valentine M, et al. Feasibility and acceptability of CD-ROM-based cognitive-behavioral treatment for binge-eating disorder. Eur Eat Disord Rev. 2007;15(3):175-184.

22. Tasca GA, Ritchie K, Conrad G, et al. Attachment scales predict outcome in a randomized controlled trial of two group therapies for binge eating disorder: an aptitude by treatment interaction. Psychother Res. 2006;16(1):106-121.

23. Wilson GT, Wilfley DE, Agras WS, et al. Psychological treatments of binge eating disorder. Arch Gen Psychiatry. 2010;67(1):94-101.

24. Cassin SE, von Ranson KM, Heng K, et al. Adapted motivational interviewing for women with binge eating disorder: a randomized controlled trial. Psychol Addict Behav. 2008;22(3):417-425.

25. Brambilla F, Samek L, Company M, et al. Multivariate therapeutic approach to binge-eating disorder: combined nutritional, psychological and pharmacological treatment. Int Clin Psychopharmacol. 2009;24(6):312-317.

26. Claudino AM, de Oliveira IR, Appolinario JC, et al. Double-blind, randomized, placebo-controlled trial of topiramate plus cognitive-behavior therapy in binge-eating disorder. J Clin Psychiatry. 2007;68(9):1324-1332.

27. Devlin MJ, Goldfein JA, Petkova E, et al. Cognitive behavioral therapy and fluoxetine as adjuncts to group behavioral therapy for binge eating disorder. Obes Res. 2005;13(6):1077-1088.

28. Molinari E, Baruffi M, Croci M, et al. Binge eating disorder in obesity: comparison of different therapeutic strategies. Eat Weight Disord. 2005;10(3):154-161.

29. Golay A, Laurent-Jaccard A, Habicht F, et al. Effect of orlistat in obese patients with binge eating disorder. Obes Res. 2005;13(10):1701-1708.

References

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

2. Hudson JI, Hiripi E, Pope HG, et al. The prevalence and correlates of eating disorders in the National Comorbidity Survey Replication. Biol Psychiatry. 2007;61(3):348-358.

3. American Psychiatric Association. Proposed revision to DSM-5: K-05 Feeding and eating disorders. http://www.dsm5.org/ProposedRevision/Pages/proposedrevision.aspx?rid=372. Updated January 31 2011. Accessed March 26, 2012.

4. Grucza RA, Pryzbeck TR, Cloninger CR. Prevalence and correlates of binge eating disorder in a community sample. Compr Psychiatry. 2007;48(2):124-131.

5. Meneghini LF, Spadola J, Florez H. Prevalence and associations of binge eating disorder in a multiethnic population with type 2 diabetes. Diabetes Care. 2006;29(12):2760.-

6. Berkman ND, Bulik CM, Brownley KA, et al. Management of eating disorders. Evidence report/technology assessment No. 135. Rockville, MD: Agency for Healthcare Research and Quality; 2006. AHRQ Publication No. 06-E010.

7. Guerdjikova AI, McElroy SL, Kotwal R, et al. High-dose escitalopram in the treatment of binge-eating disorder with obesity: a placebo-controlled monotherapy trial. Hum Psychopharmacol. 2008;23(1):1-11.

8. McElroy SL, Kotwal R, Guerdjikova AL, et al. Zonisamide in the treatment of binge eating disorder with obesity: a randomized controlled trial. J Clin Psychiatry. 2006;67(12):1897-1906.

9. McElroy SL, Hudson JI, Capece JA, et al. Topiramate for the treatment of binge eating disorder associated with obesity: a placebo-controlled study. Biol Psychiatry. 2007;61(9):1039-1048.

10. McElroy SL, Guerdjikova A, Kotwal R, et al. Atomoxetine in the treatment of binge-eating disorder: a randomized placebo-controlled trial. J Clin Psychiatry. 2007;68(3):390-398.

11. Wilfley DE, Crow SJ, Hudson JI, et al. Efficacy of sibutramine for the treatment of binge eating disorder: a randomized multicenter placebo-controlled double-blind study. Am J Psychiatry. 2008;165(1):51-58.

12. Brennan BP, Roberts JL, Fogarty KV, et al. Memantine in the treatment of binge eating disorder: an open-label, prospective trial. Int J Eat Disord. 2008;41(6):520-526.

13. Guerdjikova AI, McElroy SL, Welge JA, et al. Lamotrigine in the treatment of binge-eating disorder with obesity: a randomized, placebo-controlled monotherapy trial. Int Clin Psychopharmacol. 2009;24(3):150-158.

14. Annunziato RA, Timko CA, Crerand CE, et al. A randomized trial examining differential meal replacement adherence in a weight loss maintenance program after one-year follow-up. Eat Behav. 2009;10(3):176-183.

15. Ashton K, Drerup M, Windover A, et al. Brief, four-session group CBT reduces binge eating behaviors among bariatric surgery candidates. Surg Obes Relat Dis. 2009;5(2):257-262.

16. Dingemans AE, Spinhoven P, van Furth EF. Predictors and mediators of treatment outcome in patients with binge eating disorder. Behav Res Ther. 2007;45(11):2551-2562.

17. Friederich HC, Schild S, Wild B, et al. Treatment outcome in people with subthreshold compared with full-syndrome binge eating disorder. Obesity. 2007;15(2):283-287.

18. Grilo CM, Masheb RM. A randomized controlled comparison of guided self-help cognitive behavioral therapy and behavioral weight loss for binge eating disorder. Behav Res Ther. 2005;43(11):1509-1525.

19. Ricca V, Castellini G, Mannucci E, et al. Comparison of individual and group cognitive behavioral therapy for binge eating disorder. A randomized, three-year follow-up study. Appetite. 2010;55(3):656-665.

20. Schlup B, Munsch S, Meyer AH, et al. The efficacy of a short version of a cognitive-behavioral treatment followed by booster sessions for binge eating disorder. Behav Res Ther. 2009;47(7):628-635.

21. Shapiro JR, Reba-Harrelson L, Dymek-Valentine M, et al. Feasibility and acceptability of CD-ROM-based cognitive-behavioral treatment for binge-eating disorder. Eur Eat Disord Rev. 2007;15(3):175-184.

22. Tasca GA, Ritchie K, Conrad G, et al. Attachment scales predict outcome in a randomized controlled trial of two group therapies for binge eating disorder: an aptitude by treatment interaction. Psychother Res. 2006;16(1):106-121.

23. Wilson GT, Wilfley DE, Agras WS, et al. Psychological treatments of binge eating disorder. Arch Gen Psychiatry. 2010;67(1):94-101.

24. Cassin SE, von Ranson KM, Heng K, et al. Adapted motivational interviewing for women with binge eating disorder: a randomized controlled trial. Psychol Addict Behav. 2008;22(3):417-425.

25. Brambilla F, Samek L, Company M, et al. Multivariate therapeutic approach to binge-eating disorder: combined nutritional, psychological and pharmacological treatment. Int Clin Psychopharmacol. 2009;24(6):312-317.

26. Claudino AM, de Oliveira IR, Appolinario JC, et al. Double-blind, randomized, placebo-controlled trial of topiramate plus cognitive-behavior therapy in binge-eating disorder. J Clin Psychiatry. 2007;68(9):1324-1332.

27. Devlin MJ, Goldfein JA, Petkova E, et al. Cognitive behavioral therapy and fluoxetine as adjuncts to group behavioral therapy for binge eating disorder. Obes Res. 2005;13(6):1077-1088.

28. Molinari E, Baruffi M, Croci M, et al. Binge eating disorder in obesity: comparison of different therapeutic strategies. Eat Weight Disord. 2005;10(3):154-161.

29. Golay A, Laurent-Jaccard A, Habicht F, et al. Effect of orlistat in obese patients with binge eating disorder. Obes Res. 2005;13(10):1701-1708.

Issue
Current Psychiatry - 11(05)
Issue
Current Psychiatry - 11(05)
Page Number
32-39
Page Number
32-39
Publications
Publications
Topics
Article Type
Display Headline
Binge eating disorder: Evidence-based treatments
Display Headline
Binge eating disorder: Evidence-based treatments
Legacy Keywords
binge eating disorder; BED; treatments; pharmacotherapy; cognitive behavioral therapy; weight; treatments
Legacy Keywords
binge eating disorder; BED; treatments; pharmacotherapy; cognitive behavioral therapy; weight; treatments
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Generalized anxiety disorder: Helping patients overcome worry

Article Type
Changed
Tue, 12/11/2018 - 15:04
Display Headline
Generalized anxiety disorder: Helping patients overcome worry

Discuss this article at www.facebook.com/CurrentPsychiatry

Mrs. M, age 44, is a married mother of 2 who presents to the psychiatric clinic with increased anxiety that recently has become intolerable, stating “I can’t stop my head.” She has experienced anxiety for “as long as I can remember.” She was a shy, anxious child who worried about her parents’ health. Her anxiety worsened at college, where she first sought care. She was prescribed diazepam as needed. The next semester, she had a depressive episode, treated with imipramine, 75 mg/d, which she tolerated poorly.

Mrs. M has received episodic supportive therapy since college. She has been plagued by bouts of anxiety and worry, with insomnia, tension, and fatigue. She worries about financial, career, family, and safety issues and has a phobia of spiders. Her family and friends often comment about her excessive worry, and it has strained her marriage and career; she was passed over for a promotion in part because of her anxiousness. Mrs. M also has experienced several depressive episodes.

Mrs. M has sought medical care for various non-specific somatic complaints; all laboratory tests were normal. Approximately 10 years ago, Mrs. M’s primary care physician prescribed fluoxetine, 20 mg/d, but Mrs. M stopped taking it after a few days, stating she felt “more anxious and jittery.”

To meet DSM-IV-TR diagnostic criteria for generalized anxiety disorder (GAD), patients must experience anxiety and worry that they find difficult to control. The worry and anxiety occur more days than not for at least 6 months and cause clinically significant distress and impairment (Table 1).1 These diagnostic criteria are being reevaluated—the DSM-5 Anxiety Work Group has proposed renaming the condition generalized worry disorder, specifying that only 2 domains need to be impacted by worry, shortening the required time frame of impairment from 6 months to 3 months, and including at least 1 behavioral change spawned by excessive worry.2 Although provisional, these recommendations suggest DSM-5 will include changes to GAD when it is published in 2013.

Table 1

DSM-IV-TR diagnostic criteria for generalized anxiety disorder

  1. Excessive anxiety and worry (apprehensive expectation), occurring more days than not for at least 6 months, about a number of events or activities (such as work or school performance)
  2. The person finds it difficult to control the worry
  3. The anxiety and worry are associated with ≥3 of the following 6 symptoms (with at least some symptoms present for more days than not for the past 6 months):
    1. restlessness or feeling keyed up or on edge
    2. being easily fatigued
    3. difficulty concentrating or mind going blank
    4. irritability
    5. muscle tension
    6. sleep disturbance (difficulty falling or staying asleep, or restless, unsatisfying sleep)
  4. The focus of the anxiety and worry is not confined to features of other axis I disorders (eg, social phobia, OCD, PTSD, etc.)
  5. The anxiety, worry, or physical symptoms cause clinically significant distress or impairment in social, occupational, or other important areas of functioning
  6. The disturbance is not due to the direct physiological effects of a substance (eg, a drug of abuse, a medication) or a general medical condition (eg, hyperthyroidism), and does not occur exclusively during a mood disorder, a psychotic disorder, or a pervasive developmental disorder
OCD: obsessive-compulsive disorder; PTSD: posttraumatic stress disorder
Source: Reference 1

A common, chronic condition

In the United States, the lifetime prevalence of GAD is 5.7%.3 It is twice as common in women. Although GAD can occur at any age, 75% of patients develop it before age 47; the median age is 31.3,4 Patients who present with GAD later in life have a better prognosis.3,4

Approximately 90% of GAD patients will meet criteria for another axis I disorder.3 When GAD patients present for treatment, social phobia and panic disorder are the most common comorbid psychiatric disorders. The lifetime prevalence of a mood disorder among GAD patients is 62%, but as few as 6% of GAD patients will meet criteria for a mood disorder at presentation.3 The onset of GAD usually precedes depression.5,6

Patients with GAD often first seek treatment from their primary care provider.7 A useful screening tool is the GAD-7 (Table 2).8 This instrument has a specificity of 92% and sensitivity of 76% for GAD for patients who score ≥8.7 Although the GAD-7 cannot confirm a GAD diagnosis, it can prompt clinicians to conduct a more structured interview. Because higher scores correlate with more severe symptoms, the GAD-7 can be used to measure progress.

Table 2

Screening for generalized anxiety disorder: The GAD-7

Over the last 2 weeks, how often have you been bothered by any of the following problems?Not at allSeveral daysMore than half the daysNearly every day
1.Feeling nervous, anxious, or on edge0123
2.Not being able to stop or control worrying0123
3.Worrying too much about different things0123
4.Trouble relaxing0123
5.Being so restless that it is hard to sit still0123
6.Becoming easily annoyed or irritable0123
7.Feeling afraid as if something awful might happen0123
GAD: generalized anxiety disorder
Source: Reference 8
 

 

Differential diagnosis

GAD typically has a chronic course with fluctuating symptom severity over the patient’s lifespan. Assess patients who present with anxiety for medical conditions that mimic GAD. These include:

  • endocrine (hyperthyroidism), metabolic (electrolyte abnormalities), respiratory (asthma), neurologic (seizure disorder), or cardiac (arrhythmia) conditions
  • nutritional deficiencies, especially of B vitamins and folate
  • ingestion of substances or medications that may cause anxiety, such as caffeine or amphetamines.

A thorough history, medication review, and physical examination—as well as routine tests such as metabolic panel, complete blood count, thyroid function tests, urine drug screen, and electrocardiography—will capture most of these potential etiologies. In addition to ruling out medical causes, also assess for comorbid psychiatric conditions before reaching a diagnosis.

Evidence-based treatments

The treatment armamentarium for GAD includes psychotherapy and pharmacotherapy; complementary and alternative medicine (CAM) modalities may be useful adjunctive treatments. Which approach to use is determined by clinical judgment and the patient’s symptom severity and preferences. Combination therapy consisting of psychotherapy and medication often is appropriate.

Psychotherapy. Cognitive-behavioral therapy (CBT) is the preferred form of psychotherapy for GAD because it results in sustained improvements for patients with anxiety.5,9 Other modalities that may be effective include psychodynamic psychotherapy, mindfulness-based therapy, and interpersonal psychotherapy.10,11

Pharmacotherapy. As few as one-quarter of patients with GAD receive medications at appropriate dose and duration.12 Antidepressants are a first-line pharmacotherapy.5 Selective serotonin reuptake inhibitors (SSRIs) and serotonin-norepinephrine reuptake inhibitors are highly effective for treating GAD.5 Paroxetine, escitalopram, duloxetine, and venlafaxine are FDA-approved for GAD, but other SSRIs also are used as primary treatment.5 Assuming the selected agent is tolerable and efficacious, a treatment course of 12 months is recommended.13

Benzodiazepines promote binding of the neuroinhibitory transmitter γ-aminobutyric acid and enhance chloride ion influx, thus reducing anxiety. Benzodiazepines have been widely used because of their rapid onset of action and effectiveness in managing anxiety, but their role in long-term management of GAD is unclear because these medications increase the risk of addiction, cognitive dulling, memory impairment, psychomotor retardation, and respiratory depression when combined with other CNS depressants such as alcohol and opiates. Before prescribing a benzodiazepine, conduct a thorough risk-benefit analysis and obtain informed consent. Long-term benzodiazepine monotherapy is not recommended.5,14

Hydroxyzine is an alternative to benzodiazepines.15 It works as an antihistamine and is FDA-approved for psychogenic neurosis, a Freudian distinction encompassing anxiety derived from psychological rather than physiological factors.

The azapirone buspirone is a nonaddictive, generally non-sedating 5-HT1A agonist. Although anecdotally some psychiatrists may report limited clinical utility, many analyses found azapirones, including buspirone, were effective for GAD,16,17 particularly for patients with comorbid depression.18

Tricyclic antidepressants are not a first-line choice because of their side effect profile and potential for drug-drug interactions. Nonetheless, some research suggests imipramine may be a reasonable option for GAD.14,19

Although not FDA-approved for GAD, anticonvulsant and antipsychotic medications may be reasonable adjunctive agents for patients with refractory GAD. Studies have suggested gabapentin20 and quetiapine21 as options.

Investigational treatments. Glutaminergic transmission is being investigated as a target for pharmacotherapy for GAD. In an 8-week, open-label trial, 12 of 15 GAD patients responded to the antiglutamatergic agent riluzole, 100 mg/d, and 8 patients achieved remission.22 In another study, pregabalin, which promotes calcium channel blockade, significantly reduced patients’ scores on the Hamilton Anxiety Rating Scale.23 However, this medication is a schedule V controlled substance and little is known about its long-term effects. Researchers had proposed that inhibition of corticotropin-releasing factor (CRF) may help reduce anxiety, but in a double-blind, placebo-controlled trial, they found that the CRF antagonist pexacerfont was no more effective than placebo.24

CAM treatments. A meta-analysis found that compared with placebo, kava extract (Piper methysticum) effectively reduced anxiety symptoms.25 However, considering its risk for hepatotoxicity, kava is not a recommended treatment.26 Although valerian, St. John’s wort, and passionflower have been used to manage GAD, there is insufficient evidence of their effectiveness and safety.26 No strong evidence supports nutritional supplements such as ginger, amino acids, and omega-3 fatty acids (fish oils) as treatment for GAD. Although there’s limited research on resistance training, aromatherapy, yoga, meditation, or acupuncture for treating anxiety, consider these treatments if your patient finds them helpful, because generally they are not contraindicated.

 

 

CASE CONTINUED: An antidepressant and CBT

Mrs. M reluctantly agrees to a trial of sertraline, 50 mg/d. She refuses a prescription for clonazepam because she is afraid of drug dependence but accepts a referral for CBT. Two days later, she calls the clinic and says she is more anxious and wants to stop the sertraline. The psychiatrist reassures her and reduces the dosage to 25 mg/d.

Mrs. M’s spike of anxiety resolves by her 2-week follow-up appointment and sertraline is titrated to 200 mg/d. Her irritability, anxiety, and mood improve within 2 months. The worry does not completely resolve, but she is much improved at 6 months, and the focus of her therapy shifts to her marriage.

Related Resources

  • Goldberg D, Kendler KS, Sirvatka PJ, et al, eds. Diagnostic issues in depression and generalized anxiety disorder—refining the research agenda for DSM-V. Arlington, VA: American Psychiatric Publishing; 2010.
  • Fricchione G. Generalized anxiety disorder. N Engl J Med. 2004;351(7):675-682.

Drug Brand Names

  • Buspirone • Buspar
  • Clonazepam •Klonopin
  • Diazepam • Valium
  • Duloxetine • Cymbalta
  • Escitalopram • Lexapro
  • Fluoxetine • Prozac
  • Gabapentin• Neurontin
  • Hydroxyzine • Vistaril, Atarax
  • Imipramine • Tofranil
  • Paroxetine • Paxil
  • Pregabalin • Lyrica
  • Quetiapine • Seroquel
  • Riluzole • Rilutek
  • Sertraline • Zoloft
  • Venlafaxine • Effexor

Disclosure

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

References

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

2. Andrews G, Hobbs MJ, Borkovec TD, et al. Generalized worry disorder: a review of DSM-IV generalized anxiety disorder and options for DSM-V. Depress Anxiety. 2010;27(2):134-147.

3. Turk CL, Mennin DS. Phenomenology of generalized anxiety disorder. Psychiatr Ann. 2011;41(2):72-78.

4. Kessler RC, Chiu WT, Demler O, et al. Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62(6):617-627.

5. Ballenger JC, Davidson JR, Lecrubier Y, et al. Consensus statement on generalized anxiety disorder from the International Consensus Group on Depression and Anxiety. J Clin Psychiatry. 2001;6(suppl 11):53-58.

6. Kessler RC, Keller MB, Wittchen HU. The epidemiology of generalized anxiety disorder. Psychiatr Clin North Am. 2001;24(1):19-39.

7. Kavan MG, Elsasser G, Barone EJ. Generalized anxiety disorder: practical assessment and management. Am Fam Physician. 2009;79(9):785-791.

8. Spitzer RL, Kroenke K, Williams JBW, et al. A brief measure for assessing generalized anxiety disorder. Arch Intern Med. 2006;166(10):1092-1097.

9. Newman MG, Borkovec TD. Cognitive-behavioral treatment of generalized anxiety disorder. Clin Psychol. 1995;48(4):5-7.

10. Leichsenring F, Salzer S, Jaeger U, et al. Short-term psychodynamic psychotherapy and cognitive-behavioral therapy in generalized anxiety disorder: a randomized, controlled trial. Am J Psychiatry. 2009;166(8):875-881.

11. Evans S, Ferrando S, Findler M, et al. Mindfulness-based cognitive therapy for generalized anxiety disorder. J Anxiety Disord. 2008;22(4):716-721.

12. Stein MB, Sherbourne CD, Craske MG, et al. Quality of care for primary care patients with anxiety disorders. Am J Psychiatry. 2004;161(12):2230-2237.

13. Fricchione G. Generalized anxiety disorder. N Engl J Med. 2004;351(7):675-682.

14. Rickels K, Downing R, Schweizer E, et al. Antidepressants for the treatment of generalized anxiety disorder. A placebo-controlled comparison of imipramine, trazodone, and diazepam. Arch Gen Psychiatry. 1993;50(11):884-895.

15. Guaiana G, Barbui C, Cipriani A. Hydroxyzine for generalised anxiety disorder. Cochrane Database Syst Rev. 2010;(12):CD006815.-

16. Gammans RE, Stringfellow JC, Hvizdos AJ, et al. Use of buspirone in patients with generalized anxiety disorder and coexisting depressive symptoms. A meta-analysis of eight randomized, controlled studies. Neuropsychobiology. 1992;25(4):193-201.

17. Chessick CA, Allen MH, Thase M, et al. Azapirones for generalized anxiety disorder. Cochrane Database Syst Rev. 2006;(3):CD006115.-

18. Sramek JJ, Tansman M, Suri A, et al. Efficacy of buspirone in generalized anxiety disorder with coexisting mild depressive symptoms. J Clin Psychiatry. 1996;57(7):287-291.

19. Rickels K, DeMartinis N, García-España F, et al. Imipramine and buspirone in treatment of patients with generalized anxiety disorder who are discontinuing long-term benzodiazepine therapy. Am J Psychiatry. 2000;157(12):1973-1979.

20. Pollack MH, Matthews J, Scott EL. Gabapentin as a potential treatment for anxiety disorders. Am J Psychiatry. 1998;155(7):992-993.

21. Khan A, Joyce M, Atkinson S, et al. A randomized, double-blind study of once-daily extended release quetiapine fumarate (quetiapine XR) monotherapy in patients with generalized anxiety disorder. J Clin Psychopharmacol. 2011;31(4):418-428.

22. Mathew SJ, Amiel JM, Coplan JD, et al. Open-label trial of riluzole in generalized anxiety disorder. Am J Psychiatry. 2005;162(12):2379-2381.

23. Pande AC, Crockatt JG, Feltner DE, et al. Pregabalin in generalized anxiety disorder: a placebo-controlled trial. Am J Psychiatry. 2003;160(3):533-540.

24. Coric V, Feldman HH, Oren DA, et al. Multicenter, randomized, double-blind, active comparator and placebo-controlled trial of a corticotropin-releasing factor receptor-1 antagonist in generalized anxiety disorder. Depress Anxiety. 2010;27(5):417-425.

25. Pittler MH, Ernst E. Kava extract for treating anxiety. Cochrane Database Syst Rev. 2003;(1):CD003383.-

26. Zoberi K, Pollard CA. Treating anxiety without SSRIs. J Fam Pract. 2010;59(3):148-154.

Article PDF
Author and Disclosure Information

Matthew J. Barry, DO
Lead Psychiatrist, Rochester Veterans Affairs Outpatient Clinic, Canandaigua Veterans Affairs Medical Center, Canandaigua, NY

Issue
Current Psychiatry - 11(05)
Publications
Topics
Page Number
40-44
Legacy Keywords
generalized anxiety disorder; anxiety; differential diagnosis; GAD-7; psychotherapy; pharmacotherapy; glutaminergic transmission; complimentary and alternative medicine; CAM treatments
Sections
Author and Disclosure Information

Matthew J. Barry, DO
Lead Psychiatrist, Rochester Veterans Affairs Outpatient Clinic, Canandaigua Veterans Affairs Medical Center, Canandaigua, NY

Author and Disclosure Information

Matthew J. Barry, DO
Lead Psychiatrist, Rochester Veterans Affairs Outpatient Clinic, Canandaigua Veterans Affairs Medical Center, Canandaigua, NY

Article PDF
Article PDF

Discuss this article at www.facebook.com/CurrentPsychiatry

Mrs. M, age 44, is a married mother of 2 who presents to the psychiatric clinic with increased anxiety that recently has become intolerable, stating “I can’t stop my head.” She has experienced anxiety for “as long as I can remember.” She was a shy, anxious child who worried about her parents’ health. Her anxiety worsened at college, where she first sought care. She was prescribed diazepam as needed. The next semester, she had a depressive episode, treated with imipramine, 75 mg/d, which she tolerated poorly.

Mrs. M has received episodic supportive therapy since college. She has been plagued by bouts of anxiety and worry, with insomnia, tension, and fatigue. She worries about financial, career, family, and safety issues and has a phobia of spiders. Her family and friends often comment about her excessive worry, and it has strained her marriage and career; she was passed over for a promotion in part because of her anxiousness. Mrs. M also has experienced several depressive episodes.

Mrs. M has sought medical care for various non-specific somatic complaints; all laboratory tests were normal. Approximately 10 years ago, Mrs. M’s primary care physician prescribed fluoxetine, 20 mg/d, but Mrs. M stopped taking it after a few days, stating she felt “more anxious and jittery.”

To meet DSM-IV-TR diagnostic criteria for generalized anxiety disorder (GAD), patients must experience anxiety and worry that they find difficult to control. The worry and anxiety occur more days than not for at least 6 months and cause clinically significant distress and impairment (Table 1).1 These diagnostic criteria are being reevaluated—the DSM-5 Anxiety Work Group has proposed renaming the condition generalized worry disorder, specifying that only 2 domains need to be impacted by worry, shortening the required time frame of impairment from 6 months to 3 months, and including at least 1 behavioral change spawned by excessive worry.2 Although provisional, these recommendations suggest DSM-5 will include changes to GAD when it is published in 2013.

Table 1

DSM-IV-TR diagnostic criteria for generalized anxiety disorder

  1. Excessive anxiety and worry (apprehensive expectation), occurring more days than not for at least 6 months, about a number of events or activities (such as work or school performance)
  2. The person finds it difficult to control the worry
  3. The anxiety and worry are associated with ≥3 of the following 6 symptoms (with at least some symptoms present for more days than not for the past 6 months):
    1. restlessness or feeling keyed up or on edge
    2. being easily fatigued
    3. difficulty concentrating or mind going blank
    4. irritability
    5. muscle tension
    6. sleep disturbance (difficulty falling or staying asleep, or restless, unsatisfying sleep)
  4. The focus of the anxiety and worry is not confined to features of other axis I disorders (eg, social phobia, OCD, PTSD, etc.)
  5. The anxiety, worry, or physical symptoms cause clinically significant distress or impairment in social, occupational, or other important areas of functioning
  6. The disturbance is not due to the direct physiological effects of a substance (eg, a drug of abuse, a medication) or a general medical condition (eg, hyperthyroidism), and does not occur exclusively during a mood disorder, a psychotic disorder, or a pervasive developmental disorder
OCD: obsessive-compulsive disorder; PTSD: posttraumatic stress disorder
Source: Reference 1

A common, chronic condition

In the United States, the lifetime prevalence of GAD is 5.7%.3 It is twice as common in women. Although GAD can occur at any age, 75% of patients develop it before age 47; the median age is 31.3,4 Patients who present with GAD later in life have a better prognosis.3,4

Approximately 90% of GAD patients will meet criteria for another axis I disorder.3 When GAD patients present for treatment, social phobia and panic disorder are the most common comorbid psychiatric disorders. The lifetime prevalence of a mood disorder among GAD patients is 62%, but as few as 6% of GAD patients will meet criteria for a mood disorder at presentation.3 The onset of GAD usually precedes depression.5,6

Patients with GAD often first seek treatment from their primary care provider.7 A useful screening tool is the GAD-7 (Table 2).8 This instrument has a specificity of 92% and sensitivity of 76% for GAD for patients who score ≥8.7 Although the GAD-7 cannot confirm a GAD diagnosis, it can prompt clinicians to conduct a more structured interview. Because higher scores correlate with more severe symptoms, the GAD-7 can be used to measure progress.

Table 2

Screening for generalized anxiety disorder: The GAD-7

Over the last 2 weeks, how often have you been bothered by any of the following problems?Not at allSeveral daysMore than half the daysNearly every day
1.Feeling nervous, anxious, or on edge0123
2.Not being able to stop or control worrying0123
3.Worrying too much about different things0123
4.Trouble relaxing0123
5.Being so restless that it is hard to sit still0123
6.Becoming easily annoyed or irritable0123
7.Feeling afraid as if something awful might happen0123
GAD: generalized anxiety disorder
Source: Reference 8
 

 

Differential diagnosis

GAD typically has a chronic course with fluctuating symptom severity over the patient’s lifespan. Assess patients who present with anxiety for medical conditions that mimic GAD. These include:

  • endocrine (hyperthyroidism), metabolic (electrolyte abnormalities), respiratory (asthma), neurologic (seizure disorder), or cardiac (arrhythmia) conditions
  • nutritional deficiencies, especially of B vitamins and folate
  • ingestion of substances or medications that may cause anxiety, such as caffeine or amphetamines.

A thorough history, medication review, and physical examination—as well as routine tests such as metabolic panel, complete blood count, thyroid function tests, urine drug screen, and electrocardiography—will capture most of these potential etiologies. In addition to ruling out medical causes, also assess for comorbid psychiatric conditions before reaching a diagnosis.

Evidence-based treatments

The treatment armamentarium for GAD includes psychotherapy and pharmacotherapy; complementary and alternative medicine (CAM) modalities may be useful adjunctive treatments. Which approach to use is determined by clinical judgment and the patient’s symptom severity and preferences. Combination therapy consisting of psychotherapy and medication often is appropriate.

Psychotherapy. Cognitive-behavioral therapy (CBT) is the preferred form of psychotherapy for GAD because it results in sustained improvements for patients with anxiety.5,9 Other modalities that may be effective include psychodynamic psychotherapy, mindfulness-based therapy, and interpersonal psychotherapy.10,11

Pharmacotherapy. As few as one-quarter of patients with GAD receive medications at appropriate dose and duration.12 Antidepressants are a first-line pharmacotherapy.5 Selective serotonin reuptake inhibitors (SSRIs) and serotonin-norepinephrine reuptake inhibitors are highly effective for treating GAD.5 Paroxetine, escitalopram, duloxetine, and venlafaxine are FDA-approved for GAD, but other SSRIs also are used as primary treatment.5 Assuming the selected agent is tolerable and efficacious, a treatment course of 12 months is recommended.13

Benzodiazepines promote binding of the neuroinhibitory transmitter γ-aminobutyric acid and enhance chloride ion influx, thus reducing anxiety. Benzodiazepines have been widely used because of their rapid onset of action and effectiveness in managing anxiety, but their role in long-term management of GAD is unclear because these medications increase the risk of addiction, cognitive dulling, memory impairment, psychomotor retardation, and respiratory depression when combined with other CNS depressants such as alcohol and opiates. Before prescribing a benzodiazepine, conduct a thorough risk-benefit analysis and obtain informed consent. Long-term benzodiazepine monotherapy is not recommended.5,14

Hydroxyzine is an alternative to benzodiazepines.15 It works as an antihistamine and is FDA-approved for psychogenic neurosis, a Freudian distinction encompassing anxiety derived from psychological rather than physiological factors.

The azapirone buspirone is a nonaddictive, generally non-sedating 5-HT1A agonist. Although anecdotally some psychiatrists may report limited clinical utility, many analyses found azapirones, including buspirone, were effective for GAD,16,17 particularly for patients with comorbid depression.18

Tricyclic antidepressants are not a first-line choice because of their side effect profile and potential for drug-drug interactions. Nonetheless, some research suggests imipramine may be a reasonable option for GAD.14,19

Although not FDA-approved for GAD, anticonvulsant and antipsychotic medications may be reasonable adjunctive agents for patients with refractory GAD. Studies have suggested gabapentin20 and quetiapine21 as options.

Investigational treatments. Glutaminergic transmission is being investigated as a target for pharmacotherapy for GAD. In an 8-week, open-label trial, 12 of 15 GAD patients responded to the antiglutamatergic agent riluzole, 100 mg/d, and 8 patients achieved remission.22 In another study, pregabalin, which promotes calcium channel blockade, significantly reduced patients’ scores on the Hamilton Anxiety Rating Scale.23 However, this medication is a schedule V controlled substance and little is known about its long-term effects. Researchers had proposed that inhibition of corticotropin-releasing factor (CRF) may help reduce anxiety, but in a double-blind, placebo-controlled trial, they found that the CRF antagonist pexacerfont was no more effective than placebo.24

CAM treatments. A meta-analysis found that compared with placebo, kava extract (Piper methysticum) effectively reduced anxiety symptoms.25 However, considering its risk for hepatotoxicity, kava is not a recommended treatment.26 Although valerian, St. John’s wort, and passionflower have been used to manage GAD, there is insufficient evidence of their effectiveness and safety.26 No strong evidence supports nutritional supplements such as ginger, amino acids, and omega-3 fatty acids (fish oils) as treatment for GAD. Although there’s limited research on resistance training, aromatherapy, yoga, meditation, or acupuncture for treating anxiety, consider these treatments if your patient finds them helpful, because generally they are not contraindicated.

 

 

CASE CONTINUED: An antidepressant and CBT

Mrs. M reluctantly agrees to a trial of sertraline, 50 mg/d. She refuses a prescription for clonazepam because she is afraid of drug dependence but accepts a referral for CBT. Two days later, she calls the clinic and says she is more anxious and wants to stop the sertraline. The psychiatrist reassures her and reduces the dosage to 25 mg/d.

Mrs. M’s spike of anxiety resolves by her 2-week follow-up appointment and sertraline is titrated to 200 mg/d. Her irritability, anxiety, and mood improve within 2 months. The worry does not completely resolve, but she is much improved at 6 months, and the focus of her therapy shifts to her marriage.

Related Resources

  • Goldberg D, Kendler KS, Sirvatka PJ, et al, eds. Diagnostic issues in depression and generalized anxiety disorder—refining the research agenda for DSM-V. Arlington, VA: American Psychiatric Publishing; 2010.
  • Fricchione G. Generalized anxiety disorder. N Engl J Med. 2004;351(7):675-682.

Drug Brand Names

  • Buspirone • Buspar
  • Clonazepam •Klonopin
  • Diazepam • Valium
  • Duloxetine • Cymbalta
  • Escitalopram • Lexapro
  • Fluoxetine • Prozac
  • Gabapentin• Neurontin
  • Hydroxyzine • Vistaril, Atarax
  • Imipramine • Tofranil
  • Paroxetine • Paxil
  • Pregabalin • Lyrica
  • Quetiapine • Seroquel
  • Riluzole • Rilutek
  • Sertraline • Zoloft
  • Venlafaxine • Effexor

Disclosure

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

Discuss this article at www.facebook.com/CurrentPsychiatry

Mrs. M, age 44, is a married mother of 2 who presents to the psychiatric clinic with increased anxiety that recently has become intolerable, stating “I can’t stop my head.” She has experienced anxiety for “as long as I can remember.” She was a shy, anxious child who worried about her parents’ health. Her anxiety worsened at college, where she first sought care. She was prescribed diazepam as needed. The next semester, she had a depressive episode, treated with imipramine, 75 mg/d, which she tolerated poorly.

Mrs. M has received episodic supportive therapy since college. She has been plagued by bouts of anxiety and worry, with insomnia, tension, and fatigue. She worries about financial, career, family, and safety issues and has a phobia of spiders. Her family and friends often comment about her excessive worry, and it has strained her marriage and career; she was passed over for a promotion in part because of her anxiousness. Mrs. M also has experienced several depressive episodes.

Mrs. M has sought medical care for various non-specific somatic complaints; all laboratory tests were normal. Approximately 10 years ago, Mrs. M’s primary care physician prescribed fluoxetine, 20 mg/d, but Mrs. M stopped taking it after a few days, stating she felt “more anxious and jittery.”

To meet DSM-IV-TR diagnostic criteria for generalized anxiety disorder (GAD), patients must experience anxiety and worry that they find difficult to control. The worry and anxiety occur more days than not for at least 6 months and cause clinically significant distress and impairment (Table 1).1 These diagnostic criteria are being reevaluated—the DSM-5 Anxiety Work Group has proposed renaming the condition generalized worry disorder, specifying that only 2 domains need to be impacted by worry, shortening the required time frame of impairment from 6 months to 3 months, and including at least 1 behavioral change spawned by excessive worry.2 Although provisional, these recommendations suggest DSM-5 will include changes to GAD when it is published in 2013.

Table 1

DSM-IV-TR diagnostic criteria for generalized anxiety disorder

  1. Excessive anxiety and worry (apprehensive expectation), occurring more days than not for at least 6 months, about a number of events or activities (such as work or school performance)
  2. The person finds it difficult to control the worry
  3. The anxiety and worry are associated with ≥3 of the following 6 symptoms (with at least some symptoms present for more days than not for the past 6 months):
    1. restlessness or feeling keyed up or on edge
    2. being easily fatigued
    3. difficulty concentrating or mind going blank
    4. irritability
    5. muscle tension
    6. sleep disturbance (difficulty falling or staying asleep, or restless, unsatisfying sleep)
  4. The focus of the anxiety and worry is not confined to features of other axis I disorders (eg, social phobia, OCD, PTSD, etc.)
  5. The anxiety, worry, or physical symptoms cause clinically significant distress or impairment in social, occupational, or other important areas of functioning
  6. The disturbance is not due to the direct physiological effects of a substance (eg, a drug of abuse, a medication) or a general medical condition (eg, hyperthyroidism), and does not occur exclusively during a mood disorder, a psychotic disorder, or a pervasive developmental disorder
OCD: obsessive-compulsive disorder; PTSD: posttraumatic stress disorder
Source: Reference 1

A common, chronic condition

In the United States, the lifetime prevalence of GAD is 5.7%.3 It is twice as common in women. Although GAD can occur at any age, 75% of patients develop it before age 47; the median age is 31.3,4 Patients who present with GAD later in life have a better prognosis.3,4

Approximately 90% of GAD patients will meet criteria for another axis I disorder.3 When GAD patients present for treatment, social phobia and panic disorder are the most common comorbid psychiatric disorders. The lifetime prevalence of a mood disorder among GAD patients is 62%, but as few as 6% of GAD patients will meet criteria for a mood disorder at presentation.3 The onset of GAD usually precedes depression.5,6

Patients with GAD often first seek treatment from their primary care provider.7 A useful screening tool is the GAD-7 (Table 2).8 This instrument has a specificity of 92% and sensitivity of 76% for GAD for patients who score ≥8.7 Although the GAD-7 cannot confirm a GAD diagnosis, it can prompt clinicians to conduct a more structured interview. Because higher scores correlate with more severe symptoms, the GAD-7 can be used to measure progress.

Table 2

Screening for generalized anxiety disorder: The GAD-7

Over the last 2 weeks, how often have you been bothered by any of the following problems?Not at allSeveral daysMore than half the daysNearly every day
1.Feeling nervous, anxious, or on edge0123
2.Not being able to stop or control worrying0123
3.Worrying too much about different things0123
4.Trouble relaxing0123
5.Being so restless that it is hard to sit still0123
6.Becoming easily annoyed or irritable0123
7.Feeling afraid as if something awful might happen0123
GAD: generalized anxiety disorder
Source: Reference 8
 

 

Differential diagnosis

GAD typically has a chronic course with fluctuating symptom severity over the patient’s lifespan. Assess patients who present with anxiety for medical conditions that mimic GAD. These include:

  • endocrine (hyperthyroidism), metabolic (electrolyte abnormalities), respiratory (asthma), neurologic (seizure disorder), or cardiac (arrhythmia) conditions
  • nutritional deficiencies, especially of B vitamins and folate
  • ingestion of substances or medications that may cause anxiety, such as caffeine or amphetamines.

A thorough history, medication review, and physical examination—as well as routine tests such as metabolic panel, complete blood count, thyroid function tests, urine drug screen, and electrocardiography—will capture most of these potential etiologies. In addition to ruling out medical causes, also assess for comorbid psychiatric conditions before reaching a diagnosis.

Evidence-based treatments

The treatment armamentarium for GAD includes psychotherapy and pharmacotherapy; complementary and alternative medicine (CAM) modalities may be useful adjunctive treatments. Which approach to use is determined by clinical judgment and the patient’s symptom severity and preferences. Combination therapy consisting of psychotherapy and medication often is appropriate.

Psychotherapy. Cognitive-behavioral therapy (CBT) is the preferred form of psychotherapy for GAD because it results in sustained improvements for patients with anxiety.5,9 Other modalities that may be effective include psychodynamic psychotherapy, mindfulness-based therapy, and interpersonal psychotherapy.10,11

Pharmacotherapy. As few as one-quarter of patients with GAD receive medications at appropriate dose and duration.12 Antidepressants are a first-line pharmacotherapy.5 Selective serotonin reuptake inhibitors (SSRIs) and serotonin-norepinephrine reuptake inhibitors are highly effective for treating GAD.5 Paroxetine, escitalopram, duloxetine, and venlafaxine are FDA-approved for GAD, but other SSRIs also are used as primary treatment.5 Assuming the selected agent is tolerable and efficacious, a treatment course of 12 months is recommended.13

Benzodiazepines promote binding of the neuroinhibitory transmitter γ-aminobutyric acid and enhance chloride ion influx, thus reducing anxiety. Benzodiazepines have been widely used because of their rapid onset of action and effectiveness in managing anxiety, but their role in long-term management of GAD is unclear because these medications increase the risk of addiction, cognitive dulling, memory impairment, psychomotor retardation, and respiratory depression when combined with other CNS depressants such as alcohol and opiates. Before prescribing a benzodiazepine, conduct a thorough risk-benefit analysis and obtain informed consent. Long-term benzodiazepine monotherapy is not recommended.5,14

Hydroxyzine is an alternative to benzodiazepines.15 It works as an antihistamine and is FDA-approved for psychogenic neurosis, a Freudian distinction encompassing anxiety derived from psychological rather than physiological factors.

The azapirone buspirone is a nonaddictive, generally non-sedating 5-HT1A agonist. Although anecdotally some psychiatrists may report limited clinical utility, many analyses found azapirones, including buspirone, were effective for GAD,16,17 particularly for patients with comorbid depression.18

Tricyclic antidepressants are not a first-line choice because of their side effect profile and potential for drug-drug interactions. Nonetheless, some research suggests imipramine may be a reasonable option for GAD.14,19

Although not FDA-approved for GAD, anticonvulsant and antipsychotic medications may be reasonable adjunctive agents for patients with refractory GAD. Studies have suggested gabapentin20 and quetiapine21 as options.

Investigational treatments. Glutaminergic transmission is being investigated as a target for pharmacotherapy for GAD. In an 8-week, open-label trial, 12 of 15 GAD patients responded to the antiglutamatergic agent riluzole, 100 mg/d, and 8 patients achieved remission.22 In another study, pregabalin, which promotes calcium channel blockade, significantly reduced patients’ scores on the Hamilton Anxiety Rating Scale.23 However, this medication is a schedule V controlled substance and little is known about its long-term effects. Researchers had proposed that inhibition of corticotropin-releasing factor (CRF) may help reduce anxiety, but in a double-blind, placebo-controlled trial, they found that the CRF antagonist pexacerfont was no more effective than placebo.24

CAM treatments. A meta-analysis found that compared with placebo, kava extract (Piper methysticum) effectively reduced anxiety symptoms.25 However, considering its risk for hepatotoxicity, kava is not a recommended treatment.26 Although valerian, St. John’s wort, and passionflower have been used to manage GAD, there is insufficient evidence of their effectiveness and safety.26 No strong evidence supports nutritional supplements such as ginger, amino acids, and omega-3 fatty acids (fish oils) as treatment for GAD. Although there’s limited research on resistance training, aromatherapy, yoga, meditation, or acupuncture for treating anxiety, consider these treatments if your patient finds them helpful, because generally they are not contraindicated.

 

 

CASE CONTINUED: An antidepressant and CBT

Mrs. M reluctantly agrees to a trial of sertraline, 50 mg/d. She refuses a prescription for clonazepam because she is afraid of drug dependence but accepts a referral for CBT. Two days later, she calls the clinic and says she is more anxious and wants to stop the sertraline. The psychiatrist reassures her and reduces the dosage to 25 mg/d.

Mrs. M’s spike of anxiety resolves by her 2-week follow-up appointment and sertraline is titrated to 200 mg/d. Her irritability, anxiety, and mood improve within 2 months. The worry does not completely resolve, but she is much improved at 6 months, and the focus of her therapy shifts to her marriage.

Related Resources

  • Goldberg D, Kendler KS, Sirvatka PJ, et al, eds. Diagnostic issues in depression and generalized anxiety disorder—refining the research agenda for DSM-V. Arlington, VA: American Psychiatric Publishing; 2010.
  • Fricchione G. Generalized anxiety disorder. N Engl J Med. 2004;351(7):675-682.

Drug Brand Names

  • Buspirone • Buspar
  • Clonazepam •Klonopin
  • Diazepam • Valium
  • Duloxetine • Cymbalta
  • Escitalopram • Lexapro
  • Fluoxetine • Prozac
  • Gabapentin• Neurontin
  • Hydroxyzine • Vistaril, Atarax
  • Imipramine • Tofranil
  • Paroxetine • Paxil
  • Pregabalin • Lyrica
  • Quetiapine • Seroquel
  • Riluzole • Rilutek
  • Sertraline • Zoloft
  • Venlafaxine • Effexor

Disclosure

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

References

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

2. Andrews G, Hobbs MJ, Borkovec TD, et al. Generalized worry disorder: a review of DSM-IV generalized anxiety disorder and options for DSM-V. Depress Anxiety. 2010;27(2):134-147.

3. Turk CL, Mennin DS. Phenomenology of generalized anxiety disorder. Psychiatr Ann. 2011;41(2):72-78.

4. Kessler RC, Chiu WT, Demler O, et al. Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62(6):617-627.

5. Ballenger JC, Davidson JR, Lecrubier Y, et al. Consensus statement on generalized anxiety disorder from the International Consensus Group on Depression and Anxiety. J Clin Psychiatry. 2001;6(suppl 11):53-58.

6. Kessler RC, Keller MB, Wittchen HU. The epidemiology of generalized anxiety disorder. Psychiatr Clin North Am. 2001;24(1):19-39.

7. Kavan MG, Elsasser G, Barone EJ. Generalized anxiety disorder: practical assessment and management. Am Fam Physician. 2009;79(9):785-791.

8. Spitzer RL, Kroenke K, Williams JBW, et al. A brief measure for assessing generalized anxiety disorder. Arch Intern Med. 2006;166(10):1092-1097.

9. Newman MG, Borkovec TD. Cognitive-behavioral treatment of generalized anxiety disorder. Clin Psychol. 1995;48(4):5-7.

10. Leichsenring F, Salzer S, Jaeger U, et al. Short-term psychodynamic psychotherapy and cognitive-behavioral therapy in generalized anxiety disorder: a randomized, controlled trial. Am J Psychiatry. 2009;166(8):875-881.

11. Evans S, Ferrando S, Findler M, et al. Mindfulness-based cognitive therapy for generalized anxiety disorder. J Anxiety Disord. 2008;22(4):716-721.

12. Stein MB, Sherbourne CD, Craske MG, et al. Quality of care for primary care patients with anxiety disorders. Am J Psychiatry. 2004;161(12):2230-2237.

13. Fricchione G. Generalized anxiety disorder. N Engl J Med. 2004;351(7):675-682.

14. Rickels K, Downing R, Schweizer E, et al. Antidepressants for the treatment of generalized anxiety disorder. A placebo-controlled comparison of imipramine, trazodone, and diazepam. Arch Gen Psychiatry. 1993;50(11):884-895.

15. Guaiana G, Barbui C, Cipriani A. Hydroxyzine for generalised anxiety disorder. Cochrane Database Syst Rev. 2010;(12):CD006815.-

16. Gammans RE, Stringfellow JC, Hvizdos AJ, et al. Use of buspirone in patients with generalized anxiety disorder and coexisting depressive symptoms. A meta-analysis of eight randomized, controlled studies. Neuropsychobiology. 1992;25(4):193-201.

17. Chessick CA, Allen MH, Thase M, et al. Azapirones for generalized anxiety disorder. Cochrane Database Syst Rev. 2006;(3):CD006115.-

18. Sramek JJ, Tansman M, Suri A, et al. Efficacy of buspirone in generalized anxiety disorder with coexisting mild depressive symptoms. J Clin Psychiatry. 1996;57(7):287-291.

19. Rickels K, DeMartinis N, García-España F, et al. Imipramine and buspirone in treatment of patients with generalized anxiety disorder who are discontinuing long-term benzodiazepine therapy. Am J Psychiatry. 2000;157(12):1973-1979.

20. Pollack MH, Matthews J, Scott EL. Gabapentin as a potential treatment for anxiety disorders. Am J Psychiatry. 1998;155(7):992-993.

21. Khan A, Joyce M, Atkinson S, et al. A randomized, double-blind study of once-daily extended release quetiapine fumarate (quetiapine XR) monotherapy in patients with generalized anxiety disorder. J Clin Psychopharmacol. 2011;31(4):418-428.

22. Mathew SJ, Amiel JM, Coplan JD, et al. Open-label trial of riluzole in generalized anxiety disorder. Am J Psychiatry. 2005;162(12):2379-2381.

23. Pande AC, Crockatt JG, Feltner DE, et al. Pregabalin in generalized anxiety disorder: a placebo-controlled trial. Am J Psychiatry. 2003;160(3):533-540.

24. Coric V, Feldman HH, Oren DA, et al. Multicenter, randomized, double-blind, active comparator and placebo-controlled trial of a corticotropin-releasing factor receptor-1 antagonist in generalized anxiety disorder. Depress Anxiety. 2010;27(5):417-425.

25. Pittler MH, Ernst E. Kava extract for treating anxiety. Cochrane Database Syst Rev. 2003;(1):CD003383.-

26. Zoberi K, Pollard CA. Treating anxiety without SSRIs. J Fam Pract. 2010;59(3):148-154.

References

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

2. Andrews G, Hobbs MJ, Borkovec TD, et al. Generalized worry disorder: a review of DSM-IV generalized anxiety disorder and options for DSM-V. Depress Anxiety. 2010;27(2):134-147.

3. Turk CL, Mennin DS. Phenomenology of generalized anxiety disorder. Psychiatr Ann. 2011;41(2):72-78.

4. Kessler RC, Chiu WT, Demler O, et al. Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62(6):617-627.

5. Ballenger JC, Davidson JR, Lecrubier Y, et al. Consensus statement on generalized anxiety disorder from the International Consensus Group on Depression and Anxiety. J Clin Psychiatry. 2001;6(suppl 11):53-58.

6. Kessler RC, Keller MB, Wittchen HU. The epidemiology of generalized anxiety disorder. Psychiatr Clin North Am. 2001;24(1):19-39.

7. Kavan MG, Elsasser G, Barone EJ. Generalized anxiety disorder: practical assessment and management. Am Fam Physician. 2009;79(9):785-791.

8. Spitzer RL, Kroenke K, Williams JBW, et al. A brief measure for assessing generalized anxiety disorder. Arch Intern Med. 2006;166(10):1092-1097.

9. Newman MG, Borkovec TD. Cognitive-behavioral treatment of generalized anxiety disorder. Clin Psychol. 1995;48(4):5-7.

10. Leichsenring F, Salzer S, Jaeger U, et al. Short-term psychodynamic psychotherapy and cognitive-behavioral therapy in generalized anxiety disorder: a randomized, controlled trial. Am J Psychiatry. 2009;166(8):875-881.

11. Evans S, Ferrando S, Findler M, et al. Mindfulness-based cognitive therapy for generalized anxiety disorder. J Anxiety Disord. 2008;22(4):716-721.

12. Stein MB, Sherbourne CD, Craske MG, et al. Quality of care for primary care patients with anxiety disorders. Am J Psychiatry. 2004;161(12):2230-2237.

13. Fricchione G. Generalized anxiety disorder. N Engl J Med. 2004;351(7):675-682.

14. Rickels K, Downing R, Schweizer E, et al. Antidepressants for the treatment of generalized anxiety disorder. A placebo-controlled comparison of imipramine, trazodone, and diazepam. Arch Gen Psychiatry. 1993;50(11):884-895.

15. Guaiana G, Barbui C, Cipriani A. Hydroxyzine for generalised anxiety disorder. Cochrane Database Syst Rev. 2010;(12):CD006815.-

16. Gammans RE, Stringfellow JC, Hvizdos AJ, et al. Use of buspirone in patients with generalized anxiety disorder and coexisting depressive symptoms. A meta-analysis of eight randomized, controlled studies. Neuropsychobiology. 1992;25(4):193-201.

17. Chessick CA, Allen MH, Thase M, et al. Azapirones for generalized anxiety disorder. Cochrane Database Syst Rev. 2006;(3):CD006115.-

18. Sramek JJ, Tansman M, Suri A, et al. Efficacy of buspirone in generalized anxiety disorder with coexisting mild depressive symptoms. J Clin Psychiatry. 1996;57(7):287-291.

19. Rickels K, DeMartinis N, García-España F, et al. Imipramine and buspirone in treatment of patients with generalized anxiety disorder who are discontinuing long-term benzodiazepine therapy. Am J Psychiatry. 2000;157(12):1973-1979.

20. Pollack MH, Matthews J, Scott EL. Gabapentin as a potential treatment for anxiety disorders. Am J Psychiatry. 1998;155(7):992-993.

21. Khan A, Joyce M, Atkinson S, et al. A randomized, double-blind study of once-daily extended release quetiapine fumarate (quetiapine XR) monotherapy in patients with generalized anxiety disorder. J Clin Psychopharmacol. 2011;31(4):418-428.

22. Mathew SJ, Amiel JM, Coplan JD, et al. Open-label trial of riluzole in generalized anxiety disorder. Am J Psychiatry. 2005;162(12):2379-2381.

23. Pande AC, Crockatt JG, Feltner DE, et al. Pregabalin in generalized anxiety disorder: a placebo-controlled trial. Am J Psychiatry. 2003;160(3):533-540.

24. Coric V, Feldman HH, Oren DA, et al. Multicenter, randomized, double-blind, active comparator and placebo-controlled trial of a corticotropin-releasing factor receptor-1 antagonist in generalized anxiety disorder. Depress Anxiety. 2010;27(5):417-425.

25. Pittler MH, Ernst E. Kava extract for treating anxiety. Cochrane Database Syst Rev. 2003;(1):CD003383.-

26. Zoberi K, Pollard CA. Treating anxiety without SSRIs. J Fam Pract. 2010;59(3):148-154.

Issue
Current Psychiatry - 11(05)
Issue
Current Psychiatry - 11(05)
Page Number
40-44
Page Number
40-44
Publications
Publications
Topics
Article Type
Display Headline
Generalized anxiety disorder: Helping patients overcome worry
Display Headline
Generalized anxiety disorder: Helping patients overcome worry
Legacy Keywords
generalized anxiety disorder; anxiety; differential diagnosis; GAD-7; psychotherapy; pharmacotherapy; glutaminergic transmission; complimentary and alternative medicine; CAM treatments
Legacy Keywords
generalized anxiety disorder; anxiety; differential diagnosis; GAD-7; psychotherapy; pharmacotherapy; glutaminergic transmission; complimentary and alternative medicine; CAM treatments
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Evaluating older adults’ capacity and need for guardianship

Article Type
Changed
Tue, 12/11/2018 - 15:04
Display Headline
Evaluating older adults’ capacity and need for guardianship

Discuss this article at www.facebook.com/CurrentPsychiatry

Although forensic psychiatrists typically are consulted in complex legal matters, geriatric, consultation-liaison, and general psychiatrists are on the front lines of assessing capacity to give informed consent and need for guardianship. Psychiatrists often find such consultations daunting because residency training usually includes little to no formal training in performing psycho-legal assessments. Evaluating issues such as decision-making capacity, guardianship, and capacity to give informed consent requires a delicate balance between autonomy and beneficence. This article reviews 4 common legal issues in geriatric consultation—capacity evaluations, informed consent, guardianship, and elder abuse—and suggests a systematic approach to psycho-legal consultations in older adults.

Confidentiality and dual agency

Every psychiatrist should be familiar with basic principles of medical ethics as well as key aspects of local mental health law. Relevant ethical principles include autonomy, beneficence, confidentiality, and dual agency. A review of all these ethical issues is beyond the scope of this article, so here I highlight confidentiality and dual agency.

Confidentiality—the clinician’s obligation not to disclose private medical information—is a legal as well as an ethical requirement. A psychiatrist who performs a psycho-legal evaluation must disclose to the patient the purpose of the evaluation, that a report will be prepared, and to whom it will be submitted. Exceptions to confidentiality include medical emergencies, mandatory reporting of abuse and infectious diseases, and the duty to protect (warning police and the intended victim when a patient makes a threat).

Dual agency or dual role refers to serving as both a treating physician and a forensic evaluator. Although it is ideal to avoid serving in a dual role, sometimes it is impractical or impossible to avoid doing so, such as in guardianship or civil commitment evaluations, or in state forensic hospitals. In such cases, the psychiatrist must be aware of potential conflicts between clinical and forensic evaluations. A treating psychiatrist primarily serves his or her patient’s best interest, whereas a forensic psychiatrist primarily seeks truth.1 A treating psychiatrist is at risk of consciously or unconsciously biasing his or her psycho-legal evaluation in favor of or against the patient/litigant, depending upon the psychiatrist’s countertransference. Further, performing a psycho-legal evaluation can cause problems in ongoing treatment. A psychiatrist who testifies that his or her fiercely independent patient needs a guardian or nursing home placement will experience significant challenges in continuing to work with that patient.

4 common issues for older adults

Decision-making capacity. Although “capacity” and “competence” often are used interchangeably, “capacity” broadly refers to the ability to perform a specific task, whereas “competence” refers to the legally defined standard for performing a specific task such as making a will. “Competence” is legally determined, whereas “capacity” may be determined clinically.

Capacity usually is task-specific rather than a general construct. The existence of physical or mental illness per se does not mean that a patient lacks capacity. Rather, capacity is determined by whether an individual has specific abilities, regardless of diagnosis. Specific capacities include the ability to give informed consent, manage finances, make a will, or enter into contracts (Table 1).2-4 Appelbaum and Gutheil describe 4 components for assessing specific capacity:

  • communication of a choice
  • factual understanding of the issues
  • appreciation of the situation and its consequences
  • rational manipulation of information.5,6

Table 1

Criteria of 3 specific capacities

CapacityCriteria
Capacity to give informed consentUnderstand nature of illness and treatment
Understand risks and benefits of treatment
Understand treatment alternatives
Understand risk of refusing treatment
Testamentary capacityUnderstand that he/she is making a will
Know the nature and extent of their property
Understand the “natural objects” of their bounty and their claims upon them
Contractual capacityUnderstand the transaction
Act in a reasonable manner
Source: References 2-4

Ability to communicate a choice refers to a patient’s ability to express his or her wishes in a reasonably stable manner. Factual understanding of the issues refers to an individual’s ability to understand the relevant facts before making a decision. Appreciation of the situation and its consequences refers to a person’s ability to rationally understand the effect of decisions. Appreciation is a higher level of understanding than mere factual understanding—eg, a delusional patient who believes himself immortal may intellectually understand that a surgical procedure carries a 50% mortality risk, but may be unable to appreciate the information as it relates to him because he believes he is immortal. Rational manipulation of information refers to a patient’s reasoning process and how the patient integrates data into his or her decision-making process.5

 

 

Informed consent. In my experience, capacity to give informed consent is the most commonly requested specific capacity assessment in general medical settings. Informed consent must be knowing, voluntary, and competent. All material information—information that would cause a reasonable person to accept or reject a proposed treatment—should be communicated to the patient. Informed consent requires an understanding of the patient’s condition and indication for treatment, risks and benefits of and alternatives to treatment, and risks of declining treatment.2,3 Exceptions to informed consent include incompetence, medical emergencies, patient waiver of informed consent, and a limited therapeutic privilege (when a physician determines that the information would harm the patient).3

Several instruments can help clinicians assess patients’ capacity to give informed consent. The benefits of using a structured instrument include:

  • ensuring that specific information is covered during each evaluation
  • systematically recording a patient’s response.5

Disadvantages of using instruments include the fact that no instrument can take into account all aspects of a particular case, and some instruments are time-consuming and require training. Structured instruments can be a useful adjunct to the clinical interview in some cases, but should not substitute for it. In a review of 23 instruments for assessing decisional capacity to consent to treatment or clinical research, the MacArthur Competence Assessment Tool for Clinical Research and the MacArthur Competence Assessment Tool for Treatment had the most empirical support, although the authors noted that other instruments might be better suited to specific situations.7

Psychiatrists may be consulted when a patient refuses treatment or decides to leave the hospital against medical advice. The key issue in both situations is whether the patient has capacity to refuse treatment.8 If there is evidence that the patient is mentally ill and poses an imminent risk of suicide or violence or is unable to provide for his or her basic needs, the psychiatrist should assess whether the patient meets criteria for civil commitment.

Many clinicians employ a “sliding scale” approach to competence, requiring a lower degree of competence for consenting to low-risk, high-benefit interventions and a greater degree of competence for higher-risk procedures.5,9 Family members often serve as informal surrogate decision makers for incapacitated patients, except when there is significant family discord or no family members are available.5

Guardianship. Guardians are appointed by courts to make decisions for individuals who have been found incompetent (wards). Although its purposes are beneficent, the guardianship system could do significant harm.10 Determining that an individual is incompetent is tantamount to depriving him or her of basic personhood. In many cases, the ward loses the ability to consent to or refuse medical care, manage his or her finances, enter into contracts, marry, and determine where he or she will live. On the other hand, failing to recognize incompetence can leave a vulnerable person in danger of physical deterioration, abuse, neglect, or exploitation.

It is critical that guardianship evaluations be conducted carefully. In a review of 298 guardianship cases from 3 states, Moye and colleagues11 found that the quality of the reports was significantly better in Colorado, a state with guardianship reforms, but documentation of functional strengths and weaknesses was “particularly rare” in guardianship evaluations and prognosis often was not included. This information is relevant to judges, who need to determine which areas of function are impaired and how long the impairment is likely to last.

Guardianship evaluations often focus on general rather than specific capacity. In other words, often there is not a specific task such as consenting to surgery that the alleged incompetent person needs to perform. Rather, the question is whether an individual can manage his or her finances or make treatment decisions in general. Appelbaum and Gutheil suggest considering 6 factors when assessing general capacity:

  • awareness of the situation
  • factual understanding of the issues
  • appreciation of the likely consequences
  • rational manipulation of information
  • functioning in one’s environment
  • extent of demands on patient.5

The first 4 are closely related to the elements of specific capacity described above. Functioning in one’s environment and extent of demands on the patient attempt to anticipate the tasks that an individual will need to perform. A patient with mild dementia may be unable to manage a complex estate but can handle a bank account and a fixed income. Similarly, it is important to consider the patient’s support system. An impaired patient may function adequately with his wife’s help but may lose the capacity to live independently if his wife dies or becomes impaired.

 

 

Traditionally, guardianship has resulted in a complete loss of decision-making ability. Several state legislatures have passed laws allowing for limited guardianship, although orders for limited guardianship remain underutilized.10 Limited guardianship delineates specific areas of incompetence and limits the guardian’s decision-making authority to those areas while leaving intact the ward’s ability to make all other decisions for himself or herself.

The use of less-restrictive alternatives to guardianship—such as powers of attorney, durable powers of attorney, living wills, payees, and trusts—is increasing. A power of attorney allows a patient to authorize a specific individual to act on his or her behalf. The scope of the power of attorney can be limited, such as to manage finances or even to a specific transaction, such as selling a home or car. A durable power of attorney also allows an agent to make decisions on the patient’s behalf but becomes active only when the patient becomes incompetent. It often is used to appoint an individual to make medical decisions on behalf of an incompetent patient. Living wills allow patients to determine what treatment they would like in the event they become incompetent.

Elder abuse. An estimated 1 to 2 million adults age >65 have been abused, exploited, or neglected.12 Elder abuse includes physical abuse, neglect, emotional abuse, sexual abuse, and financial exploitation (including undue influence). Most states have mandatory reporting of elder abuse, although they vary regarding who must report and what the report must entail. Psychiatrists should be vigilant in looking for signs of elder abuse (Table 2),13 regardless of the reason for the consult.

Table 2

Signs of elder abuse: What to wlook for

Type of abuseSigns
PhysicalBruises, burns (especially circular, suggesting cigarette burns), slap marks
SexualUnexplained sexually transmitted diseases, bruises in genital area, breasts, or anal area
EmotionalWithdrawal, new-onset depression
FinancialSudden loss of property, unusual increase in spending, checks paid in large, round numbers, checks marked as gifts or loans
NeglectMalnutrition, lack of medical care, poor hygiene, pressure ulcers
Source: Reference 13

10 tips for thorough evaluations

1. Consider the context of the consultation. This includes medical factors (such as the patient’s condition, prognosis, relationship with the treatment team, and recommended course of treatment), legal factors (eg, pending litigation and relevant legal standards for issues such as guardianship), and psychosocial issues (eg, the patient’s current support structure and family conflicts).

2. Identify the legal issue and any relevant legal standards. The legal standard will inform you of the issues you need to address in the evaluation. If an attorney has consulted you, ask him or her to provide the legal standard.

3. Gather relevant collateral information, which may include interviews with family members or a review of financial or medical records.

4. Explain the purpose of the examination and the limits of confidentiality.

5. Perform a focused psychiatric evaluation, paying special attention to cognitive functioning, reasoning, and unusual thought content such as delusional beliefs.

6. Perform an interview specific to the referral issue.

7. Consider using a relevant assessment instrument.

8. Consider psychological testing, laboratory testing, imaging, or further medical evaluation. These assessments can help determine the diagnosis, the cause of any deficits in capacity, and whether any deficits are reversible.

9. Determine what opinions you are able to render. Limit opinions and remember that it may be appropriate to decline to address certain issues if there is insufficient information or if the issue is outside your area of expertise.

10. Prepare a written report. Consider the audience. Minimize the use of medical jargon and define all medical terms. State your opinions clearly and with reasonable medical certainty (in most jurisdictions, this means more likely than not). State the basis for all opinions.

For a case study that provides an example of a psycho-legal evaluation of a geriatric patient, see the Box.

Box

Case study: Unfaithfully yours?

Mr. A, age 75, recently started taking a dopaminergic agonist to treat Parkinson’s disease. He says he wants to divorce his wife of 35 years because of “scandalous affairs” she allegedly engaged in. His wife reports that he has been accusing her of having affairs with various men, including a man who recently painted their house.

On evaluation, Mr. A’s Mini-Mental State Examination score is 30/30. He has no signs of depression and his sleep patterns have not changed. There have been no changes in his spending patterns, although he no longer gives his wife money for grocery shopping, telling her to get money from her “boyfriends.” He is adamant about this decision, saying, “It’s my money and I can do with it as I please. This is still a free country, isn’t it?”

He says he has $70,000 in his individual retirement account, $20,000 in his bank account, and receives a pension of $1,785 per month. He estimates that his home is worth $200,000. His financial records essentially are consistent with his reports. He is able to perform basic calculations without difficulty and is aware of his monthly expenses. He describes his relationship with his wife by saying, “It was fine until she started screwing around.”

When asked about the likely consequences of his decision, he shrugs and says, “I guess she’ll have to get money from her boyfriends. I don’t really care who she sees as long as they stay away from me.” He denies having thoughts of harming his wife or her alleged “boyfriends.” He recognizes that his wife might divorce him, leaving him alone.

When I ask Mr. A if it is possible he is mistaken in his belief that his wife is having affairs, he says, “No, doctor. You don’t know her.” When I ask how he knows she is having affairs, he says that the painter started looking at him “funny” and that the busboy at a restaurant they frequent called his wife “dear.” He believes his wife is having sexual relations with both of these men.

Does Mr. A require a guardian? I opine that Mr. A requires a guardian of estate (to manage his property) but not a guardian of person because he is capable of making decisions about his medical care and other personal decisions. He is failing to care for his wife because of his delusional jealousy. Although cognitively intact, he is unable to appreciate the consequences of his actions or rationally manipulate information because of his delusional thinking. He believes he is “cutting off” an unfaithful spouse when, in fact, there is no evidence that she has been unfaithful. His inability to rationally manipulate information is demonstrated by the fact that he uses innocuous facts such as a busboy calling his elderly wife “dear” to support his delusion that she was having affairs.

I note that his psychosis is reversible because it is likely due to his antiparkinsonian regimen. However, he declines both a dose reduction in his medication and antipsychotic treatment. I note that should his psychosis resolve, he may regain financial decision-making capacity.

 

 

Related Resources

Disclosure

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

Acknowledgement

The author thanks forensic psychiatry fellow Abhishek Jain, MD for reviewing the article and offering editorial suggestions.

References

1. American Academy of Psychiatry and the Law. Ethics guidelines for the practice of forensic psychiatry. http://www.aapl.org/ethics.htm. Adopted May 2005. Accessed February 24 2012.

2. Blank K. Legal and ethical issues. In: Sadvoy J Jarvik LF, Grossberg GT, et al, eds. Comprehensive textbook of geriatric psychiatry. 3rd ed. New York, NY: American Association of Geriatric Psychiatry; 2004:1183-1206.

3. Schwartz HI, Mack DM. Informed consent and competency. In: Rosner R ed. Principles and practice of forensic psychiatry. 2nd ed. New York, NY: Oxford University Press; 2003:97-106.

4. Ciccone RJ. Civil competencies. In: Rosner R ed. Principles and practice of forensic psychiatry. 2nd ed. New York, NY: Oxford University Press; 2003:308-315.

5. Appelbaum PS, Gutheil TG. Competence and substitute decision making. In: Appelbaum PS Gutheil TG, eds. Clinical handbook of psychiatry and the law. 4th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2007:177-213.

6. Appelbaum PS, Grisso T. Assessing patients’ capacities to consent to treatment. N Engl J Med. 1988;319(25):1635-1638.

7. Dunn LB, Nowrangi MA, Palmer BW, et al. Assessing decisional capacity for clinical research or treatment: a review of instruments. Am J Psychiatry. 2006;163(8):1323-1334.

8. Resnick PJ, Sorrentino R. Forensic issues in consultation-liaison psychiatry. Psychiatric Times. 2005;23(14).-

9. Drane JF. The many faces of competency. Hastings Cent Rep. 1985;15(2):17-21.

10. Wright JL. Guardianship for your own good: improving the well being of respondents and wards in the USA. Int J Law Psychiatry. 2010;33(5-6):350-368.

11. Moye J, Wood S, Edelstein B, et al. Clinical evidence in guardianship of older adults is inadequate: findings from a tri-state study. Gerontologist. 2007;47(5):604-612.

12. National Center on Elder Abuse. Fact sheet: elder abuse prevalence and incidence. http://www.ncea.aoa.gov/NCEAroot/Main_Site/pdf/publication/FinalStatistics050331.pdf. Accessed February 24, 2012.

13. National Center on Elder Abuse. Why should I care about elder abuse? http://www.ncea.aoa.gov/Main_Site/pdf/publication/NCEA_WhatIsAbuse-2010.pdf. Published March 3 2010. Accessed February 24, 2012.

Article PDF
Author and Disclosure Information

Sherif Soliman, MD
Assistant Clinical Professor, Department of Psychiatry, Case Western Reserve University, Cleveland, OH, Northcoast Behavioral Healthcare, Northfield, OH

Issue
Current Psychiatry - 11(04)
Publications
Page Number
39-54
Legacy Keywords
capacity; assessment; geriatrics; legal; decision-making; informed consent; guardianship; elder abuse
Sections
Author and Disclosure Information

Sherif Soliman, MD
Assistant Clinical Professor, Department of Psychiatry, Case Western Reserve University, Cleveland, OH, Northcoast Behavioral Healthcare, Northfield, OH

Author and Disclosure Information

Sherif Soliman, MD
Assistant Clinical Professor, Department of Psychiatry, Case Western Reserve University, Cleveland, OH, Northcoast Behavioral Healthcare, Northfield, OH

Article PDF
Article PDF

Discuss this article at www.facebook.com/CurrentPsychiatry

Although forensic psychiatrists typically are consulted in complex legal matters, geriatric, consultation-liaison, and general psychiatrists are on the front lines of assessing capacity to give informed consent and need for guardianship. Psychiatrists often find such consultations daunting because residency training usually includes little to no formal training in performing psycho-legal assessments. Evaluating issues such as decision-making capacity, guardianship, and capacity to give informed consent requires a delicate balance between autonomy and beneficence. This article reviews 4 common legal issues in geriatric consultation—capacity evaluations, informed consent, guardianship, and elder abuse—and suggests a systematic approach to psycho-legal consultations in older adults.

Confidentiality and dual agency

Every psychiatrist should be familiar with basic principles of medical ethics as well as key aspects of local mental health law. Relevant ethical principles include autonomy, beneficence, confidentiality, and dual agency. A review of all these ethical issues is beyond the scope of this article, so here I highlight confidentiality and dual agency.

Confidentiality—the clinician’s obligation not to disclose private medical information—is a legal as well as an ethical requirement. A psychiatrist who performs a psycho-legal evaluation must disclose to the patient the purpose of the evaluation, that a report will be prepared, and to whom it will be submitted. Exceptions to confidentiality include medical emergencies, mandatory reporting of abuse and infectious diseases, and the duty to protect (warning police and the intended victim when a patient makes a threat).

Dual agency or dual role refers to serving as both a treating physician and a forensic evaluator. Although it is ideal to avoid serving in a dual role, sometimes it is impractical or impossible to avoid doing so, such as in guardianship or civil commitment evaluations, or in state forensic hospitals. In such cases, the psychiatrist must be aware of potential conflicts between clinical and forensic evaluations. A treating psychiatrist primarily serves his or her patient’s best interest, whereas a forensic psychiatrist primarily seeks truth.1 A treating psychiatrist is at risk of consciously or unconsciously biasing his or her psycho-legal evaluation in favor of or against the patient/litigant, depending upon the psychiatrist’s countertransference. Further, performing a psycho-legal evaluation can cause problems in ongoing treatment. A psychiatrist who testifies that his or her fiercely independent patient needs a guardian or nursing home placement will experience significant challenges in continuing to work with that patient.

4 common issues for older adults

Decision-making capacity. Although “capacity” and “competence” often are used interchangeably, “capacity” broadly refers to the ability to perform a specific task, whereas “competence” refers to the legally defined standard for performing a specific task such as making a will. “Competence” is legally determined, whereas “capacity” may be determined clinically.

Capacity usually is task-specific rather than a general construct. The existence of physical or mental illness per se does not mean that a patient lacks capacity. Rather, capacity is determined by whether an individual has specific abilities, regardless of diagnosis. Specific capacities include the ability to give informed consent, manage finances, make a will, or enter into contracts (Table 1).2-4 Appelbaum and Gutheil describe 4 components for assessing specific capacity:

  • communication of a choice
  • factual understanding of the issues
  • appreciation of the situation and its consequences
  • rational manipulation of information.5,6

Table 1

Criteria of 3 specific capacities

CapacityCriteria
Capacity to give informed consentUnderstand nature of illness and treatment
Understand risks and benefits of treatment
Understand treatment alternatives
Understand risk of refusing treatment
Testamentary capacityUnderstand that he/she is making a will
Know the nature and extent of their property
Understand the “natural objects” of their bounty and their claims upon them
Contractual capacityUnderstand the transaction
Act in a reasonable manner
Source: References 2-4

Ability to communicate a choice refers to a patient’s ability to express his or her wishes in a reasonably stable manner. Factual understanding of the issues refers to an individual’s ability to understand the relevant facts before making a decision. Appreciation of the situation and its consequences refers to a person’s ability to rationally understand the effect of decisions. Appreciation is a higher level of understanding than mere factual understanding—eg, a delusional patient who believes himself immortal may intellectually understand that a surgical procedure carries a 50% mortality risk, but may be unable to appreciate the information as it relates to him because he believes he is immortal. Rational manipulation of information refers to a patient’s reasoning process and how the patient integrates data into his or her decision-making process.5

 

 

Informed consent. In my experience, capacity to give informed consent is the most commonly requested specific capacity assessment in general medical settings. Informed consent must be knowing, voluntary, and competent. All material information—information that would cause a reasonable person to accept or reject a proposed treatment—should be communicated to the patient. Informed consent requires an understanding of the patient’s condition and indication for treatment, risks and benefits of and alternatives to treatment, and risks of declining treatment.2,3 Exceptions to informed consent include incompetence, medical emergencies, patient waiver of informed consent, and a limited therapeutic privilege (when a physician determines that the information would harm the patient).3

Several instruments can help clinicians assess patients’ capacity to give informed consent. The benefits of using a structured instrument include:

  • ensuring that specific information is covered during each evaluation
  • systematically recording a patient’s response.5

Disadvantages of using instruments include the fact that no instrument can take into account all aspects of a particular case, and some instruments are time-consuming and require training. Structured instruments can be a useful adjunct to the clinical interview in some cases, but should not substitute for it. In a review of 23 instruments for assessing decisional capacity to consent to treatment or clinical research, the MacArthur Competence Assessment Tool for Clinical Research and the MacArthur Competence Assessment Tool for Treatment had the most empirical support, although the authors noted that other instruments might be better suited to specific situations.7

Psychiatrists may be consulted when a patient refuses treatment or decides to leave the hospital against medical advice. The key issue in both situations is whether the patient has capacity to refuse treatment.8 If there is evidence that the patient is mentally ill and poses an imminent risk of suicide or violence or is unable to provide for his or her basic needs, the psychiatrist should assess whether the patient meets criteria for civil commitment.

Many clinicians employ a “sliding scale” approach to competence, requiring a lower degree of competence for consenting to low-risk, high-benefit interventions and a greater degree of competence for higher-risk procedures.5,9 Family members often serve as informal surrogate decision makers for incapacitated patients, except when there is significant family discord or no family members are available.5

Guardianship. Guardians are appointed by courts to make decisions for individuals who have been found incompetent (wards). Although its purposes are beneficent, the guardianship system could do significant harm.10 Determining that an individual is incompetent is tantamount to depriving him or her of basic personhood. In many cases, the ward loses the ability to consent to or refuse medical care, manage his or her finances, enter into contracts, marry, and determine where he or she will live. On the other hand, failing to recognize incompetence can leave a vulnerable person in danger of physical deterioration, abuse, neglect, or exploitation.

It is critical that guardianship evaluations be conducted carefully. In a review of 298 guardianship cases from 3 states, Moye and colleagues11 found that the quality of the reports was significantly better in Colorado, a state with guardianship reforms, but documentation of functional strengths and weaknesses was “particularly rare” in guardianship evaluations and prognosis often was not included. This information is relevant to judges, who need to determine which areas of function are impaired and how long the impairment is likely to last.

Guardianship evaluations often focus on general rather than specific capacity. In other words, often there is not a specific task such as consenting to surgery that the alleged incompetent person needs to perform. Rather, the question is whether an individual can manage his or her finances or make treatment decisions in general. Appelbaum and Gutheil suggest considering 6 factors when assessing general capacity:

  • awareness of the situation
  • factual understanding of the issues
  • appreciation of the likely consequences
  • rational manipulation of information
  • functioning in one’s environment
  • extent of demands on patient.5

The first 4 are closely related to the elements of specific capacity described above. Functioning in one’s environment and extent of demands on the patient attempt to anticipate the tasks that an individual will need to perform. A patient with mild dementia may be unable to manage a complex estate but can handle a bank account and a fixed income. Similarly, it is important to consider the patient’s support system. An impaired patient may function adequately with his wife’s help but may lose the capacity to live independently if his wife dies or becomes impaired.

 

 

Traditionally, guardianship has resulted in a complete loss of decision-making ability. Several state legislatures have passed laws allowing for limited guardianship, although orders for limited guardianship remain underutilized.10 Limited guardianship delineates specific areas of incompetence and limits the guardian’s decision-making authority to those areas while leaving intact the ward’s ability to make all other decisions for himself or herself.

The use of less-restrictive alternatives to guardianship—such as powers of attorney, durable powers of attorney, living wills, payees, and trusts—is increasing. A power of attorney allows a patient to authorize a specific individual to act on his or her behalf. The scope of the power of attorney can be limited, such as to manage finances or even to a specific transaction, such as selling a home or car. A durable power of attorney also allows an agent to make decisions on the patient’s behalf but becomes active only when the patient becomes incompetent. It often is used to appoint an individual to make medical decisions on behalf of an incompetent patient. Living wills allow patients to determine what treatment they would like in the event they become incompetent.

Elder abuse. An estimated 1 to 2 million adults age >65 have been abused, exploited, or neglected.12 Elder abuse includes physical abuse, neglect, emotional abuse, sexual abuse, and financial exploitation (including undue influence). Most states have mandatory reporting of elder abuse, although they vary regarding who must report and what the report must entail. Psychiatrists should be vigilant in looking for signs of elder abuse (Table 2),13 regardless of the reason for the consult.

Table 2

Signs of elder abuse: What to wlook for

Type of abuseSigns
PhysicalBruises, burns (especially circular, suggesting cigarette burns), slap marks
SexualUnexplained sexually transmitted diseases, bruises in genital area, breasts, or anal area
EmotionalWithdrawal, new-onset depression
FinancialSudden loss of property, unusual increase in spending, checks paid in large, round numbers, checks marked as gifts or loans
NeglectMalnutrition, lack of medical care, poor hygiene, pressure ulcers
Source: Reference 13

10 tips for thorough evaluations

1. Consider the context of the consultation. This includes medical factors (such as the patient’s condition, prognosis, relationship with the treatment team, and recommended course of treatment), legal factors (eg, pending litigation and relevant legal standards for issues such as guardianship), and psychosocial issues (eg, the patient’s current support structure and family conflicts).

2. Identify the legal issue and any relevant legal standards. The legal standard will inform you of the issues you need to address in the evaluation. If an attorney has consulted you, ask him or her to provide the legal standard.

3. Gather relevant collateral information, which may include interviews with family members or a review of financial or medical records.

4. Explain the purpose of the examination and the limits of confidentiality.

5. Perform a focused psychiatric evaluation, paying special attention to cognitive functioning, reasoning, and unusual thought content such as delusional beliefs.

6. Perform an interview specific to the referral issue.

7. Consider using a relevant assessment instrument.

8. Consider psychological testing, laboratory testing, imaging, or further medical evaluation. These assessments can help determine the diagnosis, the cause of any deficits in capacity, and whether any deficits are reversible.

9. Determine what opinions you are able to render. Limit opinions and remember that it may be appropriate to decline to address certain issues if there is insufficient information or if the issue is outside your area of expertise.

10. Prepare a written report. Consider the audience. Minimize the use of medical jargon and define all medical terms. State your opinions clearly and with reasonable medical certainty (in most jurisdictions, this means more likely than not). State the basis for all opinions.

For a case study that provides an example of a psycho-legal evaluation of a geriatric patient, see the Box.

Box

Case study: Unfaithfully yours?

Mr. A, age 75, recently started taking a dopaminergic agonist to treat Parkinson’s disease. He says he wants to divorce his wife of 35 years because of “scandalous affairs” she allegedly engaged in. His wife reports that he has been accusing her of having affairs with various men, including a man who recently painted their house.

On evaluation, Mr. A’s Mini-Mental State Examination score is 30/30. He has no signs of depression and his sleep patterns have not changed. There have been no changes in his spending patterns, although he no longer gives his wife money for grocery shopping, telling her to get money from her “boyfriends.” He is adamant about this decision, saying, “It’s my money and I can do with it as I please. This is still a free country, isn’t it?”

He says he has $70,000 in his individual retirement account, $20,000 in his bank account, and receives a pension of $1,785 per month. He estimates that his home is worth $200,000. His financial records essentially are consistent with his reports. He is able to perform basic calculations without difficulty and is aware of his monthly expenses. He describes his relationship with his wife by saying, “It was fine until she started screwing around.”

When asked about the likely consequences of his decision, he shrugs and says, “I guess she’ll have to get money from her boyfriends. I don’t really care who she sees as long as they stay away from me.” He denies having thoughts of harming his wife or her alleged “boyfriends.” He recognizes that his wife might divorce him, leaving him alone.

When I ask Mr. A if it is possible he is mistaken in his belief that his wife is having affairs, he says, “No, doctor. You don’t know her.” When I ask how he knows she is having affairs, he says that the painter started looking at him “funny” and that the busboy at a restaurant they frequent called his wife “dear.” He believes his wife is having sexual relations with both of these men.

Does Mr. A require a guardian? I opine that Mr. A requires a guardian of estate (to manage his property) but not a guardian of person because he is capable of making decisions about his medical care and other personal decisions. He is failing to care for his wife because of his delusional jealousy. Although cognitively intact, he is unable to appreciate the consequences of his actions or rationally manipulate information because of his delusional thinking. He believes he is “cutting off” an unfaithful spouse when, in fact, there is no evidence that she has been unfaithful. His inability to rationally manipulate information is demonstrated by the fact that he uses innocuous facts such as a busboy calling his elderly wife “dear” to support his delusion that she was having affairs.

I note that his psychosis is reversible because it is likely due to his antiparkinsonian regimen. However, he declines both a dose reduction in his medication and antipsychotic treatment. I note that should his psychosis resolve, he may regain financial decision-making capacity.

 

 

Related Resources

Disclosure

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

Acknowledgement

The author thanks forensic psychiatry fellow Abhishek Jain, MD for reviewing the article and offering editorial suggestions.

Discuss this article at www.facebook.com/CurrentPsychiatry

Although forensic psychiatrists typically are consulted in complex legal matters, geriatric, consultation-liaison, and general psychiatrists are on the front lines of assessing capacity to give informed consent and need for guardianship. Psychiatrists often find such consultations daunting because residency training usually includes little to no formal training in performing psycho-legal assessments. Evaluating issues such as decision-making capacity, guardianship, and capacity to give informed consent requires a delicate balance between autonomy and beneficence. This article reviews 4 common legal issues in geriatric consultation—capacity evaluations, informed consent, guardianship, and elder abuse—and suggests a systematic approach to psycho-legal consultations in older adults.

Confidentiality and dual agency

Every psychiatrist should be familiar with basic principles of medical ethics as well as key aspects of local mental health law. Relevant ethical principles include autonomy, beneficence, confidentiality, and dual agency. A review of all these ethical issues is beyond the scope of this article, so here I highlight confidentiality and dual agency.

Confidentiality—the clinician’s obligation not to disclose private medical information—is a legal as well as an ethical requirement. A psychiatrist who performs a psycho-legal evaluation must disclose to the patient the purpose of the evaluation, that a report will be prepared, and to whom it will be submitted. Exceptions to confidentiality include medical emergencies, mandatory reporting of abuse and infectious diseases, and the duty to protect (warning police and the intended victim when a patient makes a threat).

Dual agency or dual role refers to serving as both a treating physician and a forensic evaluator. Although it is ideal to avoid serving in a dual role, sometimes it is impractical or impossible to avoid doing so, such as in guardianship or civil commitment evaluations, or in state forensic hospitals. In such cases, the psychiatrist must be aware of potential conflicts between clinical and forensic evaluations. A treating psychiatrist primarily serves his or her patient’s best interest, whereas a forensic psychiatrist primarily seeks truth.1 A treating psychiatrist is at risk of consciously or unconsciously biasing his or her psycho-legal evaluation in favor of or against the patient/litigant, depending upon the psychiatrist’s countertransference. Further, performing a psycho-legal evaluation can cause problems in ongoing treatment. A psychiatrist who testifies that his or her fiercely independent patient needs a guardian or nursing home placement will experience significant challenges in continuing to work with that patient.

4 common issues for older adults

Decision-making capacity. Although “capacity” and “competence” often are used interchangeably, “capacity” broadly refers to the ability to perform a specific task, whereas “competence” refers to the legally defined standard for performing a specific task such as making a will. “Competence” is legally determined, whereas “capacity” may be determined clinically.

Capacity usually is task-specific rather than a general construct. The existence of physical or mental illness per se does not mean that a patient lacks capacity. Rather, capacity is determined by whether an individual has specific abilities, regardless of diagnosis. Specific capacities include the ability to give informed consent, manage finances, make a will, or enter into contracts (Table 1).2-4 Appelbaum and Gutheil describe 4 components for assessing specific capacity:

  • communication of a choice
  • factual understanding of the issues
  • appreciation of the situation and its consequences
  • rational manipulation of information.5,6

Table 1

Criteria of 3 specific capacities

CapacityCriteria
Capacity to give informed consentUnderstand nature of illness and treatment
Understand risks and benefits of treatment
Understand treatment alternatives
Understand risk of refusing treatment
Testamentary capacityUnderstand that he/she is making a will
Know the nature and extent of their property
Understand the “natural objects” of their bounty and their claims upon them
Contractual capacityUnderstand the transaction
Act in a reasonable manner
Source: References 2-4

Ability to communicate a choice refers to a patient’s ability to express his or her wishes in a reasonably stable manner. Factual understanding of the issues refers to an individual’s ability to understand the relevant facts before making a decision. Appreciation of the situation and its consequences refers to a person’s ability to rationally understand the effect of decisions. Appreciation is a higher level of understanding than mere factual understanding—eg, a delusional patient who believes himself immortal may intellectually understand that a surgical procedure carries a 50% mortality risk, but may be unable to appreciate the information as it relates to him because he believes he is immortal. Rational manipulation of information refers to a patient’s reasoning process and how the patient integrates data into his or her decision-making process.5

 

 

Informed consent. In my experience, capacity to give informed consent is the most commonly requested specific capacity assessment in general medical settings. Informed consent must be knowing, voluntary, and competent. All material information—information that would cause a reasonable person to accept or reject a proposed treatment—should be communicated to the patient. Informed consent requires an understanding of the patient’s condition and indication for treatment, risks and benefits of and alternatives to treatment, and risks of declining treatment.2,3 Exceptions to informed consent include incompetence, medical emergencies, patient waiver of informed consent, and a limited therapeutic privilege (when a physician determines that the information would harm the patient).3

Several instruments can help clinicians assess patients’ capacity to give informed consent. The benefits of using a structured instrument include:

  • ensuring that specific information is covered during each evaluation
  • systematically recording a patient’s response.5

Disadvantages of using instruments include the fact that no instrument can take into account all aspects of a particular case, and some instruments are time-consuming and require training. Structured instruments can be a useful adjunct to the clinical interview in some cases, but should not substitute for it. In a review of 23 instruments for assessing decisional capacity to consent to treatment or clinical research, the MacArthur Competence Assessment Tool for Clinical Research and the MacArthur Competence Assessment Tool for Treatment had the most empirical support, although the authors noted that other instruments might be better suited to specific situations.7

Psychiatrists may be consulted when a patient refuses treatment or decides to leave the hospital against medical advice. The key issue in both situations is whether the patient has capacity to refuse treatment.8 If there is evidence that the patient is mentally ill and poses an imminent risk of suicide or violence or is unable to provide for his or her basic needs, the psychiatrist should assess whether the patient meets criteria for civil commitment.

Many clinicians employ a “sliding scale” approach to competence, requiring a lower degree of competence for consenting to low-risk, high-benefit interventions and a greater degree of competence for higher-risk procedures.5,9 Family members often serve as informal surrogate decision makers for incapacitated patients, except when there is significant family discord or no family members are available.5

Guardianship. Guardians are appointed by courts to make decisions for individuals who have been found incompetent (wards). Although its purposes are beneficent, the guardianship system could do significant harm.10 Determining that an individual is incompetent is tantamount to depriving him or her of basic personhood. In many cases, the ward loses the ability to consent to or refuse medical care, manage his or her finances, enter into contracts, marry, and determine where he or she will live. On the other hand, failing to recognize incompetence can leave a vulnerable person in danger of physical deterioration, abuse, neglect, or exploitation.

It is critical that guardianship evaluations be conducted carefully. In a review of 298 guardianship cases from 3 states, Moye and colleagues11 found that the quality of the reports was significantly better in Colorado, a state with guardianship reforms, but documentation of functional strengths and weaknesses was “particularly rare” in guardianship evaluations and prognosis often was not included. This information is relevant to judges, who need to determine which areas of function are impaired and how long the impairment is likely to last.

Guardianship evaluations often focus on general rather than specific capacity. In other words, often there is not a specific task such as consenting to surgery that the alleged incompetent person needs to perform. Rather, the question is whether an individual can manage his or her finances or make treatment decisions in general. Appelbaum and Gutheil suggest considering 6 factors when assessing general capacity:

  • awareness of the situation
  • factual understanding of the issues
  • appreciation of the likely consequences
  • rational manipulation of information
  • functioning in one’s environment
  • extent of demands on patient.5

The first 4 are closely related to the elements of specific capacity described above. Functioning in one’s environment and extent of demands on the patient attempt to anticipate the tasks that an individual will need to perform. A patient with mild dementia may be unable to manage a complex estate but can handle a bank account and a fixed income. Similarly, it is important to consider the patient’s support system. An impaired patient may function adequately with his wife’s help but may lose the capacity to live independently if his wife dies or becomes impaired.

 

 

Traditionally, guardianship has resulted in a complete loss of decision-making ability. Several state legislatures have passed laws allowing for limited guardianship, although orders for limited guardianship remain underutilized.10 Limited guardianship delineates specific areas of incompetence and limits the guardian’s decision-making authority to those areas while leaving intact the ward’s ability to make all other decisions for himself or herself.

The use of less-restrictive alternatives to guardianship—such as powers of attorney, durable powers of attorney, living wills, payees, and trusts—is increasing. A power of attorney allows a patient to authorize a specific individual to act on his or her behalf. The scope of the power of attorney can be limited, such as to manage finances or even to a specific transaction, such as selling a home or car. A durable power of attorney also allows an agent to make decisions on the patient’s behalf but becomes active only when the patient becomes incompetent. It often is used to appoint an individual to make medical decisions on behalf of an incompetent patient. Living wills allow patients to determine what treatment they would like in the event they become incompetent.

Elder abuse. An estimated 1 to 2 million adults age >65 have been abused, exploited, or neglected.12 Elder abuse includes physical abuse, neglect, emotional abuse, sexual abuse, and financial exploitation (including undue influence). Most states have mandatory reporting of elder abuse, although they vary regarding who must report and what the report must entail. Psychiatrists should be vigilant in looking for signs of elder abuse (Table 2),13 regardless of the reason for the consult.

Table 2

Signs of elder abuse: What to wlook for

Type of abuseSigns
PhysicalBruises, burns (especially circular, suggesting cigarette burns), slap marks
SexualUnexplained sexually transmitted diseases, bruises in genital area, breasts, or anal area
EmotionalWithdrawal, new-onset depression
FinancialSudden loss of property, unusual increase in spending, checks paid in large, round numbers, checks marked as gifts or loans
NeglectMalnutrition, lack of medical care, poor hygiene, pressure ulcers
Source: Reference 13

10 tips for thorough evaluations

1. Consider the context of the consultation. This includes medical factors (such as the patient’s condition, prognosis, relationship with the treatment team, and recommended course of treatment), legal factors (eg, pending litigation and relevant legal standards for issues such as guardianship), and psychosocial issues (eg, the patient’s current support structure and family conflicts).

2. Identify the legal issue and any relevant legal standards. The legal standard will inform you of the issues you need to address in the evaluation. If an attorney has consulted you, ask him or her to provide the legal standard.

3. Gather relevant collateral information, which may include interviews with family members or a review of financial or medical records.

4. Explain the purpose of the examination and the limits of confidentiality.

5. Perform a focused psychiatric evaluation, paying special attention to cognitive functioning, reasoning, and unusual thought content such as delusional beliefs.

6. Perform an interview specific to the referral issue.

7. Consider using a relevant assessment instrument.

8. Consider psychological testing, laboratory testing, imaging, or further medical evaluation. These assessments can help determine the diagnosis, the cause of any deficits in capacity, and whether any deficits are reversible.

9. Determine what opinions you are able to render. Limit opinions and remember that it may be appropriate to decline to address certain issues if there is insufficient information or if the issue is outside your area of expertise.

10. Prepare a written report. Consider the audience. Minimize the use of medical jargon and define all medical terms. State your opinions clearly and with reasonable medical certainty (in most jurisdictions, this means more likely than not). State the basis for all opinions.

For a case study that provides an example of a psycho-legal evaluation of a geriatric patient, see the Box.

Box

Case study: Unfaithfully yours?

Mr. A, age 75, recently started taking a dopaminergic agonist to treat Parkinson’s disease. He says he wants to divorce his wife of 35 years because of “scandalous affairs” she allegedly engaged in. His wife reports that he has been accusing her of having affairs with various men, including a man who recently painted their house.

On evaluation, Mr. A’s Mini-Mental State Examination score is 30/30. He has no signs of depression and his sleep patterns have not changed. There have been no changes in his spending patterns, although he no longer gives his wife money for grocery shopping, telling her to get money from her “boyfriends.” He is adamant about this decision, saying, “It’s my money and I can do with it as I please. This is still a free country, isn’t it?”

He says he has $70,000 in his individual retirement account, $20,000 in his bank account, and receives a pension of $1,785 per month. He estimates that his home is worth $200,000. His financial records essentially are consistent with his reports. He is able to perform basic calculations without difficulty and is aware of his monthly expenses. He describes his relationship with his wife by saying, “It was fine until she started screwing around.”

When asked about the likely consequences of his decision, he shrugs and says, “I guess she’ll have to get money from her boyfriends. I don’t really care who she sees as long as they stay away from me.” He denies having thoughts of harming his wife or her alleged “boyfriends.” He recognizes that his wife might divorce him, leaving him alone.

When I ask Mr. A if it is possible he is mistaken in his belief that his wife is having affairs, he says, “No, doctor. You don’t know her.” When I ask how he knows she is having affairs, he says that the painter started looking at him “funny” and that the busboy at a restaurant they frequent called his wife “dear.” He believes his wife is having sexual relations with both of these men.

Does Mr. A require a guardian? I opine that Mr. A requires a guardian of estate (to manage his property) but not a guardian of person because he is capable of making decisions about his medical care and other personal decisions. He is failing to care for his wife because of his delusional jealousy. Although cognitively intact, he is unable to appreciate the consequences of his actions or rationally manipulate information because of his delusional thinking. He believes he is “cutting off” an unfaithful spouse when, in fact, there is no evidence that she has been unfaithful. His inability to rationally manipulate information is demonstrated by the fact that he uses innocuous facts such as a busboy calling his elderly wife “dear” to support his delusion that she was having affairs.

I note that his psychosis is reversible because it is likely due to his antiparkinsonian regimen. However, he declines both a dose reduction in his medication and antipsychotic treatment. I note that should his psychosis resolve, he may regain financial decision-making capacity.

 

 

Related Resources

Disclosure

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

Acknowledgement

The author thanks forensic psychiatry fellow Abhishek Jain, MD for reviewing the article and offering editorial suggestions.

References

1. American Academy of Psychiatry and the Law. Ethics guidelines for the practice of forensic psychiatry. http://www.aapl.org/ethics.htm. Adopted May 2005. Accessed February 24 2012.

2. Blank K. Legal and ethical issues. In: Sadvoy J Jarvik LF, Grossberg GT, et al, eds. Comprehensive textbook of geriatric psychiatry. 3rd ed. New York, NY: American Association of Geriatric Psychiatry; 2004:1183-1206.

3. Schwartz HI, Mack DM. Informed consent and competency. In: Rosner R ed. Principles and practice of forensic psychiatry. 2nd ed. New York, NY: Oxford University Press; 2003:97-106.

4. Ciccone RJ. Civil competencies. In: Rosner R ed. Principles and practice of forensic psychiatry. 2nd ed. New York, NY: Oxford University Press; 2003:308-315.

5. Appelbaum PS, Gutheil TG. Competence and substitute decision making. In: Appelbaum PS Gutheil TG, eds. Clinical handbook of psychiatry and the law. 4th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2007:177-213.

6. Appelbaum PS, Grisso T. Assessing patients’ capacities to consent to treatment. N Engl J Med. 1988;319(25):1635-1638.

7. Dunn LB, Nowrangi MA, Palmer BW, et al. Assessing decisional capacity for clinical research or treatment: a review of instruments. Am J Psychiatry. 2006;163(8):1323-1334.

8. Resnick PJ, Sorrentino R. Forensic issues in consultation-liaison psychiatry. Psychiatric Times. 2005;23(14).-

9. Drane JF. The many faces of competency. Hastings Cent Rep. 1985;15(2):17-21.

10. Wright JL. Guardianship for your own good: improving the well being of respondents and wards in the USA. Int J Law Psychiatry. 2010;33(5-6):350-368.

11. Moye J, Wood S, Edelstein B, et al. Clinical evidence in guardianship of older adults is inadequate: findings from a tri-state study. Gerontologist. 2007;47(5):604-612.

12. National Center on Elder Abuse. Fact sheet: elder abuse prevalence and incidence. http://www.ncea.aoa.gov/NCEAroot/Main_Site/pdf/publication/FinalStatistics050331.pdf. Accessed February 24, 2012.

13. National Center on Elder Abuse. Why should I care about elder abuse? http://www.ncea.aoa.gov/Main_Site/pdf/publication/NCEA_WhatIsAbuse-2010.pdf. Published March 3 2010. Accessed February 24, 2012.

References

1. American Academy of Psychiatry and the Law. Ethics guidelines for the practice of forensic psychiatry. http://www.aapl.org/ethics.htm. Adopted May 2005. Accessed February 24 2012.

2. Blank K. Legal and ethical issues. In: Sadvoy J Jarvik LF, Grossberg GT, et al, eds. Comprehensive textbook of geriatric psychiatry. 3rd ed. New York, NY: American Association of Geriatric Psychiatry; 2004:1183-1206.

3. Schwartz HI, Mack DM. Informed consent and competency. In: Rosner R ed. Principles and practice of forensic psychiatry. 2nd ed. New York, NY: Oxford University Press; 2003:97-106.

4. Ciccone RJ. Civil competencies. In: Rosner R ed. Principles and practice of forensic psychiatry. 2nd ed. New York, NY: Oxford University Press; 2003:308-315.

5. Appelbaum PS, Gutheil TG. Competence and substitute decision making. In: Appelbaum PS Gutheil TG, eds. Clinical handbook of psychiatry and the law. 4th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2007:177-213.

6. Appelbaum PS, Grisso T. Assessing patients’ capacities to consent to treatment. N Engl J Med. 1988;319(25):1635-1638.

7. Dunn LB, Nowrangi MA, Palmer BW, et al. Assessing decisional capacity for clinical research or treatment: a review of instruments. Am J Psychiatry. 2006;163(8):1323-1334.

8. Resnick PJ, Sorrentino R. Forensic issues in consultation-liaison psychiatry. Psychiatric Times. 2005;23(14).-

9. Drane JF. The many faces of competency. Hastings Cent Rep. 1985;15(2):17-21.

10. Wright JL. Guardianship for your own good: improving the well being of respondents and wards in the USA. Int J Law Psychiatry. 2010;33(5-6):350-368.

11. Moye J, Wood S, Edelstein B, et al. Clinical evidence in guardianship of older adults is inadequate: findings from a tri-state study. Gerontologist. 2007;47(5):604-612.

12. National Center on Elder Abuse. Fact sheet: elder abuse prevalence and incidence. http://www.ncea.aoa.gov/NCEAroot/Main_Site/pdf/publication/FinalStatistics050331.pdf. Accessed February 24, 2012.

13. National Center on Elder Abuse. Why should I care about elder abuse? http://www.ncea.aoa.gov/Main_Site/pdf/publication/NCEA_WhatIsAbuse-2010.pdf. Published March 3 2010. Accessed February 24, 2012.

Issue
Current Psychiatry - 11(04)
Issue
Current Psychiatry - 11(04)
Page Number
39-54
Page Number
39-54
Publications
Publications
Article Type
Display Headline
Evaluating older adults’ capacity and need for guardianship
Display Headline
Evaluating older adults’ capacity and need for guardianship
Legacy Keywords
capacity; assessment; geriatrics; legal; decision-making; informed consent; guardianship; elder abuse
Legacy Keywords
capacity; assessment; geriatrics; legal; decision-making; informed consent; guardianship; elder abuse
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Premenstrual dysphoric disorder: How to alleviate her suffering

Article Type
Changed
Tue, 12/11/2018 - 15:04
Display Headline
Premenstrual dysphoric disorder: How to alleviate her suffering

Approximately 75% of women experience a premenstrual change in emotional or physical symptoms commonly referred to as premenstrual syndrome (PMS). These symptoms—including increased irritability, tension, depressed mood, and somatic complaints such as breast tenderness and bloating—often are mild to moderate and cause minimal distress.1 However, approximately 3% to 9% of women experience moderate to severe premenstrual mood symptoms that meet criteria for premenstrual dysphoric disorder (PMDD).2

PMDD includes depressed or labile mood, anxiety, irritability, anger, insomnia, difficulty concentrating, and other symptoms that occur exclusively during the 2 weeks before menses and cause significant deterioration in daily functioning. Women with PMDD use general and mental health services more often than women without the condition.3 They may experience impairment in marital and parental relationships as severe as that experienced by women with recurrent or chronic major depression.2

PMDD often responds to treatment. Unfortunately, many women with PMDD do not seek treatment, and up to 90% may go undiagnosed.4 In this article, we review the prevalence, etiology, diagnosis, and treatment of PMDD.

A complex disorder

A distinguishing characteristic of PMDD is the timing of symptom onset. In women with PMDD, mood symptoms occur only during the luteal phase of the menstrual cycle (ovulation until onset of menses) and resolve after menstruation onset. Women with PMDD report normal mood and functioning during the follicular phase of the menstrual cycle (first day of the menstrual cycle until ovulation).

Although PMS and PMDD criteria share affective and somatic symptoms, more symptoms are required for a PMDD diagnosis, and symptoms often are more severe.5 As defined in DSM-IV-TR (Table),6 PMDD has a broader range of symptoms than PMS and includes symptoms not included in the American College of Obstetrics and Gynecology criteria for PMS,7 such as impaired concentration, appetite, and sleep (hypersomnia or insomnia); and mood lability. PMDD symptoms must occur only during the 2 weeks preceding menses, although on average symptoms last 6 days and severity usually peaks in the 2 days before menses.1 The prevalence of subthreshold PMDD is fairly common; approximately 19% of women will meet some—but not all—DSM-IV-TR criteria for PMDD.3

In a revision proposed for DSM-5, PMDD would be included as a mood disorder, which represents a significant change from DSM-IV-TR, where it is listed in the appendix as “research criteria.”8 In addition, in oral contraceptive users, a PMDD diagnosis should not be made unless the premenstrual symptoms are reported to be present and as severe when the woman is not taking the oral contraceptive.8

Comorbidity with other axis I disorders such as major depressive disorder (MDD), bipolar disorder (BD), and anxiety disorders is high.9-11 Women with an MDD history have the highest correlation with PMDD,9 and worsening premenstrual mood symptoms are more common in women with BD.12 Payne et al11 found that premenstrual symptoms were reported by twice as many women diagnosed with mood disorders (68%) than women without a psychiatric diagnosis (34%). Moreover, 38% to 46% of women with PMDD have comorbid seasonal affective disorder, and 11% to 38% report a comorbid anxiety disorder.12 Women with PMDD and a history of MDD have lower cortisol concentrations than non-PMDD women.10 Although interventions for PMDD and a comorbid axis I disorder may be similar, it is important to consider both when planning treatment.

Abuse, trauma, and PMDD. An association between PMS/PMDD and a history of sexual and physical abuse is well-documented.13 Studies have reported abuse histories among almost 60% of women with PMDD,14 although studies comparing abuse and trauma in PMDD vs non-PMDD women have been small. A recent study found that trauma and posttraumatic stress disorder are independently associated with PMDD and premenstrual symptoms.15

Evidence suggests that a history of abuse is associated with specific biological sequelae in PMDD women, particularly with respect to hypothalamic-pituitary-thyroid axis measures and noradrenergic activity.16-18 Women with PMDD and a history of sexual abuse show:

  • markedly elevated triiodothyronine (T3) concentrations (the more biologically potent thyroid hormone) that appear to result from increased conversion of thyroxine (T4) to T316
  • lower circulating plasma norepinephrine concentrations17
  • greater resting and stress-induced heart rates and systolic blood pressure compared with non-abused PMDD women, an effect that is eliminated by clonidine (an α-2 adrenergic receptor agonist).18

One study showed that PMDD women with abuse histories had higher blood pressure measurements at rest and during stress and exhibited greater vascular tone than non-abused women; these effects were not seen in non-PMDD women with similar abuse histories.14 This body of evidence is consistent with the concept that PMDD is a stress-related disorder,19 and that a history of abuse is prevalent and may identify a clinically distinct subgroup of PMDD women with respect to thyroid axis and adrenergic physiology. Screening PMDD patients for abuse histories may help manage the disorder.

 

 

For a discussion of the etiology of PMDD, see Box 1.

Table

DSM-IV-TR research criteria for PMDD

  1. In most menstrual cycles of the past year, ≥5 of the following symptoms must be present for most of the time during the last week of the luteal phase, begin to remit within a few days after the onset of the follicular phase, and are absent in the week postmenses, with ≥1 of the symptoms being either 1, 2, 3, or 4:
  2. The disturbance markedly interferes with work or school or with usual social activities and relationships with others (eg, avoidance of social activities, decreased productivity and efficiency at work or school)
  3. The disturbance is not merely an exacerbation of the symptoms of another disorder, such as major depressive disorder, panic disorder, dysthymic disorder, or a personality disorder (although it may be superimposed on any of these disorders)
  4. Criteria A, B, and C must be confirmed by prospective daily ratings during ≥2 consecutive symptomatic cycles (The diagnosis may be made provisionally prior to this confirmation)
Source: Reference 6

Box 1

Reproductive hormones in PMDD etiology

Although questions remain about the pathogenesis of premenstrual dysphoric disorder (PMDD), literature documents the role of gonadal steroids (estrogen and progesterone) in the etiology of premenstrual syndrome (PMS)/PMDD and suggests that women with PMDD are differentially sensitive to the normal physiologic fluctuations of gonadal hormones throughout the menstrual cycle.a

The first half of the menstrual cycle—the follicular phase—begins with increasing levels of follicular stimulating hormone (FSH) leading to maturity of the ovarian follicle. Once the follicle is ripe, the luteal phase of the menstrual cycle begins with a surge in luteinizing hormone (LH), which results in ovulation of the mature follicle, followed by increased secretion of progesterone, followed by increased estrogen secretion. The system is regulated via negative feedback, and high levels of progesterone decrease gonadotropin-releasing hormone (GnRH) pulse frequency, which leads to decreased secretion of FSH and LH, and subsequent decline of estrogen and progesterone. If the ovarian follicle is not fertilized, menstruation begins and FSH levels rise again, initiating the follicular phase of the menstrual cycle.

Fluctuations in reproductive steroid levels have been implicated in the etiology of PMDD from studies showing that oophorectomy and ovulation inhibitors (GnRH agonists) relieve symptoms.b Some researchers proposed that symptoms are related to the drop of progesterone in the late luteal phase; however, many women have symptoms that start at ovulation or during the early luteal phase before the fall in progesterone concentrations.c PMS symptoms may occur independently of the mid-to-late luteal phase.d Because production of gonadal steroids does not differ between women with or without PMS or PMDD,e it may be that follicular or periovulatory changes in levels of estradiol or progesterone secretion trigger symptoms of PMDD in susceptible women, while women without PMDD appear to be immune to these effects of gonadal steroids. This idea is supported by a study showing that pharmacologic induction of a hypogonadal state eliminates symptoms in most women with severe PMS, while “adding back” estrogen or progesterone within the context of hypogonadism elicits return of PMS symptoms in those with PMS but not in controls.a

Abnormalities in serotonin levels also may contribute to PMDD.f In 1 study, a serotonin receptor antagonist precipitated return of symptoms within 24 hours of administration in women with PMDD but not in controls.g PMDD symptoms also can be evoked by depleting the serotonin precursor tryptophan.h When women with PMDD received paroxetine at different phases of their menstrual cycle, they showed fluctuations in serotonergic function across their cycles; these fluctuations were not seen in controls.i Other neurotransmitters implicated in PMDD include γ-aminobutyric acid (GABA),j glutamate,k lower levels of cortisol and beta-endorphins,l and an abnormal stress response.m

Other studies have focused on differing concentrations of luteal phase hormonesn and gene associations. Two studies suggested that PMDD is heritableo,p and other studies have looked at the association between specific psychological traits that are more prominent in PMDD and single nucleotide polymorphisms in the estrogen receptor alpha gene.q,r

Thyroid hormones also may play a role in the etiology of PMS/PMDD. Thyroid function tests have shown greater variability in women with PMS vs controls,s although this variability appears to be limited to women with a sexual abuse history.t Other studies have evaluated hormones regulated across the circadian and sleep-wake cycles, including melatonin, cortisol, thyroid-stimulating hormone, and prolactin, which suggests that although levels of these hormones may not differ between women with PMDD and controls, the timing of their excretion may vary.s Additionally, women with PMDD are characterized by prefrontal brain asymmetry on electroencephalography that also is evident in patients with major depressive disorder.u

There also may be dysregulation of allopregnanolone (ALLO) in women with PMDD.v,w ALLO is a metabolite of progesterone that is a neurosteroid produced in the brain as well as in the ovary and adrenals.v It produces anxiolytic effects by acting as a modulator of GABA receptors.x In PMDD, ALLO levels may influence the severity of premenstrual symptoms.w

References

  1. Schmidt PJ, Nieman LK, Danaceau MA, et al. Differential behavioral effects of gonadal steroids in women with and in those without premenstrual syndrome. N Engl J Med. 1998;338(4):209-216.
  2. Muse KN, Cetel NS, Futterman LA, et al. The premenstrual syndrome. Effects of “medical ovariectomy.” N Engl J Med. 1984;311(21):1345-1349.
  3. Yonkers KA, O’Brien PM, Eriksson E. Premenstrual syndrome. Lancet. 2008;371(9619):1200-1210.
  4. Schmidt PJ, Nieman LK, Grover GN, et al. Lack of effect of induced menses on symptoms in women with premenstrual syndrome. N Engl J Med. 1991;324(17):1174-1179.
  5. Rubinow DR, Schmidt PJ. The neuroendocrinology of menstrual cycle mood disorders. Ann N Y Acad Sci. 1995;771:648-659.
  6. Steiner M, Pearlstein T. Premenstrual dysphoria and the serotonin system: pathophysiology and treatment. J Clin Psychiatry. 2000;61(suppl 12):17-21.
  7. Roca CA, Schmidt PJ, Smith MJ, et al. Effects of metergoline on symptoms in women with premenstrual dysphoric disorder. Am J Psychiatry. 2002;159(11):1876-1881.
  8. Menkes DB, Coates DC, Fawcett JP. Acute tryptophan depletion aggravates premenstrual syndrome. J Affect Disord. 1994;32(1):37-44.
  9. Inoue Y, Terao T, Iwata N, et al. Fluctuating serotonergic function in premenstrual dysphoric disorder and premenstrual syndrome: findings from neuroendocrine challenge tests. Psychopharmacology (Berl). 2007;190(2):213-219.
  10. Epperson CN, Haga K, Mason GF, et al. Cortical gamma-aminobutyric acid levels across the menstrual cycle in healthy women and those with premenstrual dysphoric disorder: a proton magnetic resonance spectroscopy study. Arch Gen Psychiatry. 2002;59(9):851-858.
  11. Batra NA, Seres-Mailo J, Hanstock C, et al. Proton magnetic resonance spectroscopy measurement of brain glutamate levels in premenstrual dysphoric disorder. Biol Psychiatry. 2008;63(12):1178-1184.
  12. Straneva PA, Maixner W, Light KC, et al. Menstrual cycle, beta-endorphins, and pain sensitivity in premenstrual dysphoric disorder. Health Psychol. 2002;21(4):358-367.
  13. Epperson CN, Pittman B, Czarkowski KA, et al. Luteal-phase accentuation of acoustic startle response in women with premenstrual dysphoric disorder. Neuropsychopharmacology. 2007;32(10):2190-2198.
  14. Thys-Jacobs S, McMahon D, Bilezikian JP. Differences in free estradiol and sex hormone-binding globulin in women with and without premenstrual dysphoric disorder. J Clin Endocrinol Metab. 2008;93(1):96-102.
  15. Payne JL, Klein SR, Zamoiski RB, et al. Premenstrual mood symptoms: study of familiality and personality correlates in mood disorder pedigrees. Arch Womens Ment Health. 2009;12(1):27-34.
  16. Kendler KS, Karkowski LM, Corey LA, et al. Longitudinal population-based twin study of retrospectively reported premenstrual symptoms and lifetime major depression. Am J Psychiatry. 1998;155(9):1234-1240.
  17. Miller A, Vo H, Huo L, et al. Estrogen receptor alpha (ESR-1) associations with psychological traits in women with PMDD and controls. J Psychiatr Res. 2010;44(12):788-794.
  18. Huo L, Straub RE, Roca C, et al. Risk for premenstrual dysphoric disorder is associated with genetic variation in ESR1, the estrogen receptor alpha gene. Biol Psychiatry. 2007;62(8):925-933.
  19. Girdler SS, Pedersen CA, Light KC. Thyroid axis function during the menstrual cycle in women with premenstrual syndrome. Psychoneuroendocrinology. 1995;20(4):395-403.
  20. Girdler SS, Thompson KS, Light KC, et al. Historical sexual abuse and current thyroid axis profiles in women with premenstrual dysphoric disorder. Psychosom Med. 2004;66(3):403-410.
  21. Accortt EE, Stewart JL, Coan JA, et al. Prefrontal brain asymmetry and pre-menstrual dysphoric disorder symptomatology. J Affect Disord. 2011;128(1-2):178-183.
  22. Paul SM, Purdy RH. Neuroactive steroids. FASEB J. 1992;6(6):2311-2322.
  23. Girdler SS, Straneva PA, Light KC, et al. Allopregnanolone levels and reactivity to mental stress in premenstrual dysphoric disorder. Biol Psychiatry. 2001;49(9):788-797.
  24. Brot MD, Akwa Y, Purdy RH, et al. The anxiolytic-like effects of the neurosteroid allopregnanolone: interactions with GABA(A) receptors. Eur J Pharmacol. 1997;325(1):1-7.
 

 

Mood charting aids diagnosis

A PMDD diagnosis requires prospective daily monitoring of symptoms for ≥2 consecutive months. Because only 25% to 35% of women who present with PMDD meet diagnostic criteria when prospective daily monitoring is used,20 it is important for patients to keep a daily diary of PMDD symptoms to distinguish the disorder from PMS (Box 2). The Prospective Record of the Impact and Severity of Premenstrual Symptoms calendar and the Daily Record of Severity of Problems (DRSPP)21 may help make the diagnosis.

The widely used DRSPP allows clinicians to quantify the severity of physical, emotional, and behavioral symptoms and may be the easiest to use in clinical practice because it creates a graphic representation of cyclical symptom changes. The DRSPP includes all PMDD symptoms and severity ratings21 and is recognized as a valid instrument for diagnosing PMDD. Another option is a revised visual analogue scale. Lastly, a new revised Premenstrual Tension Syndrome (PMTS) rating scale, which combines the PMTS Observer rating scale plus multiple visual analogue scales, shows promise as a tool to assess PMDD symptoms.

Box 2

PMS or PMDD? Charting symptoms over menstrual cycles

To distinguish premenstrual syndrome (PMS) from premenstrual dysphoric disorder (PMDD), premenstrual exacerbation of an underlying psychiatric disorder, general medical conditions, or other disorders with no association to the menstrual cycle, it is necessary to have patients conduct daily symptom charting over 2 menstrual cycles. This charting should include documentation of emotional, behavioral, and physical symptoms. PMDD can be differentiated from PMS by the severity and number of symptoms. In PMDD, 1 of the symptoms must be a mood disturbance (depressed, anxious, labile, and/or irritable). For a sample form used for PMDD charting, the Daily Record of Severity of Problems, see http://pmdd.factsforhealth.org/drsp/drsp_month.pdf.

Treatment options

Hormonal interventions. Attempts to treat PMS with progesterone during the luteal phase have been largely unsuccessful, although progesterone is approved to treat PMS in the United Kingdom. Long-acting gonadotropin-releasing hormone (GnRH) agonists are effective but result in medical menopause with its accompanying symptoms, which puts women at risk for osteoporosis.22 Approximately 60% to 70% of women with PMDD respond to leuprolide (a GnRH agonist), but it is difficult to predict who will respond; daily mood self-ratings of sadness, anxiety, and irritability predict a positive response to leuprolide with high probability.23 Side effects of GnRH agonists (hot flashes, night sweats, vaginal dryness, etc.) can be tempered by “adding back” some estrogen with a hormonal agent with progestational activity to reduce the risks of unopposed estrogen (ie, endometrial hyperplasia).24

Surgical bilateral oophorectomy is effective but extremely invasive, especially in younger women in whom removal of ovaries generally is inadvisable. Patients should receive a trial of a GnRH agonist before a surgical intervention, because oophorectomy may not reduce symptoms and is irreversible. Oophorectomy also would require hormone replacement therapy.

High-dose estrogen as transdermal patches or subcutaneous implants to inhibit ovulation is effective, but because of the risks of unopposed estrogen, a progestin would be needed. Risks of estrogen therapy (alone and in combination with progestins) include increased risk of endometrial cancer, coronary heart disease, breast cancer, stroke, and pulmonary embolism.25 Danazol, a synthetic androgen and gonadotropin inhibitor, effectively blocks ovulation, but side effects include hirsutism and possible teratogenicity.26 Although these hormonal manipulations may effectively treat PMDD, none are considered practical.

The use of combined oral contraceptives (estrogen and progestin) is common. Although continuous cycle oral contraceptives often are recommended for PMDD, limited evidence supports their use; studies have been mostly negative.27,28 A recent review of 4 studies of a continuous oral contraceptive (levonorgestrel 90 mcg/ethinyl estradiol 20 mcg) for PMDD and PMS had more promising results, although the results were highly variable among studies and a large placebo effect was observed.29

A combination oral contraceptive, drospirenone/ethinyl estradiol, is FDA-approved for treating PMDD in women seeking hormonal contraception because it has shown efficacy compared with placebo, with reported improvements in perceived productivity, social activities, and interpersonal relationships.30 The nature of hormone delivery (ie, a reduction in the pill-free interval from 7 to 4 days) in drospirenone/ethinyl estradiol may contribute to its efficacy because a reduced pill-free interval minimizes the degree of follicular recruitment and subsequent estrogen production and cyclicity seen with standard oral contraceptive.31

 

 

Antidepressants have been shown to effectively ameliorate affective and physical symptoms and improve quality of life and psychosocial function in patients with PMS and PMDD. The response rates for selective serotonin reuptake inhibitors (SSRIs) in PMDD treatment vary from 60% to 90%, vs 30% to 40% for placebo.32 A 2009 Cochrane review found SSRIs reduced premenstrual symptoms compared with placebo.33 However, a literature review suggested that the percentage of women with PMDD who respond to SSRIs or continuous oral contraceptives is lower than the percentage of women who do not respond at all, once the placebo effect is taken into account, and that approximately 40% of women with PMDD do not respond to SSRIs.34 A small study found that citalopram may be effective for women with PMDD who did not respond to a prior SSRI.35

However, only antidepressants that affect serotonergic—not noradrenergic—transmission are effective in PMDD.22 These include:

  • the tricyclic antidepressant clomipramine
  • the SSRIs citalopram, escitalopram, fluoxetine, paroxetine, and sertraline
  • the serotonin-noradrenergic reuptake inhibitor venlafaxine.

It appears that in PMDD, serotonergic agents play a role other than their antidepressant effect.36 The effect of these agents is rapid in PMS/PMDD; women with PMDD who take antidepressants often experience reduced symptoms within the first menstrual cycle, whereas in MDD the onset of action can take weeks or months.37

Although why onset of antidepressant action is quick in PMDD is unclear, rapid onset allows for several dosing options. Some women prefer continuous dosing throughout the month because they do not have to keep track of ovulation. Dosing antidepressants only in the luteal phase (taking the antidepressant from ovulation onset to the start of menses) is an effective treatment strategy.38 Many women prefer to take medication for only 2 weeks per month, which can decrease side effects and lower treatment costs. Alternatively, symptom-onset dosing—initiating the antidepressant when PMDD symptoms begin and stopping at menses onset or within 3 days thereafter—has shown promising results.39,40 Paroxetine, sertraline, and fluoxetine are FDA-approved for PMDD as continuous or intermittent regimens, although using fluoxetine intermittently may not make sense because its biologically active metabolite has an extended half-life.37

Other treatments. Dietary interventions, psychotherapy, vitamins, bright light treatment, and spironolactone have been assessed for PMS/PMDD, although for many evidence-based findings are lacking (Box 3).

See the Bibliography below for studies that support using antidepressants to treat PMDD

Box 3

Beyond hormones and antidepressants: Other treatments for PMDD

Two reviews of 10 randomized controlled trials (RCTs) that evaluated 62 herbs, vitamins, and mineral treatments for premenstrual symptoms found efficacy for chasteberry (Vitex agnus-castus), calcium, and vitamin B6 but not for primrose oil, magnesium oxide, or St. John’s wort.a,b A study comparing fluoxetine with chasteberry found a similar percentage of patients responded to either agent (68% vs 58%, respectively).c Another study showed calcium resulted in a 48% reduction in premenstrual symptoms from baseline, compared with a 30% reduction with placebo.d Bright light treatment significantly reduced depression ratings in women with premenstrual dysphoric disorder (PMDD).e Compared with placebo, the aldosterone antagonist spironolactone improved irritability, depression, feelings of swelling, breast tenderness, and food craving in women with premenstrual syndrome (PMS).f

A recent systematic review of 7 trials of cognitive-behavioral therapy (CBT) for PMDD, including 3 RCTs, showed a lack of a statistically significant effect.g However, a separate review of RCTs of alternative treatments for PMDD—5 of which included CBT—suggested that CBT may be beneficial in reducing premenstrual symptoms, but the evidence was low quality.h

References

  1. Dante G, Facchinetti F. Herbal treatments for alleviating premenstrual symptoms: a systematic review. J Psychosom Obstet Gynaecol. 2011;32(1):42-51.
  2. Whelan AM, Jurgens TM, Naylor H. Herbs, vitamins and minerals in the treatment of premenstrual syndrome: a systematic review. Can J Clin Pharmacol. 2009;16(3):e407-e429.
  3. Atmaca M, Kumru S, Tezcan E. Fluoxetine versus Vitex agnus castus extract in the treatment of premenstrual dysphoric disorder. Hum Psychopharmacol. 2003;18(3):191-195.
  4. Thys-Jacobs S, Starkey P, Bernstein D, et al. Calcium carbonate and the premenstrual syndrome: effects on premenstrual and menstrual symptoms. Premenstrual Syndrome Study Group. Am J Obstet Gynecol. 1998;179(2):444-452.
  5. Parry BL, Berga SL, Mostofi N, et al. Morning versus evening bright light treatment of late luteal phase dysphoric disorder. Am J Psychiatry. 1989;146(9):1215-1217.
  6. Wang M, Hammarbäck S, Lindhe BA, et al. Treatment of premenstrual syndrome by spironolactone: a double-blind, placebo-controlled study. Acta Obstet Gynecol Scand. 1995;74(10):803-808.
  7. Lustyk MK, Gerrish WG, Shaver S, et al. Cognitive-behavioral therapy for premenstrual syndrome and premenstrual dysphoric disorder: a systematic review. Arch Womens Ment Health. 2009;12(2):85-96.
  8. Busse JW, Montori VM, Krasnik C, et al. Psychological intervention for premenstrual syndrome: a meta-analysis of randomized controlled trials. Psychother Psychosom. 2009;78(1):6-15.
 

 


Bibliography

Evidence supporting antidepressants as treatment for PMDD

Cohen LS, Miner C, Brown EW, et al. Premenstrual daily fluoxetine for premenstrual dysphoric disorder: a placebo-controlled, clinical trial using computerized diaries. Obstet Gynecol. 2002;100(3):435-444.

Eriksson E, Ekman A, Sinclair S, et al. Escitalopram administered in the luteal phase exerts a marked and dose-dependent effect in premenstrual dysphoric disorder. J Clin Psychopharmacol. 2008;28(2):195-202.

Eriksson E, Hedberg MA, Andersch B, et al. The serotonin reuptake inhibitor paroxetin is superior to the noradrenaline reuptake inhibitor maprotiline in the treatment of premenstrual syndrome. Neuropsychopharmacology. 1995;12(2):167-176.

Menkes DB, Taghavi E, Mason PA, et al. Fluoxetine treatment of severe premenstrual syndrome. BMJ. 1992;305(6849):346-347.

Miner C, Brown E, McCray S, et al. Weekly luteal-phase dosing with enteric-coated fluoxetine 90 mg in premenstrual dysphoric disorder: a randomized, double-blind, placebo-controlled clinical trial. Clin Ther. 2002;24(3):417-433.

Ozeren S, Corakçi A, Yücesoy I, et al. Fluoxetine in the treatment of premenstrual syndrome. Eur J Obstet Gynecol Reprod Biol. 1997;73(2):167-170.

Pearlstein TB, Stone AB, Lund SA, et al. Comparison of fluoxetine, bupropion, and placebo in the treatment of premenstrual dysphoric disorder. J Clin Psychopharmacol. 1997;17(4):261-266.

Ravindran LN, Woods SA, Steiner M, et al. Symptom-onset dosing with citalopram in the treatment of premenstrual dysphoric disorder (PMDD): a case series. Arch Womens Ment Health. 2007;10(3):125-127.

Steiner M, Brown E, Trzepacz P, et al. Fluoxetine improves functional work capacity in women with premenstrual dysphoric disorder. Arch Womens Ment Health. 2003;6(1):71-77.

Stone AB, Pearlstein TB, Brown WA. Fluoxetine in the treatment of late luteal phase dysphoric disorder. J Clin Psychiatry. 1991;52(7):290-293.

Sundblad C, Hedberg MA, Eriksson E. Clomipramine administered during the luteal phase reduces the symptoms of premenstrual syndrome: a placebo-controlled trial. Neuropsychopharmacology. 1993;9(2):133-145.

Sundblad C, Modigh K, Andersch B, et al. Clomipramine effectively reduces premenstrual irritability and dysphoria: a placebo-controlled trial. Acta Psychiatr Scand. 1992;85(1):39-47.

Su TP, Schmidt PJ, Danaceau MA, et al. Fluoxetine in the treatment of premenstrual dysphoria. Neuropsychopharmacology. 1997; 16(5):346-356.

Wikander I, Sundblad C, Andersch B, et al. Citalopram in premenstrual dysphoria: is intermittent treatment during luteal phases more effective than continuous medication throughout the menstrual cycle? J Clin Psychopharmacol. 1998;18(5):390-398.

Treatment selection

To enhance compliance and improve the likelihood of successful treatment, tailor treatment decisions to your patient’s needs. Carefully discuss with your patient the evidence-based literature to select the best option for her. Factors to consider when counseling patients include:

  • the patient’s age, cigarette smoking habits, and body mass index, which may contraindicate oral contraceptives
  • does the patient have regular cycle lengths?
  • can she adhere to an on-off schedule? If so, intermittent SSRI dosing may be a good treatment option
  • does the patient have irregular cycles?
  • is there evidence that symptoms persist into the follicular phase, albeit at a lower level? If so, continuous SSRI dosing may be the best option.

Related Resources

Drug Brand Names

  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Clonidine • Catapres, others
  • Danazol • Danocrine
  • Drospirenone/ethinyl estradiol • Yaz
  • Escitalopram • Lexapro
  • Fluoxetine • Prozac
  • Leuprolide • Lupron
  • Levonorgestrel/ethinyl estradiol • Seasonale, others
  • Paroxetine • Paxil
  • Progesterone • Prometrium
  • Sertraline • Zoloft
  • Spironolactone • Aldactone
  • Venlafaxine • Effexor

Disclosures

Drs. Wakil and Girdler report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products

Dr. Meltzer-Brody receives research/grant support from AstraZeneca, The Foundation of Hope, and the National Institutes of Health.

References

1. Yonkers KA, O’Brien PM, Eriksson E. Premenstrual syndrome. Lancet. 2008;371(9619):1200-1210.

2. Halbreich U, Borenstein J, Pearlstein T, et al. The prevalence, impairment, impact and burden of premenstrual dysphoric disorder (PMS/PMDD). Psychoneuroendocrinology. 2008;28(suppl 3):1-23.

3. Wittchen HU, Becker E, Lieb R, et al. Prevalence, incidence and stability of premenstrual dysphoric disorder in the community. Psychol Med. 2002;32(1):119-132.

4. Hylan TR, Sundell K, Judge R. The impact of premenstrual symptomatology on functioning and treatment-seeking behavior: experience from the United States United Kingdom, and France. J Womens Health Gend Based Med. 1999;8(8):1043-1052.

5. Biggs WS, Demuth RH. Premenstrual syndrome and premenstrual dysphoric disorder. Am Fam Physician. 2011;84(8):918-924.

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

7. ACOG Practice Bulletin. Clinical management guidelines for obstetrician-gynecologists. Number 15 April 2000. Premenstrual syndrome. Obstet Gynecol. 2000;95:1-9.

8. American Psychiatric Association. Proposed draft revisions to DSM disorders and criteria. DSM-5 development. http://www.dsm5.org/proposedrevision/Pages/Default.aspx. Accessed February 23 2012.

9. Yonkers KA. The association between premenstrual dysphoric disorder and other mood disorders. J Clin Psychiatry. 1997;58(suppl 15):19-25.

10. Klatzkin RR, Lindgren ME, Forneris CA, et al. Histories of major depression and premenstrual dysphoric disorder: evidence for phenotypic differences. Biol Psychol. 2010;84(2):235-247.

11. Payne JL, Roy PS, Murphy-Eberenz K, et al. Reproductive cycle-associated mood symptoms in women with major depression and bipolar disorder. J Affect Disord. 2007;99(1-3):221-229.

12. Kim DR, Gyulai L, Freeman EW, et al. Premenstrual dysphoric disorder and psychiatric co-morbidity. Arch Womens Ment Health. 2004;7(1):37-47.

13. Golding JM, Taylor DL, Menard L, et al. Prevalence of sexual abuse history in a sample of women seeking treatment for premenstrual syndrome. J Psychosom Obstet Gynaecol. 2000;21(2):69-80.

14. Girdler SS, Leserman J, Bunevicius R, et al. Persistent alterations in biological profiles in women with abuse histories: influence of premenstrual dysphoric disorder. Health Psychol. 2007;26(2):201-213.

15. Pilver CE, Levy BR, Libby DJ, et al. Posttraumatic stress disorder and trauma characteristics are correlates of premenstrual dysphoric disorder. Arch Womens Ment Health. 2011;14(5):383-393.

16. Girdler SS, Thompson KS, Light KC, et al. Historical sexual abuse and current thyroid axis profiles in women with premenstrual dysphoric disorder. Psychosom Med. 2004;66(3):403-410.

17. Girdler SS, Sherwood A, Hinderliter AL, et al. Biological correlates of abuse in women with premenstrual dysphoric disorder and healthy controls. Psychosom Med. 2003;65(5):849-856.

18. Bunevicius R, Hinderliter AL, Light KC, et al. Histories of sexual abuse are associated with differential effects of clonidine on autonomic function in women with premenstrual dysphoric disorder. Biol Psychol. 2005;69(3):281-296.

19. Perkonigg A, Yonkers KA, Pfister H, et al. Risk factors for premenstrual dysphoric disorder in a community sample of young women: the role of traumatic events and posttraumatic stress disorder. J Clin Psychiatry. 2004;65(10):1314-1322.

20. Girdler SS, Pedersen CA, Stern RA, et al. Menstrual cycle and premenstrual syndrome: modifiers of cardiovascular reactivity in women. Health Psychol. 1993;12(3):180-192.

21. Endicott J, Nee J, Harrison W. Daily Record of Severity of Problems (DRSP): reliability and validity. Arch Womens Ment Health. 2006;9(1):41-49.

22. Cunningham J, Yonkers KA, O’Brien S, et al. Update on research and treatment of premenstrual dysphoric disorder. Harv Rev Psychiatry. 2009;17(2):120-137.

23. Pincus SM, Alam S, Rubinow DR, et al. Predicting response to leuprolide of women with premenstrual dysphoric disorder by daily mood rating dynamics. J Psychiatr Res. 2011;45(3):386-394.

24. Wyatt KM, Dimmock PW, Ismail KM, et al. The effectiveness of GnRHa with and without ‘add-back’ therapy in treating premenstrual syndrome: a meta analysis. BJOG. 2004;111(6):585-593.

25. Rossouw JE, Anderson GL, Prentice RL, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288(3):321-333.

26. Hahn PM, Van Vugt DA, Reid RL. A randomized placebo-controlled, crossover trial of danazol for the treatment of premenstrual syndrome. Psychoneuroendocrinology. 1995;20(2):193-209.

27. Bancroft J, Rennie D. The impact of oral contraceptives on the experience of perimenstrual mood clumsiness, food craving and other symptoms. J Psychosom Res. 1993;37(2):195-202.

28. Graham CA, Sherwin BB. A prospective treatment study of premenstrual symptoms using a triphasic oral contraceptive. J Psychosom Res. 1992;36(3):257-266.

29. Freeman EW, Halbreich U, Grubb GS, et al. An overview of four studies of a continuous oral contraceptive (levonorgestrel 90 mcg/ethinyl estradiol 20 mcg) on premenstrual dysphoric disorder and premenstrual syndrome [published online ahead of print December 5, 2011]. Contraception. doi: 10.1016/j.contraception.2011.09.010.

30. Lopez LM, Kaptein AA, Helmerhorst FM. Oral contraceptives containing drospirenone for premenstrual syndrome. Cochrane Database Syst Rev. 2009;(2):CD006586.

31. Sullivan H, Furniss H, Spona J, et al. Effect of 21-day and 24-day oral contraceptive regimens containing gestodene (60 microg) andethinyl estradiol (15 microg) on ovarian activity. Fertil Steril. 1999;72(1):115-120.

32. Yonkers KA, Clark RH, Trivedi MH. The psychopharmacological treatment of nonmajor mood disorders. Mod Probl Pharmacopsychiatry. 1997;25:146-166.

33. Brown J, O’Brien PM, Marjoribanks J, et al. Selective serotonin reuptake inhibitors for premenstrual syndrome. Cochrane Database Syst Rev. 2009;(2):CD001396.

34. Halbreich U. Selective serotonin reuptake inhibitors and initial oral contraceptives for the treatment of PMDD: effective but not enough. CNS Spectr. 2008;13(7):566-572.

35. Freeman EW, Jabara S, Sondheimer SJ, et al. Citalopram in PMS patients with prior SSRI treatment failure: a preliminary study. J Womens Health Gend Based Med. 2002;11(5):

459-464.

36. Freeman EW, Rickels K, Sondheimer SJ, et al. Differential response to antidepressants in women with premenstrual syndrome/premenstrual dysphoric disorder: a randomized controlled trial. Arch Gen Psychiatry. 1999;56(10):932-939.

37. Steiner M, Pearlstein T, Cohen LS, et al. Expert guidelines for the treatment of severe PMS, PMDD, and comorbidities: the role of SSRIs. J Womens Health (Larchmt). 2006;15(1):57-69.

38. Freeman EW. Luteal phase administration of agents for the treatment of premenstrual dysphoric disorder. CNS Drugs. 2004;18(7):453-468.

39. Yonkers KA, Holthausen GA, Poschman K, et al. Symptom-onset treatment for women with premenstrual dysphoric disorder. J Clin Psychopharmacol. 2006;26(2):198-202.

40. Freeman EW, Sondheimer SJ, Sammel MD, et al. A preliminary study of luteal phase versus symptom-onset dosing with escitalopram for premenstrual dysphoric disorder. J Clin Psychiatry. 2005;66(6):769-773.

Article PDF
Author and Disclosure Information

Laura Wakil, MD
Third-Year Psychiatry Resident, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC
Samantha Meltzer-Brody, MD, MPH
Director, Perinatal Psychiatry Program, University of North Carolina Center for Women’s Mood Disorders, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC
Susan Girdler, PhD
Director, UNC Stress and Health Research Program, Menstrually Related Mood Disorders Program, University of North Carolina Center for Women’s Mood Disorders, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC

Issue
Current Psychiatry - 11(04)
Publications
Page Number
22-41
Legacy Keywords
Premenstrual dysphoric disorder; PMDD; mood charting; hormonal intervention; antidepressants;
Sections
Author and Disclosure Information

Laura Wakil, MD
Third-Year Psychiatry Resident, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC
Samantha Meltzer-Brody, MD, MPH
Director, Perinatal Psychiatry Program, University of North Carolina Center for Women’s Mood Disorders, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC
Susan Girdler, PhD
Director, UNC Stress and Health Research Program, Menstrually Related Mood Disorders Program, University of North Carolina Center for Women’s Mood Disorders, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC

Author and Disclosure Information

Laura Wakil, MD
Third-Year Psychiatry Resident, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC
Samantha Meltzer-Brody, MD, MPH
Director, Perinatal Psychiatry Program, University of North Carolina Center for Women’s Mood Disorders, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC
Susan Girdler, PhD
Director, UNC Stress and Health Research Program, Menstrually Related Mood Disorders Program, University of North Carolina Center for Women’s Mood Disorders, Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC

Article PDF
Article PDF

Approximately 75% of women experience a premenstrual change in emotional or physical symptoms commonly referred to as premenstrual syndrome (PMS). These symptoms—including increased irritability, tension, depressed mood, and somatic complaints such as breast tenderness and bloating—often are mild to moderate and cause minimal distress.1 However, approximately 3% to 9% of women experience moderate to severe premenstrual mood symptoms that meet criteria for premenstrual dysphoric disorder (PMDD).2

PMDD includes depressed or labile mood, anxiety, irritability, anger, insomnia, difficulty concentrating, and other symptoms that occur exclusively during the 2 weeks before menses and cause significant deterioration in daily functioning. Women with PMDD use general and mental health services more often than women without the condition.3 They may experience impairment in marital and parental relationships as severe as that experienced by women with recurrent or chronic major depression.2

PMDD often responds to treatment. Unfortunately, many women with PMDD do not seek treatment, and up to 90% may go undiagnosed.4 In this article, we review the prevalence, etiology, diagnosis, and treatment of PMDD.

A complex disorder

A distinguishing characteristic of PMDD is the timing of symptom onset. In women with PMDD, mood symptoms occur only during the luteal phase of the menstrual cycle (ovulation until onset of menses) and resolve after menstruation onset. Women with PMDD report normal mood and functioning during the follicular phase of the menstrual cycle (first day of the menstrual cycle until ovulation).

Although PMS and PMDD criteria share affective and somatic symptoms, more symptoms are required for a PMDD diagnosis, and symptoms often are more severe.5 As defined in DSM-IV-TR (Table),6 PMDD has a broader range of symptoms than PMS and includes symptoms not included in the American College of Obstetrics and Gynecology criteria for PMS,7 such as impaired concentration, appetite, and sleep (hypersomnia or insomnia); and mood lability. PMDD symptoms must occur only during the 2 weeks preceding menses, although on average symptoms last 6 days and severity usually peaks in the 2 days before menses.1 The prevalence of subthreshold PMDD is fairly common; approximately 19% of women will meet some—but not all—DSM-IV-TR criteria for PMDD.3

In a revision proposed for DSM-5, PMDD would be included as a mood disorder, which represents a significant change from DSM-IV-TR, where it is listed in the appendix as “research criteria.”8 In addition, in oral contraceptive users, a PMDD diagnosis should not be made unless the premenstrual symptoms are reported to be present and as severe when the woman is not taking the oral contraceptive.8

Comorbidity with other axis I disorders such as major depressive disorder (MDD), bipolar disorder (BD), and anxiety disorders is high.9-11 Women with an MDD history have the highest correlation with PMDD,9 and worsening premenstrual mood symptoms are more common in women with BD.12 Payne et al11 found that premenstrual symptoms were reported by twice as many women diagnosed with mood disorders (68%) than women without a psychiatric diagnosis (34%). Moreover, 38% to 46% of women with PMDD have comorbid seasonal affective disorder, and 11% to 38% report a comorbid anxiety disorder.12 Women with PMDD and a history of MDD have lower cortisol concentrations than non-PMDD women.10 Although interventions for PMDD and a comorbid axis I disorder may be similar, it is important to consider both when planning treatment.

Abuse, trauma, and PMDD. An association between PMS/PMDD and a history of sexual and physical abuse is well-documented.13 Studies have reported abuse histories among almost 60% of women with PMDD,14 although studies comparing abuse and trauma in PMDD vs non-PMDD women have been small. A recent study found that trauma and posttraumatic stress disorder are independently associated with PMDD and premenstrual symptoms.15

Evidence suggests that a history of abuse is associated with specific biological sequelae in PMDD women, particularly with respect to hypothalamic-pituitary-thyroid axis measures and noradrenergic activity.16-18 Women with PMDD and a history of sexual abuse show:

  • markedly elevated triiodothyronine (T3) concentrations (the more biologically potent thyroid hormone) that appear to result from increased conversion of thyroxine (T4) to T316
  • lower circulating plasma norepinephrine concentrations17
  • greater resting and stress-induced heart rates and systolic blood pressure compared with non-abused PMDD women, an effect that is eliminated by clonidine (an α-2 adrenergic receptor agonist).18

One study showed that PMDD women with abuse histories had higher blood pressure measurements at rest and during stress and exhibited greater vascular tone than non-abused women; these effects were not seen in non-PMDD women with similar abuse histories.14 This body of evidence is consistent with the concept that PMDD is a stress-related disorder,19 and that a history of abuse is prevalent and may identify a clinically distinct subgroup of PMDD women with respect to thyroid axis and adrenergic physiology. Screening PMDD patients for abuse histories may help manage the disorder.

 

 

For a discussion of the etiology of PMDD, see Box 1.

Table

DSM-IV-TR research criteria for PMDD

  1. In most menstrual cycles of the past year, ≥5 of the following symptoms must be present for most of the time during the last week of the luteal phase, begin to remit within a few days after the onset of the follicular phase, and are absent in the week postmenses, with ≥1 of the symptoms being either 1, 2, 3, or 4:
  2. The disturbance markedly interferes with work or school or with usual social activities and relationships with others (eg, avoidance of social activities, decreased productivity and efficiency at work or school)
  3. The disturbance is not merely an exacerbation of the symptoms of another disorder, such as major depressive disorder, panic disorder, dysthymic disorder, or a personality disorder (although it may be superimposed on any of these disorders)
  4. Criteria A, B, and C must be confirmed by prospective daily ratings during ≥2 consecutive symptomatic cycles (The diagnosis may be made provisionally prior to this confirmation)
Source: Reference 6

Box 1

Reproductive hormones in PMDD etiology

Although questions remain about the pathogenesis of premenstrual dysphoric disorder (PMDD), literature documents the role of gonadal steroids (estrogen and progesterone) in the etiology of premenstrual syndrome (PMS)/PMDD and suggests that women with PMDD are differentially sensitive to the normal physiologic fluctuations of gonadal hormones throughout the menstrual cycle.a

The first half of the menstrual cycle—the follicular phase—begins with increasing levels of follicular stimulating hormone (FSH) leading to maturity of the ovarian follicle. Once the follicle is ripe, the luteal phase of the menstrual cycle begins with a surge in luteinizing hormone (LH), which results in ovulation of the mature follicle, followed by increased secretion of progesterone, followed by increased estrogen secretion. The system is regulated via negative feedback, and high levels of progesterone decrease gonadotropin-releasing hormone (GnRH) pulse frequency, which leads to decreased secretion of FSH and LH, and subsequent decline of estrogen and progesterone. If the ovarian follicle is not fertilized, menstruation begins and FSH levels rise again, initiating the follicular phase of the menstrual cycle.

Fluctuations in reproductive steroid levels have been implicated in the etiology of PMDD from studies showing that oophorectomy and ovulation inhibitors (GnRH agonists) relieve symptoms.b Some researchers proposed that symptoms are related to the drop of progesterone in the late luteal phase; however, many women have symptoms that start at ovulation or during the early luteal phase before the fall in progesterone concentrations.c PMS symptoms may occur independently of the mid-to-late luteal phase.d Because production of gonadal steroids does not differ between women with or without PMS or PMDD,e it may be that follicular or periovulatory changes in levels of estradiol or progesterone secretion trigger symptoms of PMDD in susceptible women, while women without PMDD appear to be immune to these effects of gonadal steroids. This idea is supported by a study showing that pharmacologic induction of a hypogonadal state eliminates symptoms in most women with severe PMS, while “adding back” estrogen or progesterone within the context of hypogonadism elicits return of PMS symptoms in those with PMS but not in controls.a

Abnormalities in serotonin levels also may contribute to PMDD.f In 1 study, a serotonin receptor antagonist precipitated return of symptoms within 24 hours of administration in women with PMDD but not in controls.g PMDD symptoms also can be evoked by depleting the serotonin precursor tryptophan.h When women with PMDD received paroxetine at different phases of their menstrual cycle, they showed fluctuations in serotonergic function across their cycles; these fluctuations were not seen in controls.i Other neurotransmitters implicated in PMDD include γ-aminobutyric acid (GABA),j glutamate,k lower levels of cortisol and beta-endorphins,l and an abnormal stress response.m

Other studies have focused on differing concentrations of luteal phase hormonesn and gene associations. Two studies suggested that PMDD is heritableo,p and other studies have looked at the association between specific psychological traits that are more prominent in PMDD and single nucleotide polymorphisms in the estrogen receptor alpha gene.q,r

Thyroid hormones also may play a role in the etiology of PMS/PMDD. Thyroid function tests have shown greater variability in women with PMS vs controls,s although this variability appears to be limited to women with a sexual abuse history.t Other studies have evaluated hormones regulated across the circadian and sleep-wake cycles, including melatonin, cortisol, thyroid-stimulating hormone, and prolactin, which suggests that although levels of these hormones may not differ between women with PMDD and controls, the timing of their excretion may vary.s Additionally, women with PMDD are characterized by prefrontal brain asymmetry on electroencephalography that also is evident in patients with major depressive disorder.u

There also may be dysregulation of allopregnanolone (ALLO) in women with PMDD.v,w ALLO is a metabolite of progesterone that is a neurosteroid produced in the brain as well as in the ovary and adrenals.v It produces anxiolytic effects by acting as a modulator of GABA receptors.x In PMDD, ALLO levels may influence the severity of premenstrual symptoms.w

References

  1. Schmidt PJ, Nieman LK, Danaceau MA, et al. Differential behavioral effects of gonadal steroids in women with and in those without premenstrual syndrome. N Engl J Med. 1998;338(4):209-216.
  2. Muse KN, Cetel NS, Futterman LA, et al. The premenstrual syndrome. Effects of “medical ovariectomy.” N Engl J Med. 1984;311(21):1345-1349.
  3. Yonkers KA, O’Brien PM, Eriksson E. Premenstrual syndrome. Lancet. 2008;371(9619):1200-1210.
  4. Schmidt PJ, Nieman LK, Grover GN, et al. Lack of effect of induced menses on symptoms in women with premenstrual syndrome. N Engl J Med. 1991;324(17):1174-1179.
  5. Rubinow DR, Schmidt PJ. The neuroendocrinology of menstrual cycle mood disorders. Ann N Y Acad Sci. 1995;771:648-659.
  6. Steiner M, Pearlstein T. Premenstrual dysphoria and the serotonin system: pathophysiology and treatment. J Clin Psychiatry. 2000;61(suppl 12):17-21.
  7. Roca CA, Schmidt PJ, Smith MJ, et al. Effects of metergoline on symptoms in women with premenstrual dysphoric disorder. Am J Psychiatry. 2002;159(11):1876-1881.
  8. Menkes DB, Coates DC, Fawcett JP. Acute tryptophan depletion aggravates premenstrual syndrome. J Affect Disord. 1994;32(1):37-44.
  9. Inoue Y, Terao T, Iwata N, et al. Fluctuating serotonergic function in premenstrual dysphoric disorder and premenstrual syndrome: findings from neuroendocrine challenge tests. Psychopharmacology (Berl). 2007;190(2):213-219.
  10. Epperson CN, Haga K, Mason GF, et al. Cortical gamma-aminobutyric acid levels across the menstrual cycle in healthy women and those with premenstrual dysphoric disorder: a proton magnetic resonance spectroscopy study. Arch Gen Psychiatry. 2002;59(9):851-858.
  11. Batra NA, Seres-Mailo J, Hanstock C, et al. Proton magnetic resonance spectroscopy measurement of brain glutamate levels in premenstrual dysphoric disorder. Biol Psychiatry. 2008;63(12):1178-1184.
  12. Straneva PA, Maixner W, Light KC, et al. Menstrual cycle, beta-endorphins, and pain sensitivity in premenstrual dysphoric disorder. Health Psychol. 2002;21(4):358-367.
  13. Epperson CN, Pittman B, Czarkowski KA, et al. Luteal-phase accentuation of acoustic startle response in women with premenstrual dysphoric disorder. Neuropsychopharmacology. 2007;32(10):2190-2198.
  14. Thys-Jacobs S, McMahon D, Bilezikian JP. Differences in free estradiol and sex hormone-binding globulin in women with and without premenstrual dysphoric disorder. J Clin Endocrinol Metab. 2008;93(1):96-102.
  15. Payne JL, Klein SR, Zamoiski RB, et al. Premenstrual mood symptoms: study of familiality and personality correlates in mood disorder pedigrees. Arch Womens Ment Health. 2009;12(1):27-34.
  16. Kendler KS, Karkowski LM, Corey LA, et al. Longitudinal population-based twin study of retrospectively reported premenstrual symptoms and lifetime major depression. Am J Psychiatry. 1998;155(9):1234-1240.
  17. Miller A, Vo H, Huo L, et al. Estrogen receptor alpha (ESR-1) associations with psychological traits in women with PMDD and controls. J Psychiatr Res. 2010;44(12):788-794.
  18. Huo L, Straub RE, Roca C, et al. Risk for premenstrual dysphoric disorder is associated with genetic variation in ESR1, the estrogen receptor alpha gene. Biol Psychiatry. 2007;62(8):925-933.
  19. Girdler SS, Pedersen CA, Light KC. Thyroid axis function during the menstrual cycle in women with premenstrual syndrome. Psychoneuroendocrinology. 1995;20(4):395-403.
  20. Girdler SS, Thompson KS, Light KC, et al. Historical sexual abuse and current thyroid axis profiles in women with premenstrual dysphoric disorder. Psychosom Med. 2004;66(3):403-410.
  21. Accortt EE, Stewart JL, Coan JA, et al. Prefrontal brain asymmetry and pre-menstrual dysphoric disorder symptomatology. J Affect Disord. 2011;128(1-2):178-183.
  22. Paul SM, Purdy RH. Neuroactive steroids. FASEB J. 1992;6(6):2311-2322.
  23. Girdler SS, Straneva PA, Light KC, et al. Allopregnanolone levels and reactivity to mental stress in premenstrual dysphoric disorder. Biol Psychiatry. 2001;49(9):788-797.
  24. Brot MD, Akwa Y, Purdy RH, et al. The anxiolytic-like effects of the neurosteroid allopregnanolone: interactions with GABA(A) receptors. Eur J Pharmacol. 1997;325(1):1-7.
 

 

Mood charting aids diagnosis

A PMDD diagnosis requires prospective daily monitoring of symptoms for ≥2 consecutive months. Because only 25% to 35% of women who present with PMDD meet diagnostic criteria when prospective daily monitoring is used,20 it is important for patients to keep a daily diary of PMDD symptoms to distinguish the disorder from PMS (Box 2). The Prospective Record of the Impact and Severity of Premenstrual Symptoms calendar and the Daily Record of Severity of Problems (DRSPP)21 may help make the diagnosis.

The widely used DRSPP allows clinicians to quantify the severity of physical, emotional, and behavioral symptoms and may be the easiest to use in clinical practice because it creates a graphic representation of cyclical symptom changes. The DRSPP includes all PMDD symptoms and severity ratings21 and is recognized as a valid instrument for diagnosing PMDD. Another option is a revised visual analogue scale. Lastly, a new revised Premenstrual Tension Syndrome (PMTS) rating scale, which combines the PMTS Observer rating scale plus multiple visual analogue scales, shows promise as a tool to assess PMDD symptoms.

Box 2

PMS or PMDD? Charting symptoms over menstrual cycles

To distinguish premenstrual syndrome (PMS) from premenstrual dysphoric disorder (PMDD), premenstrual exacerbation of an underlying psychiatric disorder, general medical conditions, or other disorders with no association to the menstrual cycle, it is necessary to have patients conduct daily symptom charting over 2 menstrual cycles. This charting should include documentation of emotional, behavioral, and physical symptoms. PMDD can be differentiated from PMS by the severity and number of symptoms. In PMDD, 1 of the symptoms must be a mood disturbance (depressed, anxious, labile, and/or irritable). For a sample form used for PMDD charting, the Daily Record of Severity of Problems, see http://pmdd.factsforhealth.org/drsp/drsp_month.pdf.

Treatment options

Hormonal interventions. Attempts to treat PMS with progesterone during the luteal phase have been largely unsuccessful, although progesterone is approved to treat PMS in the United Kingdom. Long-acting gonadotropin-releasing hormone (GnRH) agonists are effective but result in medical menopause with its accompanying symptoms, which puts women at risk for osteoporosis.22 Approximately 60% to 70% of women with PMDD respond to leuprolide (a GnRH agonist), but it is difficult to predict who will respond; daily mood self-ratings of sadness, anxiety, and irritability predict a positive response to leuprolide with high probability.23 Side effects of GnRH agonists (hot flashes, night sweats, vaginal dryness, etc.) can be tempered by “adding back” some estrogen with a hormonal agent with progestational activity to reduce the risks of unopposed estrogen (ie, endometrial hyperplasia).24

Surgical bilateral oophorectomy is effective but extremely invasive, especially in younger women in whom removal of ovaries generally is inadvisable. Patients should receive a trial of a GnRH agonist before a surgical intervention, because oophorectomy may not reduce symptoms and is irreversible. Oophorectomy also would require hormone replacement therapy.

High-dose estrogen as transdermal patches or subcutaneous implants to inhibit ovulation is effective, but because of the risks of unopposed estrogen, a progestin would be needed. Risks of estrogen therapy (alone and in combination with progestins) include increased risk of endometrial cancer, coronary heart disease, breast cancer, stroke, and pulmonary embolism.25 Danazol, a synthetic androgen and gonadotropin inhibitor, effectively blocks ovulation, but side effects include hirsutism and possible teratogenicity.26 Although these hormonal manipulations may effectively treat PMDD, none are considered practical.

The use of combined oral contraceptives (estrogen and progestin) is common. Although continuous cycle oral contraceptives often are recommended for PMDD, limited evidence supports their use; studies have been mostly negative.27,28 A recent review of 4 studies of a continuous oral contraceptive (levonorgestrel 90 mcg/ethinyl estradiol 20 mcg) for PMDD and PMS had more promising results, although the results were highly variable among studies and a large placebo effect was observed.29

A combination oral contraceptive, drospirenone/ethinyl estradiol, is FDA-approved for treating PMDD in women seeking hormonal contraception because it has shown efficacy compared with placebo, with reported improvements in perceived productivity, social activities, and interpersonal relationships.30 The nature of hormone delivery (ie, a reduction in the pill-free interval from 7 to 4 days) in drospirenone/ethinyl estradiol may contribute to its efficacy because a reduced pill-free interval minimizes the degree of follicular recruitment and subsequent estrogen production and cyclicity seen with standard oral contraceptive.31

 

 

Antidepressants have been shown to effectively ameliorate affective and physical symptoms and improve quality of life and psychosocial function in patients with PMS and PMDD. The response rates for selective serotonin reuptake inhibitors (SSRIs) in PMDD treatment vary from 60% to 90%, vs 30% to 40% for placebo.32 A 2009 Cochrane review found SSRIs reduced premenstrual symptoms compared with placebo.33 However, a literature review suggested that the percentage of women with PMDD who respond to SSRIs or continuous oral contraceptives is lower than the percentage of women who do not respond at all, once the placebo effect is taken into account, and that approximately 40% of women with PMDD do not respond to SSRIs.34 A small study found that citalopram may be effective for women with PMDD who did not respond to a prior SSRI.35

However, only antidepressants that affect serotonergic—not noradrenergic—transmission are effective in PMDD.22 These include:

  • the tricyclic antidepressant clomipramine
  • the SSRIs citalopram, escitalopram, fluoxetine, paroxetine, and sertraline
  • the serotonin-noradrenergic reuptake inhibitor venlafaxine.

It appears that in PMDD, serotonergic agents play a role other than their antidepressant effect.36 The effect of these agents is rapid in PMS/PMDD; women with PMDD who take antidepressants often experience reduced symptoms within the first menstrual cycle, whereas in MDD the onset of action can take weeks or months.37

Although why onset of antidepressant action is quick in PMDD is unclear, rapid onset allows for several dosing options. Some women prefer continuous dosing throughout the month because they do not have to keep track of ovulation. Dosing antidepressants only in the luteal phase (taking the antidepressant from ovulation onset to the start of menses) is an effective treatment strategy.38 Many women prefer to take medication for only 2 weeks per month, which can decrease side effects and lower treatment costs. Alternatively, symptom-onset dosing—initiating the antidepressant when PMDD symptoms begin and stopping at menses onset or within 3 days thereafter—has shown promising results.39,40 Paroxetine, sertraline, and fluoxetine are FDA-approved for PMDD as continuous or intermittent regimens, although using fluoxetine intermittently may not make sense because its biologically active metabolite has an extended half-life.37

Other treatments. Dietary interventions, psychotherapy, vitamins, bright light treatment, and spironolactone have been assessed for PMS/PMDD, although for many evidence-based findings are lacking (Box 3).

See the Bibliography below for studies that support using antidepressants to treat PMDD

Box 3

Beyond hormones and antidepressants: Other treatments for PMDD

Two reviews of 10 randomized controlled trials (RCTs) that evaluated 62 herbs, vitamins, and mineral treatments for premenstrual symptoms found efficacy for chasteberry (Vitex agnus-castus), calcium, and vitamin B6 but not for primrose oil, magnesium oxide, or St. John’s wort.a,b A study comparing fluoxetine with chasteberry found a similar percentage of patients responded to either agent (68% vs 58%, respectively).c Another study showed calcium resulted in a 48% reduction in premenstrual symptoms from baseline, compared with a 30% reduction with placebo.d Bright light treatment significantly reduced depression ratings in women with premenstrual dysphoric disorder (PMDD).e Compared with placebo, the aldosterone antagonist spironolactone improved irritability, depression, feelings of swelling, breast tenderness, and food craving in women with premenstrual syndrome (PMS).f

A recent systematic review of 7 trials of cognitive-behavioral therapy (CBT) for PMDD, including 3 RCTs, showed a lack of a statistically significant effect.g However, a separate review of RCTs of alternative treatments for PMDD—5 of which included CBT—suggested that CBT may be beneficial in reducing premenstrual symptoms, but the evidence was low quality.h

References

  1. Dante G, Facchinetti F. Herbal treatments for alleviating premenstrual symptoms: a systematic review. J Psychosom Obstet Gynaecol. 2011;32(1):42-51.
  2. Whelan AM, Jurgens TM, Naylor H. Herbs, vitamins and minerals in the treatment of premenstrual syndrome: a systematic review. Can J Clin Pharmacol. 2009;16(3):e407-e429.
  3. Atmaca M, Kumru S, Tezcan E. Fluoxetine versus Vitex agnus castus extract in the treatment of premenstrual dysphoric disorder. Hum Psychopharmacol. 2003;18(3):191-195.
  4. Thys-Jacobs S, Starkey P, Bernstein D, et al. Calcium carbonate and the premenstrual syndrome: effects on premenstrual and menstrual symptoms. Premenstrual Syndrome Study Group. Am J Obstet Gynecol. 1998;179(2):444-452.
  5. Parry BL, Berga SL, Mostofi N, et al. Morning versus evening bright light treatment of late luteal phase dysphoric disorder. Am J Psychiatry. 1989;146(9):1215-1217.
  6. Wang M, Hammarbäck S, Lindhe BA, et al. Treatment of premenstrual syndrome by spironolactone: a double-blind, placebo-controlled study. Acta Obstet Gynecol Scand. 1995;74(10):803-808.
  7. Lustyk MK, Gerrish WG, Shaver S, et al. Cognitive-behavioral therapy for premenstrual syndrome and premenstrual dysphoric disorder: a systematic review. Arch Womens Ment Health. 2009;12(2):85-96.
  8. Busse JW, Montori VM, Krasnik C, et al. Psychological intervention for premenstrual syndrome: a meta-analysis of randomized controlled trials. Psychother Psychosom. 2009;78(1):6-15.
 

 


Bibliography

Evidence supporting antidepressants as treatment for PMDD

Cohen LS, Miner C, Brown EW, et al. Premenstrual daily fluoxetine for premenstrual dysphoric disorder: a placebo-controlled, clinical trial using computerized diaries. Obstet Gynecol. 2002;100(3):435-444.

Eriksson E, Ekman A, Sinclair S, et al. Escitalopram administered in the luteal phase exerts a marked and dose-dependent effect in premenstrual dysphoric disorder. J Clin Psychopharmacol. 2008;28(2):195-202.

Eriksson E, Hedberg MA, Andersch B, et al. The serotonin reuptake inhibitor paroxetin is superior to the noradrenaline reuptake inhibitor maprotiline in the treatment of premenstrual syndrome. Neuropsychopharmacology. 1995;12(2):167-176.

Menkes DB, Taghavi E, Mason PA, et al. Fluoxetine treatment of severe premenstrual syndrome. BMJ. 1992;305(6849):346-347.

Miner C, Brown E, McCray S, et al. Weekly luteal-phase dosing with enteric-coated fluoxetine 90 mg in premenstrual dysphoric disorder: a randomized, double-blind, placebo-controlled clinical trial. Clin Ther. 2002;24(3):417-433.

Ozeren S, Corakçi A, Yücesoy I, et al. Fluoxetine in the treatment of premenstrual syndrome. Eur J Obstet Gynecol Reprod Biol. 1997;73(2):167-170.

Pearlstein TB, Stone AB, Lund SA, et al. Comparison of fluoxetine, bupropion, and placebo in the treatment of premenstrual dysphoric disorder. J Clin Psychopharmacol. 1997;17(4):261-266.

Ravindran LN, Woods SA, Steiner M, et al. Symptom-onset dosing with citalopram in the treatment of premenstrual dysphoric disorder (PMDD): a case series. Arch Womens Ment Health. 2007;10(3):125-127.

Steiner M, Brown E, Trzepacz P, et al. Fluoxetine improves functional work capacity in women with premenstrual dysphoric disorder. Arch Womens Ment Health. 2003;6(1):71-77.

Stone AB, Pearlstein TB, Brown WA. Fluoxetine in the treatment of late luteal phase dysphoric disorder. J Clin Psychiatry. 1991;52(7):290-293.

Sundblad C, Hedberg MA, Eriksson E. Clomipramine administered during the luteal phase reduces the symptoms of premenstrual syndrome: a placebo-controlled trial. Neuropsychopharmacology. 1993;9(2):133-145.

Sundblad C, Modigh K, Andersch B, et al. Clomipramine effectively reduces premenstrual irritability and dysphoria: a placebo-controlled trial. Acta Psychiatr Scand. 1992;85(1):39-47.

Su TP, Schmidt PJ, Danaceau MA, et al. Fluoxetine in the treatment of premenstrual dysphoria. Neuropsychopharmacology. 1997; 16(5):346-356.

Wikander I, Sundblad C, Andersch B, et al. Citalopram in premenstrual dysphoria: is intermittent treatment during luteal phases more effective than continuous medication throughout the menstrual cycle? J Clin Psychopharmacol. 1998;18(5):390-398.

Treatment selection

To enhance compliance and improve the likelihood of successful treatment, tailor treatment decisions to your patient’s needs. Carefully discuss with your patient the evidence-based literature to select the best option for her. Factors to consider when counseling patients include:

  • the patient’s age, cigarette smoking habits, and body mass index, which may contraindicate oral contraceptives
  • does the patient have regular cycle lengths?
  • can she adhere to an on-off schedule? If so, intermittent SSRI dosing may be a good treatment option
  • does the patient have irregular cycles?
  • is there evidence that symptoms persist into the follicular phase, albeit at a lower level? If so, continuous SSRI dosing may be the best option.

Related Resources

Drug Brand Names

  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Clonidine • Catapres, others
  • Danazol • Danocrine
  • Drospirenone/ethinyl estradiol • Yaz
  • Escitalopram • Lexapro
  • Fluoxetine • Prozac
  • Leuprolide • Lupron
  • Levonorgestrel/ethinyl estradiol • Seasonale, others
  • Paroxetine • Paxil
  • Progesterone • Prometrium
  • Sertraline • Zoloft
  • Spironolactone • Aldactone
  • Venlafaxine • Effexor

Disclosures

Drs. Wakil and Girdler report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products

Dr. Meltzer-Brody receives research/grant support from AstraZeneca, The Foundation of Hope, and the National Institutes of Health.

Approximately 75% of women experience a premenstrual change in emotional or physical symptoms commonly referred to as premenstrual syndrome (PMS). These symptoms—including increased irritability, tension, depressed mood, and somatic complaints such as breast tenderness and bloating—often are mild to moderate and cause minimal distress.1 However, approximately 3% to 9% of women experience moderate to severe premenstrual mood symptoms that meet criteria for premenstrual dysphoric disorder (PMDD).2

PMDD includes depressed or labile mood, anxiety, irritability, anger, insomnia, difficulty concentrating, and other symptoms that occur exclusively during the 2 weeks before menses and cause significant deterioration in daily functioning. Women with PMDD use general and mental health services more often than women without the condition.3 They may experience impairment in marital and parental relationships as severe as that experienced by women with recurrent or chronic major depression.2

PMDD often responds to treatment. Unfortunately, many women with PMDD do not seek treatment, and up to 90% may go undiagnosed.4 In this article, we review the prevalence, etiology, diagnosis, and treatment of PMDD.

A complex disorder

A distinguishing characteristic of PMDD is the timing of symptom onset. In women with PMDD, mood symptoms occur only during the luteal phase of the menstrual cycle (ovulation until onset of menses) and resolve after menstruation onset. Women with PMDD report normal mood and functioning during the follicular phase of the menstrual cycle (first day of the menstrual cycle until ovulation).

Although PMS and PMDD criteria share affective and somatic symptoms, more symptoms are required for a PMDD diagnosis, and symptoms often are more severe.5 As defined in DSM-IV-TR (Table),6 PMDD has a broader range of symptoms than PMS and includes symptoms not included in the American College of Obstetrics and Gynecology criteria for PMS,7 such as impaired concentration, appetite, and sleep (hypersomnia or insomnia); and mood lability. PMDD symptoms must occur only during the 2 weeks preceding menses, although on average symptoms last 6 days and severity usually peaks in the 2 days before menses.1 The prevalence of subthreshold PMDD is fairly common; approximately 19% of women will meet some—but not all—DSM-IV-TR criteria for PMDD.3

In a revision proposed for DSM-5, PMDD would be included as a mood disorder, which represents a significant change from DSM-IV-TR, where it is listed in the appendix as “research criteria.”8 In addition, in oral contraceptive users, a PMDD diagnosis should not be made unless the premenstrual symptoms are reported to be present and as severe when the woman is not taking the oral contraceptive.8

Comorbidity with other axis I disorders such as major depressive disorder (MDD), bipolar disorder (BD), and anxiety disorders is high.9-11 Women with an MDD history have the highest correlation with PMDD,9 and worsening premenstrual mood symptoms are more common in women with BD.12 Payne et al11 found that premenstrual symptoms were reported by twice as many women diagnosed with mood disorders (68%) than women without a psychiatric diagnosis (34%). Moreover, 38% to 46% of women with PMDD have comorbid seasonal affective disorder, and 11% to 38% report a comorbid anxiety disorder.12 Women with PMDD and a history of MDD have lower cortisol concentrations than non-PMDD women.10 Although interventions for PMDD and a comorbid axis I disorder may be similar, it is important to consider both when planning treatment.

Abuse, trauma, and PMDD. An association between PMS/PMDD and a history of sexual and physical abuse is well-documented.13 Studies have reported abuse histories among almost 60% of women with PMDD,14 although studies comparing abuse and trauma in PMDD vs non-PMDD women have been small. A recent study found that trauma and posttraumatic stress disorder are independently associated with PMDD and premenstrual symptoms.15

Evidence suggests that a history of abuse is associated with specific biological sequelae in PMDD women, particularly with respect to hypothalamic-pituitary-thyroid axis measures and noradrenergic activity.16-18 Women with PMDD and a history of sexual abuse show:

  • markedly elevated triiodothyronine (T3) concentrations (the more biologically potent thyroid hormone) that appear to result from increased conversion of thyroxine (T4) to T316
  • lower circulating plasma norepinephrine concentrations17
  • greater resting and stress-induced heart rates and systolic blood pressure compared with non-abused PMDD women, an effect that is eliminated by clonidine (an α-2 adrenergic receptor agonist).18

One study showed that PMDD women with abuse histories had higher blood pressure measurements at rest and during stress and exhibited greater vascular tone than non-abused women; these effects were not seen in non-PMDD women with similar abuse histories.14 This body of evidence is consistent with the concept that PMDD is a stress-related disorder,19 and that a history of abuse is prevalent and may identify a clinically distinct subgroup of PMDD women with respect to thyroid axis and adrenergic physiology. Screening PMDD patients for abuse histories may help manage the disorder.

 

 

For a discussion of the etiology of PMDD, see Box 1.

Table

DSM-IV-TR research criteria for PMDD

  1. In most menstrual cycles of the past year, ≥5 of the following symptoms must be present for most of the time during the last week of the luteal phase, begin to remit within a few days after the onset of the follicular phase, and are absent in the week postmenses, with ≥1 of the symptoms being either 1, 2, 3, or 4:
  2. The disturbance markedly interferes with work or school or with usual social activities and relationships with others (eg, avoidance of social activities, decreased productivity and efficiency at work or school)
  3. The disturbance is not merely an exacerbation of the symptoms of another disorder, such as major depressive disorder, panic disorder, dysthymic disorder, or a personality disorder (although it may be superimposed on any of these disorders)
  4. Criteria A, B, and C must be confirmed by prospective daily ratings during ≥2 consecutive symptomatic cycles (The diagnosis may be made provisionally prior to this confirmation)
Source: Reference 6

Box 1

Reproductive hormones in PMDD etiology

Although questions remain about the pathogenesis of premenstrual dysphoric disorder (PMDD), literature documents the role of gonadal steroids (estrogen and progesterone) in the etiology of premenstrual syndrome (PMS)/PMDD and suggests that women with PMDD are differentially sensitive to the normal physiologic fluctuations of gonadal hormones throughout the menstrual cycle.a

The first half of the menstrual cycle—the follicular phase—begins with increasing levels of follicular stimulating hormone (FSH) leading to maturity of the ovarian follicle. Once the follicle is ripe, the luteal phase of the menstrual cycle begins with a surge in luteinizing hormone (LH), which results in ovulation of the mature follicle, followed by increased secretion of progesterone, followed by increased estrogen secretion. The system is regulated via negative feedback, and high levels of progesterone decrease gonadotropin-releasing hormone (GnRH) pulse frequency, which leads to decreased secretion of FSH and LH, and subsequent decline of estrogen and progesterone. If the ovarian follicle is not fertilized, menstruation begins and FSH levels rise again, initiating the follicular phase of the menstrual cycle.

Fluctuations in reproductive steroid levels have been implicated in the etiology of PMDD from studies showing that oophorectomy and ovulation inhibitors (GnRH agonists) relieve symptoms.b Some researchers proposed that symptoms are related to the drop of progesterone in the late luteal phase; however, many women have symptoms that start at ovulation or during the early luteal phase before the fall in progesterone concentrations.c PMS symptoms may occur independently of the mid-to-late luteal phase.d Because production of gonadal steroids does not differ between women with or without PMS or PMDD,e it may be that follicular or periovulatory changes in levels of estradiol or progesterone secretion trigger symptoms of PMDD in susceptible women, while women without PMDD appear to be immune to these effects of gonadal steroids. This idea is supported by a study showing that pharmacologic induction of a hypogonadal state eliminates symptoms in most women with severe PMS, while “adding back” estrogen or progesterone within the context of hypogonadism elicits return of PMS symptoms in those with PMS but not in controls.a

Abnormalities in serotonin levels also may contribute to PMDD.f In 1 study, a serotonin receptor antagonist precipitated return of symptoms within 24 hours of administration in women with PMDD but not in controls.g PMDD symptoms also can be evoked by depleting the serotonin precursor tryptophan.h When women with PMDD received paroxetine at different phases of their menstrual cycle, they showed fluctuations in serotonergic function across their cycles; these fluctuations were not seen in controls.i Other neurotransmitters implicated in PMDD include γ-aminobutyric acid (GABA),j glutamate,k lower levels of cortisol and beta-endorphins,l and an abnormal stress response.m

Other studies have focused on differing concentrations of luteal phase hormonesn and gene associations. Two studies suggested that PMDD is heritableo,p and other studies have looked at the association between specific psychological traits that are more prominent in PMDD and single nucleotide polymorphisms in the estrogen receptor alpha gene.q,r

Thyroid hormones also may play a role in the etiology of PMS/PMDD. Thyroid function tests have shown greater variability in women with PMS vs controls,s although this variability appears to be limited to women with a sexual abuse history.t Other studies have evaluated hormones regulated across the circadian and sleep-wake cycles, including melatonin, cortisol, thyroid-stimulating hormone, and prolactin, which suggests that although levels of these hormones may not differ between women with PMDD and controls, the timing of their excretion may vary.s Additionally, women with PMDD are characterized by prefrontal brain asymmetry on electroencephalography that also is evident in patients with major depressive disorder.u

There also may be dysregulation of allopregnanolone (ALLO) in women with PMDD.v,w ALLO is a metabolite of progesterone that is a neurosteroid produced in the brain as well as in the ovary and adrenals.v It produces anxiolytic effects by acting as a modulator of GABA receptors.x In PMDD, ALLO levels may influence the severity of premenstrual symptoms.w

References

  1. Schmidt PJ, Nieman LK, Danaceau MA, et al. Differential behavioral effects of gonadal steroids in women with and in those without premenstrual syndrome. N Engl J Med. 1998;338(4):209-216.
  2. Muse KN, Cetel NS, Futterman LA, et al. The premenstrual syndrome. Effects of “medical ovariectomy.” N Engl J Med. 1984;311(21):1345-1349.
  3. Yonkers KA, O’Brien PM, Eriksson E. Premenstrual syndrome. Lancet. 2008;371(9619):1200-1210.
  4. Schmidt PJ, Nieman LK, Grover GN, et al. Lack of effect of induced menses on symptoms in women with premenstrual syndrome. N Engl J Med. 1991;324(17):1174-1179.
  5. Rubinow DR, Schmidt PJ. The neuroendocrinology of menstrual cycle mood disorders. Ann N Y Acad Sci. 1995;771:648-659.
  6. Steiner M, Pearlstein T. Premenstrual dysphoria and the serotonin system: pathophysiology and treatment. J Clin Psychiatry. 2000;61(suppl 12):17-21.
  7. Roca CA, Schmidt PJ, Smith MJ, et al. Effects of metergoline on symptoms in women with premenstrual dysphoric disorder. Am J Psychiatry. 2002;159(11):1876-1881.
  8. Menkes DB, Coates DC, Fawcett JP. Acute tryptophan depletion aggravates premenstrual syndrome. J Affect Disord. 1994;32(1):37-44.
  9. Inoue Y, Terao T, Iwata N, et al. Fluctuating serotonergic function in premenstrual dysphoric disorder and premenstrual syndrome: findings from neuroendocrine challenge tests. Psychopharmacology (Berl). 2007;190(2):213-219.
  10. Epperson CN, Haga K, Mason GF, et al. Cortical gamma-aminobutyric acid levels across the menstrual cycle in healthy women and those with premenstrual dysphoric disorder: a proton magnetic resonance spectroscopy study. Arch Gen Psychiatry. 2002;59(9):851-858.
  11. Batra NA, Seres-Mailo J, Hanstock C, et al. Proton magnetic resonance spectroscopy measurement of brain glutamate levels in premenstrual dysphoric disorder. Biol Psychiatry. 2008;63(12):1178-1184.
  12. Straneva PA, Maixner W, Light KC, et al. Menstrual cycle, beta-endorphins, and pain sensitivity in premenstrual dysphoric disorder. Health Psychol. 2002;21(4):358-367.
  13. Epperson CN, Pittman B, Czarkowski KA, et al. Luteal-phase accentuation of acoustic startle response in women with premenstrual dysphoric disorder. Neuropsychopharmacology. 2007;32(10):2190-2198.
  14. Thys-Jacobs S, McMahon D, Bilezikian JP. Differences in free estradiol and sex hormone-binding globulin in women with and without premenstrual dysphoric disorder. J Clin Endocrinol Metab. 2008;93(1):96-102.
  15. Payne JL, Klein SR, Zamoiski RB, et al. Premenstrual mood symptoms: study of familiality and personality correlates in mood disorder pedigrees. Arch Womens Ment Health. 2009;12(1):27-34.
  16. Kendler KS, Karkowski LM, Corey LA, et al. Longitudinal population-based twin study of retrospectively reported premenstrual symptoms and lifetime major depression. Am J Psychiatry. 1998;155(9):1234-1240.
  17. Miller A, Vo H, Huo L, et al. Estrogen receptor alpha (ESR-1) associations with psychological traits in women with PMDD and controls. J Psychiatr Res. 2010;44(12):788-794.
  18. Huo L, Straub RE, Roca C, et al. Risk for premenstrual dysphoric disorder is associated with genetic variation in ESR1, the estrogen receptor alpha gene. Biol Psychiatry. 2007;62(8):925-933.
  19. Girdler SS, Pedersen CA, Light KC. Thyroid axis function during the menstrual cycle in women with premenstrual syndrome. Psychoneuroendocrinology. 1995;20(4):395-403.
  20. Girdler SS, Thompson KS, Light KC, et al. Historical sexual abuse and current thyroid axis profiles in women with premenstrual dysphoric disorder. Psychosom Med. 2004;66(3):403-410.
  21. Accortt EE, Stewart JL, Coan JA, et al. Prefrontal brain asymmetry and pre-menstrual dysphoric disorder symptomatology. J Affect Disord. 2011;128(1-2):178-183.
  22. Paul SM, Purdy RH. Neuroactive steroids. FASEB J. 1992;6(6):2311-2322.
  23. Girdler SS, Straneva PA, Light KC, et al. Allopregnanolone levels and reactivity to mental stress in premenstrual dysphoric disorder. Biol Psychiatry. 2001;49(9):788-797.
  24. Brot MD, Akwa Y, Purdy RH, et al. The anxiolytic-like effects of the neurosteroid allopregnanolone: interactions with GABA(A) receptors. Eur J Pharmacol. 1997;325(1):1-7.
 

 

Mood charting aids diagnosis

A PMDD diagnosis requires prospective daily monitoring of symptoms for ≥2 consecutive months. Because only 25% to 35% of women who present with PMDD meet diagnostic criteria when prospective daily monitoring is used,20 it is important for patients to keep a daily diary of PMDD symptoms to distinguish the disorder from PMS (Box 2). The Prospective Record of the Impact and Severity of Premenstrual Symptoms calendar and the Daily Record of Severity of Problems (DRSPP)21 may help make the diagnosis.

The widely used DRSPP allows clinicians to quantify the severity of physical, emotional, and behavioral symptoms and may be the easiest to use in clinical practice because it creates a graphic representation of cyclical symptom changes. The DRSPP includes all PMDD symptoms and severity ratings21 and is recognized as a valid instrument for diagnosing PMDD. Another option is a revised visual analogue scale. Lastly, a new revised Premenstrual Tension Syndrome (PMTS) rating scale, which combines the PMTS Observer rating scale plus multiple visual analogue scales, shows promise as a tool to assess PMDD symptoms.

Box 2

PMS or PMDD? Charting symptoms over menstrual cycles

To distinguish premenstrual syndrome (PMS) from premenstrual dysphoric disorder (PMDD), premenstrual exacerbation of an underlying psychiatric disorder, general medical conditions, or other disorders with no association to the menstrual cycle, it is necessary to have patients conduct daily symptom charting over 2 menstrual cycles. This charting should include documentation of emotional, behavioral, and physical symptoms. PMDD can be differentiated from PMS by the severity and number of symptoms. In PMDD, 1 of the symptoms must be a mood disturbance (depressed, anxious, labile, and/or irritable). For a sample form used for PMDD charting, the Daily Record of Severity of Problems, see http://pmdd.factsforhealth.org/drsp/drsp_month.pdf.

Treatment options

Hormonal interventions. Attempts to treat PMS with progesterone during the luteal phase have been largely unsuccessful, although progesterone is approved to treat PMS in the United Kingdom. Long-acting gonadotropin-releasing hormone (GnRH) agonists are effective but result in medical menopause with its accompanying symptoms, which puts women at risk for osteoporosis.22 Approximately 60% to 70% of women with PMDD respond to leuprolide (a GnRH agonist), but it is difficult to predict who will respond; daily mood self-ratings of sadness, anxiety, and irritability predict a positive response to leuprolide with high probability.23 Side effects of GnRH agonists (hot flashes, night sweats, vaginal dryness, etc.) can be tempered by “adding back” some estrogen with a hormonal agent with progestational activity to reduce the risks of unopposed estrogen (ie, endometrial hyperplasia).24

Surgical bilateral oophorectomy is effective but extremely invasive, especially in younger women in whom removal of ovaries generally is inadvisable. Patients should receive a trial of a GnRH agonist before a surgical intervention, because oophorectomy may not reduce symptoms and is irreversible. Oophorectomy also would require hormone replacement therapy.

High-dose estrogen as transdermal patches or subcutaneous implants to inhibit ovulation is effective, but because of the risks of unopposed estrogen, a progestin would be needed. Risks of estrogen therapy (alone and in combination with progestins) include increased risk of endometrial cancer, coronary heart disease, breast cancer, stroke, and pulmonary embolism.25 Danazol, a synthetic androgen and gonadotropin inhibitor, effectively blocks ovulation, but side effects include hirsutism and possible teratogenicity.26 Although these hormonal manipulations may effectively treat PMDD, none are considered practical.

The use of combined oral contraceptives (estrogen and progestin) is common. Although continuous cycle oral contraceptives often are recommended for PMDD, limited evidence supports their use; studies have been mostly negative.27,28 A recent review of 4 studies of a continuous oral contraceptive (levonorgestrel 90 mcg/ethinyl estradiol 20 mcg) for PMDD and PMS had more promising results, although the results were highly variable among studies and a large placebo effect was observed.29

A combination oral contraceptive, drospirenone/ethinyl estradiol, is FDA-approved for treating PMDD in women seeking hormonal contraception because it has shown efficacy compared with placebo, with reported improvements in perceived productivity, social activities, and interpersonal relationships.30 The nature of hormone delivery (ie, a reduction in the pill-free interval from 7 to 4 days) in drospirenone/ethinyl estradiol may contribute to its efficacy because a reduced pill-free interval minimizes the degree of follicular recruitment and subsequent estrogen production and cyclicity seen with standard oral contraceptive.31

 

 

Antidepressants have been shown to effectively ameliorate affective and physical symptoms and improve quality of life and psychosocial function in patients with PMS and PMDD. The response rates for selective serotonin reuptake inhibitors (SSRIs) in PMDD treatment vary from 60% to 90%, vs 30% to 40% for placebo.32 A 2009 Cochrane review found SSRIs reduced premenstrual symptoms compared with placebo.33 However, a literature review suggested that the percentage of women with PMDD who respond to SSRIs or continuous oral contraceptives is lower than the percentage of women who do not respond at all, once the placebo effect is taken into account, and that approximately 40% of women with PMDD do not respond to SSRIs.34 A small study found that citalopram may be effective for women with PMDD who did not respond to a prior SSRI.35

However, only antidepressants that affect serotonergic—not noradrenergic—transmission are effective in PMDD.22 These include:

  • the tricyclic antidepressant clomipramine
  • the SSRIs citalopram, escitalopram, fluoxetine, paroxetine, and sertraline
  • the serotonin-noradrenergic reuptake inhibitor venlafaxine.

It appears that in PMDD, serotonergic agents play a role other than their antidepressant effect.36 The effect of these agents is rapid in PMS/PMDD; women with PMDD who take antidepressants often experience reduced symptoms within the first menstrual cycle, whereas in MDD the onset of action can take weeks or months.37

Although why onset of antidepressant action is quick in PMDD is unclear, rapid onset allows for several dosing options. Some women prefer continuous dosing throughout the month because they do not have to keep track of ovulation. Dosing antidepressants only in the luteal phase (taking the antidepressant from ovulation onset to the start of menses) is an effective treatment strategy.38 Many women prefer to take medication for only 2 weeks per month, which can decrease side effects and lower treatment costs. Alternatively, symptom-onset dosing—initiating the antidepressant when PMDD symptoms begin and stopping at menses onset or within 3 days thereafter—has shown promising results.39,40 Paroxetine, sertraline, and fluoxetine are FDA-approved for PMDD as continuous or intermittent regimens, although using fluoxetine intermittently may not make sense because its biologically active metabolite has an extended half-life.37

Other treatments. Dietary interventions, psychotherapy, vitamins, bright light treatment, and spironolactone have been assessed for PMS/PMDD, although for many evidence-based findings are lacking (Box 3).

See the Bibliography below for studies that support using antidepressants to treat PMDD

Box 3

Beyond hormones and antidepressants: Other treatments for PMDD

Two reviews of 10 randomized controlled trials (RCTs) that evaluated 62 herbs, vitamins, and mineral treatments for premenstrual symptoms found efficacy for chasteberry (Vitex agnus-castus), calcium, and vitamin B6 but not for primrose oil, magnesium oxide, or St. John’s wort.a,b A study comparing fluoxetine with chasteberry found a similar percentage of patients responded to either agent (68% vs 58%, respectively).c Another study showed calcium resulted in a 48% reduction in premenstrual symptoms from baseline, compared with a 30% reduction with placebo.d Bright light treatment significantly reduced depression ratings in women with premenstrual dysphoric disorder (PMDD).e Compared with placebo, the aldosterone antagonist spironolactone improved irritability, depression, feelings of swelling, breast tenderness, and food craving in women with premenstrual syndrome (PMS).f

A recent systematic review of 7 trials of cognitive-behavioral therapy (CBT) for PMDD, including 3 RCTs, showed a lack of a statistically significant effect.g However, a separate review of RCTs of alternative treatments for PMDD—5 of which included CBT—suggested that CBT may be beneficial in reducing premenstrual symptoms, but the evidence was low quality.h

References

  1. Dante G, Facchinetti F. Herbal treatments for alleviating premenstrual symptoms: a systematic review. J Psychosom Obstet Gynaecol. 2011;32(1):42-51.
  2. Whelan AM, Jurgens TM, Naylor H. Herbs, vitamins and minerals in the treatment of premenstrual syndrome: a systematic review. Can J Clin Pharmacol. 2009;16(3):e407-e429.
  3. Atmaca M, Kumru S, Tezcan E. Fluoxetine versus Vitex agnus castus extract in the treatment of premenstrual dysphoric disorder. Hum Psychopharmacol. 2003;18(3):191-195.
  4. Thys-Jacobs S, Starkey P, Bernstein D, et al. Calcium carbonate and the premenstrual syndrome: effects on premenstrual and menstrual symptoms. Premenstrual Syndrome Study Group. Am J Obstet Gynecol. 1998;179(2):444-452.
  5. Parry BL, Berga SL, Mostofi N, et al. Morning versus evening bright light treatment of late luteal phase dysphoric disorder. Am J Psychiatry. 1989;146(9):1215-1217.
  6. Wang M, Hammarbäck S, Lindhe BA, et al. Treatment of premenstrual syndrome by spironolactone: a double-blind, placebo-controlled study. Acta Obstet Gynecol Scand. 1995;74(10):803-808.
  7. Lustyk MK, Gerrish WG, Shaver S, et al. Cognitive-behavioral therapy for premenstrual syndrome and premenstrual dysphoric disorder: a systematic review. Arch Womens Ment Health. 2009;12(2):85-96.
  8. Busse JW, Montori VM, Krasnik C, et al. Psychological intervention for premenstrual syndrome: a meta-analysis of randomized controlled trials. Psychother Psychosom. 2009;78(1):6-15.
 

 


Bibliography

Evidence supporting antidepressants as treatment for PMDD

Cohen LS, Miner C, Brown EW, et al. Premenstrual daily fluoxetine for premenstrual dysphoric disorder: a placebo-controlled, clinical trial using computerized diaries. Obstet Gynecol. 2002;100(3):435-444.

Eriksson E, Ekman A, Sinclair S, et al. Escitalopram administered in the luteal phase exerts a marked and dose-dependent effect in premenstrual dysphoric disorder. J Clin Psychopharmacol. 2008;28(2):195-202.

Eriksson E, Hedberg MA, Andersch B, et al. The serotonin reuptake inhibitor paroxetin is superior to the noradrenaline reuptake inhibitor maprotiline in the treatment of premenstrual syndrome. Neuropsychopharmacology. 1995;12(2):167-176.

Menkes DB, Taghavi E, Mason PA, et al. Fluoxetine treatment of severe premenstrual syndrome. BMJ. 1992;305(6849):346-347.

Miner C, Brown E, McCray S, et al. Weekly luteal-phase dosing with enteric-coated fluoxetine 90 mg in premenstrual dysphoric disorder: a randomized, double-blind, placebo-controlled clinical trial. Clin Ther. 2002;24(3):417-433.

Ozeren S, Corakçi A, Yücesoy I, et al. Fluoxetine in the treatment of premenstrual syndrome. Eur J Obstet Gynecol Reprod Biol. 1997;73(2):167-170.

Pearlstein TB, Stone AB, Lund SA, et al. Comparison of fluoxetine, bupropion, and placebo in the treatment of premenstrual dysphoric disorder. J Clin Psychopharmacol. 1997;17(4):261-266.

Ravindran LN, Woods SA, Steiner M, et al. Symptom-onset dosing with citalopram in the treatment of premenstrual dysphoric disorder (PMDD): a case series. Arch Womens Ment Health. 2007;10(3):125-127.

Steiner M, Brown E, Trzepacz P, et al. Fluoxetine improves functional work capacity in women with premenstrual dysphoric disorder. Arch Womens Ment Health. 2003;6(1):71-77.

Stone AB, Pearlstein TB, Brown WA. Fluoxetine in the treatment of late luteal phase dysphoric disorder. J Clin Psychiatry. 1991;52(7):290-293.

Sundblad C, Hedberg MA, Eriksson E. Clomipramine administered during the luteal phase reduces the symptoms of premenstrual syndrome: a placebo-controlled trial. Neuropsychopharmacology. 1993;9(2):133-145.

Sundblad C, Modigh K, Andersch B, et al. Clomipramine effectively reduces premenstrual irritability and dysphoria: a placebo-controlled trial. Acta Psychiatr Scand. 1992;85(1):39-47.

Su TP, Schmidt PJ, Danaceau MA, et al. Fluoxetine in the treatment of premenstrual dysphoria. Neuropsychopharmacology. 1997; 16(5):346-356.

Wikander I, Sundblad C, Andersch B, et al. Citalopram in premenstrual dysphoria: is intermittent treatment during luteal phases more effective than continuous medication throughout the menstrual cycle? J Clin Psychopharmacol. 1998;18(5):390-398.

Treatment selection

To enhance compliance and improve the likelihood of successful treatment, tailor treatment decisions to your patient’s needs. Carefully discuss with your patient the evidence-based literature to select the best option for her. Factors to consider when counseling patients include:

  • the patient’s age, cigarette smoking habits, and body mass index, which may contraindicate oral contraceptives
  • does the patient have regular cycle lengths?
  • can she adhere to an on-off schedule? If so, intermittent SSRI dosing may be a good treatment option
  • does the patient have irregular cycles?
  • is there evidence that symptoms persist into the follicular phase, albeit at a lower level? If so, continuous SSRI dosing may be the best option.

Related Resources

Drug Brand Names

  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Clonidine • Catapres, others
  • Danazol • Danocrine
  • Drospirenone/ethinyl estradiol • Yaz
  • Escitalopram • Lexapro
  • Fluoxetine • Prozac
  • Leuprolide • Lupron
  • Levonorgestrel/ethinyl estradiol • Seasonale, others
  • Paroxetine • Paxil
  • Progesterone • Prometrium
  • Sertraline • Zoloft
  • Spironolactone • Aldactone
  • Venlafaxine • Effexor

Disclosures

Drs. Wakil and Girdler report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products

Dr. Meltzer-Brody receives research/grant support from AstraZeneca, The Foundation of Hope, and the National Institutes of Health.

References

1. Yonkers KA, O’Brien PM, Eriksson E. Premenstrual syndrome. Lancet. 2008;371(9619):1200-1210.

2. Halbreich U, Borenstein J, Pearlstein T, et al. The prevalence, impairment, impact and burden of premenstrual dysphoric disorder (PMS/PMDD). Psychoneuroendocrinology. 2008;28(suppl 3):1-23.

3. Wittchen HU, Becker E, Lieb R, et al. Prevalence, incidence and stability of premenstrual dysphoric disorder in the community. Psychol Med. 2002;32(1):119-132.

4. Hylan TR, Sundell K, Judge R. The impact of premenstrual symptomatology on functioning and treatment-seeking behavior: experience from the United States United Kingdom, and France. J Womens Health Gend Based Med. 1999;8(8):1043-1052.

5. Biggs WS, Demuth RH. Premenstrual syndrome and premenstrual dysphoric disorder. Am Fam Physician. 2011;84(8):918-924.

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

7. ACOG Practice Bulletin. Clinical management guidelines for obstetrician-gynecologists. Number 15 April 2000. Premenstrual syndrome. Obstet Gynecol. 2000;95:1-9.

8. American Psychiatric Association. Proposed draft revisions to DSM disorders and criteria. DSM-5 development. http://www.dsm5.org/proposedrevision/Pages/Default.aspx. Accessed February 23 2012.

9. Yonkers KA. The association between premenstrual dysphoric disorder and other mood disorders. J Clin Psychiatry. 1997;58(suppl 15):19-25.

10. Klatzkin RR, Lindgren ME, Forneris CA, et al. Histories of major depression and premenstrual dysphoric disorder: evidence for phenotypic differences. Biol Psychol. 2010;84(2):235-247.

11. Payne JL, Roy PS, Murphy-Eberenz K, et al. Reproductive cycle-associated mood symptoms in women with major depression and bipolar disorder. J Affect Disord. 2007;99(1-3):221-229.

12. Kim DR, Gyulai L, Freeman EW, et al. Premenstrual dysphoric disorder and psychiatric co-morbidity. Arch Womens Ment Health. 2004;7(1):37-47.

13. Golding JM, Taylor DL, Menard L, et al. Prevalence of sexual abuse history in a sample of women seeking treatment for premenstrual syndrome. J Psychosom Obstet Gynaecol. 2000;21(2):69-80.

14. Girdler SS, Leserman J, Bunevicius R, et al. Persistent alterations in biological profiles in women with abuse histories: influence of premenstrual dysphoric disorder. Health Psychol. 2007;26(2):201-213.

15. Pilver CE, Levy BR, Libby DJ, et al. Posttraumatic stress disorder and trauma characteristics are correlates of premenstrual dysphoric disorder. Arch Womens Ment Health. 2011;14(5):383-393.

16. Girdler SS, Thompson KS, Light KC, et al. Historical sexual abuse and current thyroid axis profiles in women with premenstrual dysphoric disorder. Psychosom Med. 2004;66(3):403-410.

17. Girdler SS, Sherwood A, Hinderliter AL, et al. Biological correlates of abuse in women with premenstrual dysphoric disorder and healthy controls. Psychosom Med. 2003;65(5):849-856.

18. Bunevicius R, Hinderliter AL, Light KC, et al. Histories of sexual abuse are associated with differential effects of clonidine on autonomic function in women with premenstrual dysphoric disorder. Biol Psychol. 2005;69(3):281-296.

19. Perkonigg A, Yonkers KA, Pfister H, et al. Risk factors for premenstrual dysphoric disorder in a community sample of young women: the role of traumatic events and posttraumatic stress disorder. J Clin Psychiatry. 2004;65(10):1314-1322.

20. Girdler SS, Pedersen CA, Stern RA, et al. Menstrual cycle and premenstrual syndrome: modifiers of cardiovascular reactivity in women. Health Psychol. 1993;12(3):180-192.

21. Endicott J, Nee J, Harrison W. Daily Record of Severity of Problems (DRSP): reliability and validity. Arch Womens Ment Health. 2006;9(1):41-49.

22. Cunningham J, Yonkers KA, O’Brien S, et al. Update on research and treatment of premenstrual dysphoric disorder. Harv Rev Psychiatry. 2009;17(2):120-137.

23. Pincus SM, Alam S, Rubinow DR, et al. Predicting response to leuprolide of women with premenstrual dysphoric disorder by daily mood rating dynamics. J Psychiatr Res. 2011;45(3):386-394.

24. Wyatt KM, Dimmock PW, Ismail KM, et al. The effectiveness of GnRHa with and without ‘add-back’ therapy in treating premenstrual syndrome: a meta analysis. BJOG. 2004;111(6):585-593.

25. Rossouw JE, Anderson GL, Prentice RL, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288(3):321-333.

26. Hahn PM, Van Vugt DA, Reid RL. A randomized placebo-controlled, crossover trial of danazol for the treatment of premenstrual syndrome. Psychoneuroendocrinology. 1995;20(2):193-209.

27. Bancroft J, Rennie D. The impact of oral contraceptives on the experience of perimenstrual mood clumsiness, food craving and other symptoms. J Psychosom Res. 1993;37(2):195-202.

28. Graham CA, Sherwin BB. A prospective treatment study of premenstrual symptoms using a triphasic oral contraceptive. J Psychosom Res. 1992;36(3):257-266.

29. Freeman EW, Halbreich U, Grubb GS, et al. An overview of four studies of a continuous oral contraceptive (levonorgestrel 90 mcg/ethinyl estradiol 20 mcg) on premenstrual dysphoric disorder and premenstrual syndrome [published online ahead of print December 5, 2011]. Contraception. doi: 10.1016/j.contraception.2011.09.010.

30. Lopez LM, Kaptein AA, Helmerhorst FM. Oral contraceptives containing drospirenone for premenstrual syndrome. Cochrane Database Syst Rev. 2009;(2):CD006586.

31. Sullivan H, Furniss H, Spona J, et al. Effect of 21-day and 24-day oral contraceptive regimens containing gestodene (60 microg) andethinyl estradiol (15 microg) on ovarian activity. Fertil Steril. 1999;72(1):115-120.

32. Yonkers KA, Clark RH, Trivedi MH. The psychopharmacological treatment of nonmajor mood disorders. Mod Probl Pharmacopsychiatry. 1997;25:146-166.

33. Brown J, O’Brien PM, Marjoribanks J, et al. Selective serotonin reuptake inhibitors for premenstrual syndrome. Cochrane Database Syst Rev. 2009;(2):CD001396.

34. Halbreich U. Selective serotonin reuptake inhibitors and initial oral contraceptives for the treatment of PMDD: effective but not enough. CNS Spectr. 2008;13(7):566-572.

35. Freeman EW, Jabara S, Sondheimer SJ, et al. Citalopram in PMS patients with prior SSRI treatment failure: a preliminary study. J Womens Health Gend Based Med. 2002;11(5):

459-464.

36. Freeman EW, Rickels K, Sondheimer SJ, et al. Differential response to antidepressants in women with premenstrual syndrome/premenstrual dysphoric disorder: a randomized controlled trial. Arch Gen Psychiatry. 1999;56(10):932-939.

37. Steiner M, Pearlstein T, Cohen LS, et al. Expert guidelines for the treatment of severe PMS, PMDD, and comorbidities: the role of SSRIs. J Womens Health (Larchmt). 2006;15(1):57-69.

38. Freeman EW. Luteal phase administration of agents for the treatment of premenstrual dysphoric disorder. CNS Drugs. 2004;18(7):453-468.

39. Yonkers KA, Holthausen GA, Poschman K, et al. Symptom-onset treatment for women with premenstrual dysphoric disorder. J Clin Psychopharmacol. 2006;26(2):198-202.

40. Freeman EW, Sondheimer SJ, Sammel MD, et al. A preliminary study of luteal phase versus symptom-onset dosing with escitalopram for premenstrual dysphoric disorder. J Clin Psychiatry. 2005;66(6):769-773.

References

1. Yonkers KA, O’Brien PM, Eriksson E. Premenstrual syndrome. Lancet. 2008;371(9619):1200-1210.

2. Halbreich U, Borenstein J, Pearlstein T, et al. The prevalence, impairment, impact and burden of premenstrual dysphoric disorder (PMS/PMDD). Psychoneuroendocrinology. 2008;28(suppl 3):1-23.

3. Wittchen HU, Becker E, Lieb R, et al. Prevalence, incidence and stability of premenstrual dysphoric disorder in the community. Psychol Med. 2002;32(1):119-132.

4. Hylan TR, Sundell K, Judge R. The impact of premenstrual symptomatology on functioning and treatment-seeking behavior: experience from the United States United Kingdom, and France. J Womens Health Gend Based Med. 1999;8(8):1043-1052.

5. Biggs WS, Demuth RH. Premenstrual syndrome and premenstrual dysphoric disorder. Am Fam Physician. 2011;84(8):918-924.

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

7. ACOG Practice Bulletin. Clinical management guidelines for obstetrician-gynecologists. Number 15 April 2000. Premenstrual syndrome. Obstet Gynecol. 2000;95:1-9.

8. American Psychiatric Association. Proposed draft revisions to DSM disorders and criteria. DSM-5 development. http://www.dsm5.org/proposedrevision/Pages/Default.aspx. Accessed February 23 2012.

9. Yonkers KA. The association between premenstrual dysphoric disorder and other mood disorders. J Clin Psychiatry. 1997;58(suppl 15):19-25.

10. Klatzkin RR, Lindgren ME, Forneris CA, et al. Histories of major depression and premenstrual dysphoric disorder: evidence for phenotypic differences. Biol Psychol. 2010;84(2):235-247.

11. Payne JL, Roy PS, Murphy-Eberenz K, et al. Reproductive cycle-associated mood symptoms in women with major depression and bipolar disorder. J Affect Disord. 2007;99(1-3):221-229.

12. Kim DR, Gyulai L, Freeman EW, et al. Premenstrual dysphoric disorder and psychiatric co-morbidity. Arch Womens Ment Health. 2004;7(1):37-47.

13. Golding JM, Taylor DL, Menard L, et al. Prevalence of sexual abuse history in a sample of women seeking treatment for premenstrual syndrome. J Psychosom Obstet Gynaecol. 2000;21(2):69-80.

14. Girdler SS, Leserman J, Bunevicius R, et al. Persistent alterations in biological profiles in women with abuse histories: influence of premenstrual dysphoric disorder. Health Psychol. 2007;26(2):201-213.

15. Pilver CE, Levy BR, Libby DJ, et al. Posttraumatic stress disorder and trauma characteristics are correlates of premenstrual dysphoric disorder. Arch Womens Ment Health. 2011;14(5):383-393.

16. Girdler SS, Thompson KS, Light KC, et al. Historical sexual abuse and current thyroid axis profiles in women with premenstrual dysphoric disorder. Psychosom Med. 2004;66(3):403-410.

17. Girdler SS, Sherwood A, Hinderliter AL, et al. Biological correlates of abuse in women with premenstrual dysphoric disorder and healthy controls. Psychosom Med. 2003;65(5):849-856.

18. Bunevicius R, Hinderliter AL, Light KC, et al. Histories of sexual abuse are associated with differential effects of clonidine on autonomic function in women with premenstrual dysphoric disorder. Biol Psychol. 2005;69(3):281-296.

19. Perkonigg A, Yonkers KA, Pfister H, et al. Risk factors for premenstrual dysphoric disorder in a community sample of young women: the role of traumatic events and posttraumatic stress disorder. J Clin Psychiatry. 2004;65(10):1314-1322.

20. Girdler SS, Pedersen CA, Stern RA, et al. Menstrual cycle and premenstrual syndrome: modifiers of cardiovascular reactivity in women. Health Psychol. 1993;12(3):180-192.

21. Endicott J, Nee J, Harrison W. Daily Record of Severity of Problems (DRSP): reliability and validity. Arch Womens Ment Health. 2006;9(1):41-49.

22. Cunningham J, Yonkers KA, O’Brien S, et al. Update on research and treatment of premenstrual dysphoric disorder. Harv Rev Psychiatry. 2009;17(2):120-137.

23. Pincus SM, Alam S, Rubinow DR, et al. Predicting response to leuprolide of women with premenstrual dysphoric disorder by daily mood rating dynamics. J Psychiatr Res. 2011;45(3):386-394.

24. Wyatt KM, Dimmock PW, Ismail KM, et al. The effectiveness of GnRHa with and without ‘add-back’ therapy in treating premenstrual syndrome: a meta analysis. BJOG. 2004;111(6):585-593.

25. Rossouw JE, Anderson GL, Prentice RL, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288(3):321-333.

26. Hahn PM, Van Vugt DA, Reid RL. A randomized placebo-controlled, crossover trial of danazol for the treatment of premenstrual syndrome. Psychoneuroendocrinology. 1995;20(2):193-209.

27. Bancroft J, Rennie D. The impact of oral contraceptives on the experience of perimenstrual mood clumsiness, food craving and other symptoms. J Psychosom Res. 1993;37(2):195-202.

28. Graham CA, Sherwin BB. A prospective treatment study of premenstrual symptoms using a triphasic oral contraceptive. J Psychosom Res. 1992;36(3):257-266.

29. Freeman EW, Halbreich U, Grubb GS, et al. An overview of four studies of a continuous oral contraceptive (levonorgestrel 90 mcg/ethinyl estradiol 20 mcg) on premenstrual dysphoric disorder and premenstrual syndrome [published online ahead of print December 5, 2011]. Contraception. doi: 10.1016/j.contraception.2011.09.010.

30. Lopez LM, Kaptein AA, Helmerhorst FM. Oral contraceptives containing drospirenone for premenstrual syndrome. Cochrane Database Syst Rev. 2009;(2):CD006586.

31. Sullivan H, Furniss H, Spona J, et al. Effect of 21-day and 24-day oral contraceptive regimens containing gestodene (60 microg) andethinyl estradiol (15 microg) on ovarian activity. Fertil Steril. 1999;72(1):115-120.

32. Yonkers KA, Clark RH, Trivedi MH. The psychopharmacological treatment of nonmajor mood disorders. Mod Probl Pharmacopsychiatry. 1997;25:146-166.

33. Brown J, O’Brien PM, Marjoribanks J, et al. Selective serotonin reuptake inhibitors for premenstrual syndrome. Cochrane Database Syst Rev. 2009;(2):CD001396.

34. Halbreich U. Selective serotonin reuptake inhibitors and initial oral contraceptives for the treatment of PMDD: effective but not enough. CNS Spectr. 2008;13(7):566-572.

35. Freeman EW, Jabara S, Sondheimer SJ, et al. Citalopram in PMS patients with prior SSRI treatment failure: a preliminary study. J Womens Health Gend Based Med. 2002;11(5):

459-464.

36. Freeman EW, Rickels K, Sondheimer SJ, et al. Differential response to antidepressants in women with premenstrual syndrome/premenstrual dysphoric disorder: a randomized controlled trial. Arch Gen Psychiatry. 1999;56(10):932-939.

37. Steiner M, Pearlstein T, Cohen LS, et al. Expert guidelines for the treatment of severe PMS, PMDD, and comorbidities: the role of SSRIs. J Womens Health (Larchmt). 2006;15(1):57-69.

38. Freeman EW. Luteal phase administration of agents for the treatment of premenstrual dysphoric disorder. CNS Drugs. 2004;18(7):453-468.

39. Yonkers KA, Holthausen GA, Poschman K, et al. Symptom-onset treatment for women with premenstrual dysphoric disorder. J Clin Psychopharmacol. 2006;26(2):198-202.

40. Freeman EW, Sondheimer SJ, Sammel MD, et al. A preliminary study of luteal phase versus symptom-onset dosing with escitalopram for premenstrual dysphoric disorder. J Clin Psychiatry. 2005;66(6):769-773.

Issue
Current Psychiatry - 11(04)
Issue
Current Psychiatry - 11(04)
Page Number
22-41
Page Number
22-41
Publications
Publications
Article Type
Display Headline
Premenstrual dysphoric disorder: How to alleviate her suffering
Display Headline
Premenstrual dysphoric disorder: How to alleviate her suffering
Legacy Keywords
Premenstrual dysphoric disorder; PMDD; mood charting; hormonal intervention; antidepressants;
Legacy Keywords
Premenstrual dysphoric disorder; PMDD; mood charting; hormonal intervention; antidepressants;
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Getting ready for DSM-5: Psychotic disorders

Article Type
Changed
Tue, 12/11/2018 - 15:04
Display Headline
Getting ready for DSM-5: Psychotic disorders

Discuss this article at www.facebook.com/CurrentPsychiatry

In DSM-IV,1 the section on schizophrenia and other psychotic disorders includes schizophrenia (with 5 subtypes), schizophreniform disorder, schizoaffective disorder, delusional disorder, shared psychotic disorder, brief psychotic disorder, substance-induced psychotic disorder, psychotic disorder due to a general medical condition, and psychotic disorder not otherwise specified. As we consider proposed changes to DSM-5 (Table 1),2 it is useful to consider limitations in our current construct of schizophrenia.

Table 1

Psychotic disorders in DSM-5: Summary of proposed changes

Replace existing subtypes with dimensions
Include diagnosis of attenuated psychosis syndrome
Modify criteria for schizoaffective disorder
‘Delink’ catatonia from schizophrenia
Source: Reference 2

First, many etiological factors and pathophysiological processes appear relevant to what we consider schizophrenia and it is almost certain that our construct of schizophrenia encompasses not one but numerous diseases with a shared phenotype.3-5

Second, the boundary between schizophrenia and schizoaffective disorder is imprecisely defined, and a proportion of patients with schizophrenia with some mood symptoms may inappropriately receive a schizoaffective disorder diagnosis. This is compounded by the poor reliability and low diagnostic stability of a schizoaffective disorder diagnosis.6-8

Third, the current classic schizophrenia subtypes provide an inadequate description of the enormous heterogeneity of this condition. Additionally, subtype stability is low, and only the paranoid and undifferentiated subtypes are used frequently in clinical practice.

Fourth, the prominence given to Schneiderian first-rank symptoms (“bizarre” delusions or “special” hallucinations) appears misplaced.

Fifth, the current construct of schizophrenia inadequately describes the major psychopathological dimensions of the condition and stages of its evolution.8,9

Finally, the current clinical construct of schizophrenia does not match neurobiological markers and genetic findings or specific pharmacological treatment provided.5,10 Proposed DSM-5 revisions2 to the definition of schizophrenia to address these limitations are summarized below.

Schizophrenia syndrome

Proposed changes to the diagnostic criteria for schizophrenia are modest and continuity with DSM-IV is broadly maintained. Two modest changes to criterion A (active phase symptoms) are proposed:

  • Eliminate special treatment of bizarre delusions and other Schneiderian first-rank symptoms. In DSM-IV, only 1 criterion A is required if it is a bizarre delusion or hallucination. Because Schneiderian first-rank symptoms do not have diagnostic specificity and diagnosing “bizarreness” of delusions and hallucinations has low reliability, it is proposed that these positive symptoms be treated like any other with regard to their diagnostic implications.
  • Require that at least 1 of the 2 symptoms required to meet criterion A be delusions, hallucinations, or disorganized thinking. These are core positive symptoms diagnosed with high reliability and might reasonably be considered necessary for a reliable schizophrenia diagnosis.

Subtypes

The DSM-5 proposal for describing schizophrenia advocates eliminating DSM-IV schizophrenia subtypes. These subtypes have limited diagnostic stability, low reliability, and poor validity. Furthermore, except for the paranoid and undifferentiated subtypes, other subtypes rarely are used in most mental health care systems.

Schizoaffective disorder

Characterizing patients with both psychotic and mood symptoms either concurrently or at different points during their illness always has been controversial. In DSM-I and DSM-II, a diagnosis of schizophrenia, schizoaffective subtype, generally was recommended for such patients. DSM-III reversed this recommendation and specified that schizophrenia was to be diagnosed only in the absence of prominent mood symptoms. Furthermore, in DSM-III, diagnosing schizoaffective disorder was strongly discouraged, and it was the only condition in DSM-III without operational criteria. Schizoaffective disorder saw a revival in DSM-III-R that has continued through DSM-IV. In fact, in many mental health care systems, almost one-third of patients with psychotic symptoms receive a schizoaffective disorder diagnosis. One of the insidious changes to the definition of schizoaffective disorder from DSM-III to DSM-IV is that it moved from being a lifetime diagnosis to a cross-sectional diagnosis—ie, in DSM-IV, only mood/psychotic symptoms in the current episode are considered, and the longitudinal course of these symptoms in the patient’s life are ignored. The current DSM-5 proposal attempts to improve reliability of this diagnosis by providing more specific criteria and is reconceptualizing schizoaffective disorder as a longitudinal diagnosis. To this end, the most significant proposed change is to criterion C of schizoaffective disorder, which attempts to demarcate schizoaffective disorder from schizophrenia with prominent mood symptoms. Criterion C will be revised to state “symptoms that meet criteria for a mood episode are present for a majority (>50%) of the total duration of the active and residual periods of the illness.”2

Psychopathological dimensions

 

 

Schizophrenic illness is characterized by several psychopathological domains, with a distinctive course, patterns of treatment-response, and prognostic implications. The relative severity of symptom dimensions—positive, negative, mood, disorganization, motor, and cognitive—vary among patients and also within patients at different stages of their illness. Measuring the relative severity of these symptom dimensions throughout the illness course can provide clinicians with useful information about the nature of a patient’s schizophrenic illness and the specific impact of treatment on different aspects of his or her illness (Table 2). In addition to being clinically useful, dimensional measurement also should improve schizophrenia research because having dimensional information will permit studies on etiology and pathogenesis that cut across current diagnostic categories. Although field trials are evaluating 9 dimensions—delusions, hallucinations, disorganization, depression, mania, cognitive impairment, restricted emotional expression, avolition, and psychomotor—it is likely that fewer dimensions will be recommended for DSM-5, based on reliability results of these trials, clinical utility, and logistic feasibility in routine clinical settings.

Table 2

Goals of a dimensional approach to schizophrenia

Better understanding of schizophrenia
Distinct dimensions of illness
Distinct stages of illness
Elucidation of neurobiology
More precise delineation of etiology
More refined treatment development
Direction at specific dimension-endophenotype
Stage-specific treatment
Novel treatment targets

Attenuated psychosis syndrome

Some clinicians and researchers believe that many patients with schizophrenia experience unsatisfactory outcomes because we identify the illness and initiate treatment after substantial brain tissue damage has occurred. Introducing attenuated psychosis syndrome will support clinicians’ efforts to recognize mild psychotic symptoms early in their evolution and to monitor—and if necessary, intervene—during these crucial early stages. Risks include possible stigma and inappropriate use of medications and other treatments. This controversial proposal is being field tested. It is unclear if this category will be included in DSM-5 and if it does, whether it will be in the main text or the appendix.

Catatonia

Catatonia will be used as a specifier for various psychotic disorders, major mood disorders, and associated with a general medical condition. Additionally, the same criteria will be used to diagnose catatonia across DSM-5. Catatonia Not Elsewhere Classified might be added as a residual category for other conditions in which a clear catatonic syndrome is present and the parent disorder has not yet been identified.2

Other psychotic disorders

Relatively minor changes are proposed in criteria for other disorders in this section. There are likely to be changes in the text, however, that incorporate new information about these conditions generated since publication of DSM-IV-TR in 2000. Some proposed changes include:

  • deleting shared delusional disorder (folie à deux) as a separate diagnosis and instead characterizing it as a specifier for delusional disorder
  • clarifying the distinction between substance-induced psychotic disorder and other psychotic disorders accompanied by comorbid substance use.

Current status of DSM-5

Field trials are being completed and their results remain to be analyzed. Major changes being evaluated in the field trials include:

  • the impact of the change in concept and criteria for schizoaffective disorder
  • the addition of a series of psychopathology dimensions
  • the impact of adding attenuated psychosis syndrome as a new class.

Changes proposed by the Psychosis Disorders Work Group are intended to increase clinical utility (fewer diagnoses, better demarcation between disorders, greater treatment relevance [dimensions]) and modestly improve validity (more consistent with current information about the nature of various psychotic disorders), while retaining reliability in diagnosing various psychotic disorders (and improving it for schizoaffective disorder). Proposed changes are modest by and large but hope to set a better stage for a future etiopathophysiological classification.

The Psychosis Disorders Work Group’s recommendations are posted on the DSM-5 Web site2 at www.dsm5.org and are being reviewed by 2 expert committees established by the American Psychiatric Association Board of Trustees: a Scientific Review Committee and a Clinical and Public Health Implications Committee. Based on the results of the field trials, ongoing reviews, and other emerging data and discussions, additional changes to the current DSM-5 proposals may occur. DSM-5 is likely to be finalized in early 2013 and the published manual will be released in May 2013.

Related Resources

  • American Psychiatric Association. DSM-5 Development. www.dsm5.org.
  • Woods SW, McGlashan TH. The risk-benefit ratio of the proposed DSM-5 attenuated psychosis syndrome. Am J Psychiatry. 2011;168(12):1338.

Disclosure

Dr. Tandon is a member of the DSM-5 Psychotic Disorders Work Group. He is solely responsible for the content of this article.

References

1. Diagnostic and statistical manual of mental disorders, 4th ed. Washington, DC: American Psychiatric Association; 1994.

2. American Psychiatric Association. DSM-5 development. http://www.dsm5.org. Accessed March 19, 2011.

3. Tandon R, Keshavan MS, Nasrallah HA. Schizophrenia “just the facts”: what we know in 2008. Part 1: overview. Schizophr Res. 2008;100(1-3):4-19.

4. Tandon R, Keshavan MS, Nasrallah HA. Schizophrenia “just the facts” what we know in 2008. 2: epidemiology and etiology. Schizophr Res. 2008;102(1-3):1-18.

5. Keshavan MS, Tandon R, Boutros N, et al. Schizophrenia, “just the facts” what we know in 2008. Part 3: neurobiology. Schizophr Res. 2008;106(2-3):89-107.

6. Tandon R, Maj M. Nosological status and definition of schizophrenia. Some considerations for DSM-V and ICD-11. Asian Journal of Psychiatry. 2008;1(2):22-27.

7. Fiedorowicz JG, Epping EA, Flaum M. Toward defining schizophrenia as a more useful clinical construct. Curr Psychiatry Rep. 2008;10(4):344-351.

8. Tandon R, Nasrallah HA, Keshavan MS. Schizophrenia “just the facts” 4. Clinical features and conceptualization. Schizophr Res. 2009;110(1-3):1-23.

9. McGorry PD. Risk syndromes clinical staging, and DSM V: new diagnostic infrastructure for early intervention in psychiatry. Schizophr Res. 2010;120(1-3):49-53.

10. Tandon R, Nasrallah HA, Keshavan MS. Schizophrenia “just the facts” 5. Treatment and prevention. Past, present, and future. Schizophr Res. 2010;122(1-3):1-23.

Article PDF
Author and Disclosure Information


Rajiv Tandon, MD
Professor of Psychiatry, University of Florida, Gainesville, FL

Issue
Current Psychiatry - 11(04)
Publications
Topics
Page Number
E1-E4
Legacy Keywords
DSM-5; psychotic disorders; changes; schizophrenia; schizoaffective disorder;
Sections
Author and Disclosure Information


Rajiv Tandon, MD
Professor of Psychiatry, University of Florida, Gainesville, FL

Author and Disclosure Information


Rajiv Tandon, MD
Professor of Psychiatry, University of Florida, Gainesville, FL

Article PDF
Article PDF

Discuss this article at www.facebook.com/CurrentPsychiatry

In DSM-IV,1 the section on schizophrenia and other psychotic disorders includes schizophrenia (with 5 subtypes), schizophreniform disorder, schizoaffective disorder, delusional disorder, shared psychotic disorder, brief psychotic disorder, substance-induced psychotic disorder, psychotic disorder due to a general medical condition, and psychotic disorder not otherwise specified. As we consider proposed changes to DSM-5 (Table 1),2 it is useful to consider limitations in our current construct of schizophrenia.

Table 1

Psychotic disorders in DSM-5: Summary of proposed changes

Replace existing subtypes with dimensions
Include diagnosis of attenuated psychosis syndrome
Modify criteria for schizoaffective disorder
‘Delink’ catatonia from schizophrenia
Source: Reference 2

First, many etiological factors and pathophysiological processes appear relevant to what we consider schizophrenia and it is almost certain that our construct of schizophrenia encompasses not one but numerous diseases with a shared phenotype.3-5

Second, the boundary between schizophrenia and schizoaffective disorder is imprecisely defined, and a proportion of patients with schizophrenia with some mood symptoms may inappropriately receive a schizoaffective disorder diagnosis. This is compounded by the poor reliability and low diagnostic stability of a schizoaffective disorder diagnosis.6-8

Third, the current classic schizophrenia subtypes provide an inadequate description of the enormous heterogeneity of this condition. Additionally, subtype stability is low, and only the paranoid and undifferentiated subtypes are used frequently in clinical practice.

Fourth, the prominence given to Schneiderian first-rank symptoms (“bizarre” delusions or “special” hallucinations) appears misplaced.

Fifth, the current construct of schizophrenia inadequately describes the major psychopathological dimensions of the condition and stages of its evolution.8,9

Finally, the current clinical construct of schizophrenia does not match neurobiological markers and genetic findings or specific pharmacological treatment provided.5,10 Proposed DSM-5 revisions2 to the definition of schizophrenia to address these limitations are summarized below.

Schizophrenia syndrome

Proposed changes to the diagnostic criteria for schizophrenia are modest and continuity with DSM-IV is broadly maintained. Two modest changes to criterion A (active phase symptoms) are proposed:

  • Eliminate special treatment of bizarre delusions and other Schneiderian first-rank symptoms. In DSM-IV, only 1 criterion A is required if it is a bizarre delusion or hallucination. Because Schneiderian first-rank symptoms do not have diagnostic specificity and diagnosing “bizarreness” of delusions and hallucinations has low reliability, it is proposed that these positive symptoms be treated like any other with regard to their diagnostic implications.
  • Require that at least 1 of the 2 symptoms required to meet criterion A be delusions, hallucinations, or disorganized thinking. These are core positive symptoms diagnosed with high reliability and might reasonably be considered necessary for a reliable schizophrenia diagnosis.

Subtypes

The DSM-5 proposal for describing schizophrenia advocates eliminating DSM-IV schizophrenia subtypes. These subtypes have limited diagnostic stability, low reliability, and poor validity. Furthermore, except for the paranoid and undifferentiated subtypes, other subtypes rarely are used in most mental health care systems.

Schizoaffective disorder

Characterizing patients with both psychotic and mood symptoms either concurrently or at different points during their illness always has been controversial. In DSM-I and DSM-II, a diagnosis of schizophrenia, schizoaffective subtype, generally was recommended for such patients. DSM-III reversed this recommendation and specified that schizophrenia was to be diagnosed only in the absence of prominent mood symptoms. Furthermore, in DSM-III, diagnosing schizoaffective disorder was strongly discouraged, and it was the only condition in DSM-III without operational criteria. Schizoaffective disorder saw a revival in DSM-III-R that has continued through DSM-IV. In fact, in many mental health care systems, almost one-third of patients with psychotic symptoms receive a schizoaffective disorder diagnosis. One of the insidious changes to the definition of schizoaffective disorder from DSM-III to DSM-IV is that it moved from being a lifetime diagnosis to a cross-sectional diagnosis—ie, in DSM-IV, only mood/psychotic symptoms in the current episode are considered, and the longitudinal course of these symptoms in the patient’s life are ignored. The current DSM-5 proposal attempts to improve reliability of this diagnosis by providing more specific criteria and is reconceptualizing schizoaffective disorder as a longitudinal diagnosis. To this end, the most significant proposed change is to criterion C of schizoaffective disorder, which attempts to demarcate schizoaffective disorder from schizophrenia with prominent mood symptoms. Criterion C will be revised to state “symptoms that meet criteria for a mood episode are present for a majority (>50%) of the total duration of the active and residual periods of the illness.”2

Psychopathological dimensions

 

 

Schizophrenic illness is characterized by several psychopathological domains, with a distinctive course, patterns of treatment-response, and prognostic implications. The relative severity of symptom dimensions—positive, negative, mood, disorganization, motor, and cognitive—vary among patients and also within patients at different stages of their illness. Measuring the relative severity of these symptom dimensions throughout the illness course can provide clinicians with useful information about the nature of a patient’s schizophrenic illness and the specific impact of treatment on different aspects of his or her illness (Table 2). In addition to being clinically useful, dimensional measurement also should improve schizophrenia research because having dimensional information will permit studies on etiology and pathogenesis that cut across current diagnostic categories. Although field trials are evaluating 9 dimensions—delusions, hallucinations, disorganization, depression, mania, cognitive impairment, restricted emotional expression, avolition, and psychomotor—it is likely that fewer dimensions will be recommended for DSM-5, based on reliability results of these trials, clinical utility, and logistic feasibility in routine clinical settings.

Table 2

Goals of a dimensional approach to schizophrenia

Better understanding of schizophrenia
Distinct dimensions of illness
Distinct stages of illness
Elucidation of neurobiology
More precise delineation of etiology
More refined treatment development
Direction at specific dimension-endophenotype
Stage-specific treatment
Novel treatment targets

Attenuated psychosis syndrome

Some clinicians and researchers believe that many patients with schizophrenia experience unsatisfactory outcomes because we identify the illness and initiate treatment after substantial brain tissue damage has occurred. Introducing attenuated psychosis syndrome will support clinicians’ efforts to recognize mild psychotic symptoms early in their evolution and to monitor—and if necessary, intervene—during these crucial early stages. Risks include possible stigma and inappropriate use of medications and other treatments. This controversial proposal is being field tested. It is unclear if this category will be included in DSM-5 and if it does, whether it will be in the main text or the appendix.

Catatonia

Catatonia will be used as a specifier for various psychotic disorders, major mood disorders, and associated with a general medical condition. Additionally, the same criteria will be used to diagnose catatonia across DSM-5. Catatonia Not Elsewhere Classified might be added as a residual category for other conditions in which a clear catatonic syndrome is present and the parent disorder has not yet been identified.2

Other psychotic disorders

Relatively minor changes are proposed in criteria for other disorders in this section. There are likely to be changes in the text, however, that incorporate new information about these conditions generated since publication of DSM-IV-TR in 2000. Some proposed changes include:

  • deleting shared delusional disorder (folie à deux) as a separate diagnosis and instead characterizing it as a specifier for delusional disorder
  • clarifying the distinction between substance-induced psychotic disorder and other psychotic disorders accompanied by comorbid substance use.

Current status of DSM-5

Field trials are being completed and their results remain to be analyzed. Major changes being evaluated in the field trials include:

  • the impact of the change in concept and criteria for schizoaffective disorder
  • the addition of a series of psychopathology dimensions
  • the impact of adding attenuated psychosis syndrome as a new class.

Changes proposed by the Psychosis Disorders Work Group are intended to increase clinical utility (fewer diagnoses, better demarcation between disorders, greater treatment relevance [dimensions]) and modestly improve validity (more consistent with current information about the nature of various psychotic disorders), while retaining reliability in diagnosing various psychotic disorders (and improving it for schizoaffective disorder). Proposed changes are modest by and large but hope to set a better stage for a future etiopathophysiological classification.

The Psychosis Disorders Work Group’s recommendations are posted on the DSM-5 Web site2 at www.dsm5.org and are being reviewed by 2 expert committees established by the American Psychiatric Association Board of Trustees: a Scientific Review Committee and a Clinical and Public Health Implications Committee. Based on the results of the field trials, ongoing reviews, and other emerging data and discussions, additional changes to the current DSM-5 proposals may occur. DSM-5 is likely to be finalized in early 2013 and the published manual will be released in May 2013.

Related Resources

  • American Psychiatric Association. DSM-5 Development. www.dsm5.org.
  • Woods SW, McGlashan TH. The risk-benefit ratio of the proposed DSM-5 attenuated psychosis syndrome. Am J Psychiatry. 2011;168(12):1338.

Disclosure

Dr. Tandon is a member of the DSM-5 Psychotic Disorders Work Group. He is solely responsible for the content of this article.

Discuss this article at www.facebook.com/CurrentPsychiatry

In DSM-IV,1 the section on schizophrenia and other psychotic disorders includes schizophrenia (with 5 subtypes), schizophreniform disorder, schizoaffective disorder, delusional disorder, shared psychotic disorder, brief psychotic disorder, substance-induced psychotic disorder, psychotic disorder due to a general medical condition, and psychotic disorder not otherwise specified. As we consider proposed changes to DSM-5 (Table 1),2 it is useful to consider limitations in our current construct of schizophrenia.

Table 1

Psychotic disorders in DSM-5: Summary of proposed changes

Replace existing subtypes with dimensions
Include diagnosis of attenuated psychosis syndrome
Modify criteria for schizoaffective disorder
‘Delink’ catatonia from schizophrenia
Source: Reference 2

First, many etiological factors and pathophysiological processes appear relevant to what we consider schizophrenia and it is almost certain that our construct of schizophrenia encompasses not one but numerous diseases with a shared phenotype.3-5

Second, the boundary between schizophrenia and schizoaffective disorder is imprecisely defined, and a proportion of patients with schizophrenia with some mood symptoms may inappropriately receive a schizoaffective disorder diagnosis. This is compounded by the poor reliability and low diagnostic stability of a schizoaffective disorder diagnosis.6-8

Third, the current classic schizophrenia subtypes provide an inadequate description of the enormous heterogeneity of this condition. Additionally, subtype stability is low, and only the paranoid and undifferentiated subtypes are used frequently in clinical practice.

Fourth, the prominence given to Schneiderian first-rank symptoms (“bizarre” delusions or “special” hallucinations) appears misplaced.

Fifth, the current construct of schizophrenia inadequately describes the major psychopathological dimensions of the condition and stages of its evolution.8,9

Finally, the current clinical construct of schizophrenia does not match neurobiological markers and genetic findings or specific pharmacological treatment provided.5,10 Proposed DSM-5 revisions2 to the definition of schizophrenia to address these limitations are summarized below.

Schizophrenia syndrome

Proposed changes to the diagnostic criteria for schizophrenia are modest and continuity with DSM-IV is broadly maintained. Two modest changes to criterion A (active phase symptoms) are proposed:

  • Eliminate special treatment of bizarre delusions and other Schneiderian first-rank symptoms. In DSM-IV, only 1 criterion A is required if it is a bizarre delusion or hallucination. Because Schneiderian first-rank symptoms do not have diagnostic specificity and diagnosing “bizarreness” of delusions and hallucinations has low reliability, it is proposed that these positive symptoms be treated like any other with regard to their diagnostic implications.
  • Require that at least 1 of the 2 symptoms required to meet criterion A be delusions, hallucinations, or disorganized thinking. These are core positive symptoms diagnosed with high reliability and might reasonably be considered necessary for a reliable schizophrenia diagnosis.

Subtypes

The DSM-5 proposal for describing schizophrenia advocates eliminating DSM-IV schizophrenia subtypes. These subtypes have limited diagnostic stability, low reliability, and poor validity. Furthermore, except for the paranoid and undifferentiated subtypes, other subtypes rarely are used in most mental health care systems.

Schizoaffective disorder

Characterizing patients with both psychotic and mood symptoms either concurrently or at different points during their illness always has been controversial. In DSM-I and DSM-II, a diagnosis of schizophrenia, schizoaffective subtype, generally was recommended for such patients. DSM-III reversed this recommendation and specified that schizophrenia was to be diagnosed only in the absence of prominent mood symptoms. Furthermore, in DSM-III, diagnosing schizoaffective disorder was strongly discouraged, and it was the only condition in DSM-III without operational criteria. Schizoaffective disorder saw a revival in DSM-III-R that has continued through DSM-IV. In fact, in many mental health care systems, almost one-third of patients with psychotic symptoms receive a schizoaffective disorder diagnosis. One of the insidious changes to the definition of schizoaffective disorder from DSM-III to DSM-IV is that it moved from being a lifetime diagnosis to a cross-sectional diagnosis—ie, in DSM-IV, only mood/psychotic symptoms in the current episode are considered, and the longitudinal course of these symptoms in the patient’s life are ignored. The current DSM-5 proposal attempts to improve reliability of this diagnosis by providing more specific criteria and is reconceptualizing schizoaffective disorder as a longitudinal diagnosis. To this end, the most significant proposed change is to criterion C of schizoaffective disorder, which attempts to demarcate schizoaffective disorder from schizophrenia with prominent mood symptoms. Criterion C will be revised to state “symptoms that meet criteria for a mood episode are present for a majority (>50%) of the total duration of the active and residual periods of the illness.”2

Psychopathological dimensions

 

 

Schizophrenic illness is characterized by several psychopathological domains, with a distinctive course, patterns of treatment-response, and prognostic implications. The relative severity of symptom dimensions—positive, negative, mood, disorganization, motor, and cognitive—vary among patients and also within patients at different stages of their illness. Measuring the relative severity of these symptom dimensions throughout the illness course can provide clinicians with useful information about the nature of a patient’s schizophrenic illness and the specific impact of treatment on different aspects of his or her illness (Table 2). In addition to being clinically useful, dimensional measurement also should improve schizophrenia research because having dimensional information will permit studies on etiology and pathogenesis that cut across current diagnostic categories. Although field trials are evaluating 9 dimensions—delusions, hallucinations, disorganization, depression, mania, cognitive impairment, restricted emotional expression, avolition, and psychomotor—it is likely that fewer dimensions will be recommended for DSM-5, based on reliability results of these trials, clinical utility, and logistic feasibility in routine clinical settings.

Table 2

Goals of a dimensional approach to schizophrenia

Better understanding of schizophrenia
Distinct dimensions of illness
Distinct stages of illness
Elucidation of neurobiology
More precise delineation of etiology
More refined treatment development
Direction at specific dimension-endophenotype
Stage-specific treatment
Novel treatment targets

Attenuated psychosis syndrome

Some clinicians and researchers believe that many patients with schizophrenia experience unsatisfactory outcomes because we identify the illness and initiate treatment after substantial brain tissue damage has occurred. Introducing attenuated psychosis syndrome will support clinicians’ efforts to recognize mild psychotic symptoms early in their evolution and to monitor—and if necessary, intervene—during these crucial early stages. Risks include possible stigma and inappropriate use of medications and other treatments. This controversial proposal is being field tested. It is unclear if this category will be included in DSM-5 and if it does, whether it will be in the main text or the appendix.

Catatonia

Catatonia will be used as a specifier for various psychotic disorders, major mood disorders, and associated with a general medical condition. Additionally, the same criteria will be used to diagnose catatonia across DSM-5. Catatonia Not Elsewhere Classified might be added as a residual category for other conditions in which a clear catatonic syndrome is present and the parent disorder has not yet been identified.2

Other psychotic disorders

Relatively minor changes are proposed in criteria for other disorders in this section. There are likely to be changes in the text, however, that incorporate new information about these conditions generated since publication of DSM-IV-TR in 2000. Some proposed changes include:

  • deleting shared delusional disorder (folie à deux) as a separate diagnosis and instead characterizing it as a specifier for delusional disorder
  • clarifying the distinction between substance-induced psychotic disorder and other psychotic disorders accompanied by comorbid substance use.

Current status of DSM-5

Field trials are being completed and their results remain to be analyzed. Major changes being evaluated in the field trials include:

  • the impact of the change in concept and criteria for schizoaffective disorder
  • the addition of a series of psychopathology dimensions
  • the impact of adding attenuated psychosis syndrome as a new class.

Changes proposed by the Psychosis Disorders Work Group are intended to increase clinical utility (fewer diagnoses, better demarcation between disorders, greater treatment relevance [dimensions]) and modestly improve validity (more consistent with current information about the nature of various psychotic disorders), while retaining reliability in diagnosing various psychotic disorders (and improving it for schizoaffective disorder). Proposed changes are modest by and large but hope to set a better stage for a future etiopathophysiological classification.

The Psychosis Disorders Work Group’s recommendations are posted on the DSM-5 Web site2 at www.dsm5.org and are being reviewed by 2 expert committees established by the American Psychiatric Association Board of Trustees: a Scientific Review Committee and a Clinical and Public Health Implications Committee. Based on the results of the field trials, ongoing reviews, and other emerging data and discussions, additional changes to the current DSM-5 proposals may occur. DSM-5 is likely to be finalized in early 2013 and the published manual will be released in May 2013.

Related Resources

  • American Psychiatric Association. DSM-5 Development. www.dsm5.org.
  • Woods SW, McGlashan TH. The risk-benefit ratio of the proposed DSM-5 attenuated psychosis syndrome. Am J Psychiatry. 2011;168(12):1338.

Disclosure

Dr. Tandon is a member of the DSM-5 Psychotic Disorders Work Group. He is solely responsible for the content of this article.

References

1. Diagnostic and statistical manual of mental disorders, 4th ed. Washington, DC: American Psychiatric Association; 1994.

2. American Psychiatric Association. DSM-5 development. http://www.dsm5.org. Accessed March 19, 2011.

3. Tandon R, Keshavan MS, Nasrallah HA. Schizophrenia “just the facts”: what we know in 2008. Part 1: overview. Schizophr Res. 2008;100(1-3):4-19.

4. Tandon R, Keshavan MS, Nasrallah HA. Schizophrenia “just the facts” what we know in 2008. 2: epidemiology and etiology. Schizophr Res. 2008;102(1-3):1-18.

5. Keshavan MS, Tandon R, Boutros N, et al. Schizophrenia, “just the facts” what we know in 2008. Part 3: neurobiology. Schizophr Res. 2008;106(2-3):89-107.

6. Tandon R, Maj M. Nosological status and definition of schizophrenia. Some considerations for DSM-V and ICD-11. Asian Journal of Psychiatry. 2008;1(2):22-27.

7. Fiedorowicz JG, Epping EA, Flaum M. Toward defining schizophrenia as a more useful clinical construct. Curr Psychiatry Rep. 2008;10(4):344-351.

8. Tandon R, Nasrallah HA, Keshavan MS. Schizophrenia “just the facts” 4. Clinical features and conceptualization. Schizophr Res. 2009;110(1-3):1-23.

9. McGorry PD. Risk syndromes clinical staging, and DSM V: new diagnostic infrastructure for early intervention in psychiatry. Schizophr Res. 2010;120(1-3):49-53.

10. Tandon R, Nasrallah HA, Keshavan MS. Schizophrenia “just the facts” 5. Treatment and prevention. Past, present, and future. Schizophr Res. 2010;122(1-3):1-23.

References

1. Diagnostic and statistical manual of mental disorders, 4th ed. Washington, DC: American Psychiatric Association; 1994.

2. American Psychiatric Association. DSM-5 development. http://www.dsm5.org. Accessed March 19, 2011.

3. Tandon R, Keshavan MS, Nasrallah HA. Schizophrenia “just the facts”: what we know in 2008. Part 1: overview. Schizophr Res. 2008;100(1-3):4-19.

4. Tandon R, Keshavan MS, Nasrallah HA. Schizophrenia “just the facts” what we know in 2008. 2: epidemiology and etiology. Schizophr Res. 2008;102(1-3):1-18.

5. Keshavan MS, Tandon R, Boutros N, et al. Schizophrenia, “just the facts” what we know in 2008. Part 3: neurobiology. Schizophr Res. 2008;106(2-3):89-107.

6. Tandon R, Maj M. Nosological status and definition of schizophrenia. Some considerations for DSM-V and ICD-11. Asian Journal of Psychiatry. 2008;1(2):22-27.

7. Fiedorowicz JG, Epping EA, Flaum M. Toward defining schizophrenia as a more useful clinical construct. Curr Psychiatry Rep. 2008;10(4):344-351.

8. Tandon R, Nasrallah HA, Keshavan MS. Schizophrenia “just the facts” 4. Clinical features and conceptualization. Schizophr Res. 2009;110(1-3):1-23.

9. McGorry PD. Risk syndromes clinical staging, and DSM V: new diagnostic infrastructure for early intervention in psychiatry. Schizophr Res. 2010;120(1-3):49-53.

10. Tandon R, Nasrallah HA, Keshavan MS. Schizophrenia “just the facts” 5. Treatment and prevention. Past, present, and future. Schizophr Res. 2010;122(1-3):1-23.

Issue
Current Psychiatry - 11(04)
Issue
Current Psychiatry - 11(04)
Page Number
E1-E4
Page Number
E1-E4
Publications
Publications
Topics
Article Type
Display Headline
Getting ready for DSM-5: Psychotic disorders
Display Headline
Getting ready for DSM-5: Psychotic disorders
Legacy Keywords
DSM-5; psychotic disorders; changes; schizophrenia; schizoaffective disorder;
Legacy Keywords
DSM-5; psychotic disorders; changes; schizophrenia; schizoaffective disorder;
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Benzodiazepines: A versatile clinical tool

Article Type
Changed
Tue, 12/11/2018 - 15:04
Display Headline
Benzodiazepines: A versatile clinical tool

Since the discovery of chlordiazepoxide in the 1950s, benzodiazepines have revolutionized the treatment of anxiety and insomnia, largely because of their improved safety profile compared with barbiturates, formerly the preferred sedative-hypnotic.1 In addition to their anxiolytic and sedative-hypnotic effects, benzodiazepines exhibit anterograde amnesia, anticonvulsant, and muscle relaxant properties.1 Psychiatrists use benzodiazepines to treat anxiety and sleep disorders, acute agitation, alcohol withdrawal, catatonia, and psychotropic side effects such as akathisia. This article highlights the evidence for using benzodiazepines in anxiety and other disorders and why they generally should not be used for obsessive-compulsive disorder and posttraumatic stress disorder (Box 1).

Box 1

When not to use benzodiazepines: OCD and PTSD

Current evidence indicates little support for using benzodiazepines for obsessive-compulsive disorder (OCD). The American Psychiatric Association (APA) and the World Federation of Biological Psychiatry do not recommend benzodiazepines for treating OCD because of a lack of evidence for efficacy.a,b An earlier study suggested clonazepam monotherapy was effective for OCDc; however, a more recent study did not show a benefit on rate of response or degree of symptom improvement.d Augmentation strategies with benzodiazepines also do not appear to be beneficial for OCD management. A recent double-blind, placebo-controlled study failed to demonstrate faster symptom improvement by augmenting sertraline with clonazepam, although the study had a small sample size and high drop-out rate.e

Because benzodiazepines have negligible action on core posttraumatic stress disorder (PTSD) symptoms (re-experiencing, avoidance, and hyperarousal), selective serotonin reuptake inhibitors and other agents largely have supplanted them for PTSD treatment.f Use of benzodiazepines for PTSD is associated with withdrawal symptoms, more severe symptoms after discontinuation, and possible disinhibition, and may interfere with patients’ efforts to integrate trauma experiences. Although benzodiazepines may reduce distress associated with acute trauma, there is evidence—in clinical studies and animal models—that early benzodiazepine administration fails to prevent PTSD and may increase its incidence.g The International Consensus Group on Depression and Anxiety, the APA, and the British Association for Psychopharmacology all highlight the limited role, if any, for benzodiazepines in PTSD.h-j

References

  1. Bandelow B, Zohar J, Hollander E, et al. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the pharmacological treatment of anxiety, obsessive-compulsive and post-traumatic stress disorders - first revision. World J Biol Psychiatry. 2008;9(4):248-312.
  2. American Psychiatric Association. Practice guideline for the treatment of patients with obsessive-compulsive disorder. Arlington, VA: American Psychiatric Publishing, Inc.; 2007.
  3. Hewlett WA, Vinogradov S, Agras WS. Clomipramine, clonazepam, and clonidine treatment of obsessive compulsive disorder. J Clin Psychopharmacol. 1992;12(6):420-430.
  4. Hollander E, Kaplan A, Stahl SM. A double-blind, placebo-controlled trial of clonazepam in obsessive-compulsive disorder. World J Biol Psychiatry. 2003;4(1):30-34.
  5. Crockett BA, Churchill E, Davidson JR. A double-blind combination study of clonazepam and sertraline in OCD. Ann Clin Psychiatry. 2004;16(3):127-132.
  6. Argyropoulos SV, Sandford JJ, Nutt DJ. The psychobiology of anxiolytic drugs. Part 2: pharmacological treatments of anxiety. Pharmacol Ther. 2000;88(3):213-227.
  7. Matar MA, Zohar J, Kaplan Z, et al. Alprazolam treatment immediately after stress exposure interferes with the normal HPA-stress response and increases vulnerability to subsequent stress in an animal model of PTSD. Eur Neuropsychopharmacol. 2009;19(4):283-295.
  8. Ballenger JC, Davidson JR, Lecrubier Y, et al. Consensus statement update on posttraumatic stress disorder from the international consensus group on depression and anxiety. J Clin Psychiatry. 2004;65(suppl 1):55-62.
  9. Ursano RJ, Bell C, Eth S, et al. Practice guideline for the treatment of patients with acute stress disorder and posttraumatic stress disorder. Am J Psychiatry. 2004;161(11 suppl):3-31.
  10. Baldwin DS, Anderson IM, Nutt DJ, et al. Evidence-based guidelines for the pharmacological treatment of anxiety disorders: recommendations from the British Association for Psychopharmacology. J Psychopharmacol. 2005;19(6):567-596.

Pharmacokinetic properties

Most benzodiazepines are considered to have similar efficacy; therefore, selection is based on pharmacokinetic considerations. Table 1 compares the indication, onset, and half-life of 12 commonly used benzodiazepines.2-6 Although Table 1 lists approximate equivalent doses, studies report inconsistent data. These are approximations only and should not be used independently to make therapy decisions.

Table 1

Oral benzodiazepines: Indications, onset, half-life, and equivalent doses

DrugFDA-approved indication(s)Onset of actionApproximate half-life (hours) in healthy adultsApproximate equivalent dose (mg)aComments
AlprazolamAnxiety disorders, panic disorderIntermediate6.3 to 26.9 (IR), 10.7 to 15.8 (XR)0.5Increased risk for abuse because of greater lipid solubility
ChlordiazepoxideAnxiety disorders, acute alcohol withdrawal, preoperative apprehension and anxietyIntermediate24 to 4810Risk for accumulation because of long-acting metabolites (desmethyldiazepam, oxazepam)
ClonazepamSeizure disorders, panic disorderIntermediate18 to 500.25 to 0.5Use caution in patients with liver disease
ClorazepateAnxiety, seizures, acute alcohol withdrawalFast40 to 507.5Risk for accumulation because of long-acting metabolites (desmethyldiazepam, oxazepam)
DiazepamAnxiety disorders, acute alcohol withdrawal, muscle spasms, convulsive disordersVery fast20 to 1005Risk for accumulation because of long-acting metabolites (temazepam, desmethyldiazepam, oxazepam). Increased risk for abuse because of quick onset
EstazolamInsomniaIntermediate10 to 240.3 to 2None
FlurazepamInsomniaIntermediate47 to 10030Avoid in geriatric patients or patients with liver impairment
LorazepamAnxietyIntermediate10 to 201Preferred for patients with liver impairment and geriatric patients
OxazepamAnxiety, acute alcohol withdrawalSlow to intermediate5 to 2030Preferred for patients with liver impairment and geriatric patients
QuazepamInsomniaIntermediate39 to 735 to 15Risk for accumulation because of long-acting metabolites (desmethyldiazepam, oxazepam)
TemazepamInsomniaIntermediate3.5 to 18.430Preferred for patients with liver impairment and geriatric patients
TriazolamInsomniaFast1.5 to 5.50.25Lacks active metabolites
IR: immediate release; XR: extended release
aInterpret with caution, conflicting data exist
Source: References 2-6
 

 

A diverse range of indications

Alcohol withdrawal. Benzodiazepines are the treatment of choice for alcohol withdrawal syndrome, particularly to prevent seizures.7 Research supports symptom-triggered therapy using the revised Clinical Institute Withdrawal Assessment for Alcohol. Benzodiazepines reduce CNS sympathetic hyperactivity to mitigate withdrawal from alcohol by decreasing tachycardia, tremor, insomnia, agitation, and anxiety. Furthermore, these agents provide prophylaxis against serious sequelae such as seizures and delirium.

Insomnia. The American Academy of Sleep Medicine considers benzodiazepine receptor agonists (BzRAs, which include benzodiazepines and non-benzodiazepines) and ramelteon first-line pharmacotherapy for primary insomnia.8 However, pharmacologic treatment should be short-term. Agents with short to intermediate half-lives and rapid onset, such as triazolam, can aid sleep initiation. Those with longer half-lives, such as temazepam, could address sleep maintenance. If a patient does not respond to the initial agent, try another medication within the same class, because patients may respond differently. Use lower starting doses in geriatric patients.9 Closely monitor for adverse effects, rebound insomnia, and potential abuse or tolerance. Identify comorbid conditions and medications that may impair sleep, and address them accordingly.

Psychological and behavioral treatments given over 4 to 8 weeks can yield stable sleep improvements for up to 2 years. If available, these interventions may be considered first-line for treating insomnia because of their lasting effects compared with BzRAs.10

Generalized anxiety disorder (GAD). Benzodiazepines effectively treat GAD because they work quickly and are well tolerated. However, there are better first-line treatment options when considering efficacy studies and dependence and tolerance concerns. One effect-size comparison of 21 double-blind, placebo-controlled trials showed that the efficacy of selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), and pregabalin are comparable to benzodiazepines.11 Benzodiazepines can be used in the first 2 to 3 weeks after initiating antidepressants to alleviate and prevent worsening of anxiety that may occur at the start of antidepressant therapy. Recent treatment guidelines recommend benzodiazepines as a second-line treatment or for treatment-resistant GAD in patients who do not have a substance abuse history.12,13

Panic disorder. Efficacy of benzodiazepines for panic disorder is comparable to SSRIs, SNRIs, and tricyclic antidepressants (TCAs). SSRIs and SNRIs are considered first-line treatments for panic disorder because of their favorable side effect profile.14 In practice, benzodiazepines often are combined with SSRIs, SNRIs, or TCAs. A randomized controlled trial demonstrated that paroxetine and clonazepam (mean dose 1.6 mg/d at 5 weeks) resulted in a more rapid response compared with paroxetine alone, although this difference lasted only a few weeks.15 Furthermore, this study suggested that brief treatment with clonazepam followed by a taper is as effective as sustained treatment with paroxetine and clonazepam.15

There is a lack of high-quality data on combining cognitive-behavioral therapy (CBT) and benzodiazepines for panic disorder, although a Cochrane Review found that adding a benzodiazepine to CBT did not lead to a significant difference in response compared with psychotherapy alone.16 A recent randomized controlled trial demonstrated that tapering benzodiazepines combined with CBT was associated with successful discontinuation of the drug and prevented return of panic symptoms.17

Social anxiety. A meta-analysis found that for treating social anxiety, benzodiazepines have better efficacy than SSRIs, monoamine oxidase inhibitors, and anticonvulsants.18 Longer-acting benzodiazepines may be more effective than shorter-acting agents. One study of patients with social anxiety showed a 38% response rate for alprazolam vs 20% for placebo over 12 weeks, and a similar 10-week study demonstrated a 73% recovery rate with clonazepam vs 22% for placebo.19 In addition, studies have observed that patients can be maintained on clonazepam for up to 2 years without symptom relapse and will tolerate slow-taper discontinuation.18,20 Sedation and drowsiness can be lessened by limiting clonazepam doses to 2 to 3 mg/d.

Akathisia and tremor. Akathisia, a syndrome of motor restlessness and inner turmoil, is associated with antipsychotics but can occur with SSRIs. Reducing the dosage or switching to another, usually less potent agent often can relieve akathisia. When these remedies are not tenable, consider benzodiazepines along with other medications—including beta blockers and anticholinergic agents—with demonstrated efficacy in reducing akathisia symptoms. Lorazepam, diazepam, and clonazepam have demonstrated efficacy for relieving akathisia in comparison studies with placebo, propranolol, and diphenhydramine.21,22

Drug-induced postural tremor can occur with several psychotropics, including lithium, valproic acid, antidepressants, and antipsychotics. A tremor is considered mild if a patient can drink a glass of water with 1 hand without spilling and severe if holding a glass with 2 hands is difficult. Propranolol is most commonly prescribed for these tremors, but alprazolam and clonazepam have demonstrated efficacy, either as monotherapy or coadministered with a beta blocker.23

 

 

Acute agitation. Agitated patients often have acute psychosis and/or mania or dyscontrol secondary to axis II disorders.24 Patients may be paranoid, hostile, disruptive, and combative. Rapidly initiating medication can prevent the need for more restrictive measures, such as seclusion or restraint. Antipsychotics—especially high-potency agents such as haloperidol—and benzodiazepines, as monotherapy or in combination, are a mainstay treatment. Although treatment protocols favor atypical antipsychotics over typical antipsychotics, benzodiazepines are a viable option because of their anxiolytic and sedative effects. Advantages of benzodiazepine monotherapy include decreased extrapyramidal symptoms, greater patient acceptance/preference, and increased sedation compared with antipsychotics. Lorazepam, 1 to 2 mg intramuscularly (IM) or orally, is well tolerated because of its favorable drug-drug interaction profile and lack of significant cardiac side effects. Benzodiazepines can cause respiratory depression in patients with chronic lung disease and additive sedation secondary to opiates, other sedatives/hypnotics, or alcohol. Behavioral disinhibition is rare and is associated with preexisting CNS pathology or mental retardation.25 The IM olanzapine package insert warns against coadministering IM lorazepam because of additive cardiorespiratory depressive effects and excessive somnolence.26

Catatonia. The characteristic symptoms of catatonia are immobility, negativism, muteness, and failure to eat or drink. Benzodiazepines improve these symptoms in approximately 70% to 80% of catatonic patients with affective disorders. Response rates are lower in catatonia in patients with schizophrenia.27 If catatonia in a patient with psychosis is missed, giving antipsychotics before benzodiazepines may worsen catatonic symptoms or precipitate neuroleptic malignant syndrome in some cases. When you suspect a patient has catatonia, start with lorazepam, 1 to 2 mg IV or IM, and examine the patient for diminishing catatonic signs within 1 to 2 hours. If catatonia signs lessen, begin regularly scheduled lorazepam, with dosing varying by age—be more cautious in geriatric patients—and symptom severity. Titrate benzodiazepines for stuporous patients more slowly (eg, 1 mg 3 times a day as a starting dose) than for excited catatonic patients. Lorazepam can be increased gradually as tolerated; it is not unusual for patients to require up to 8 to 12 mg/d. Electroconvulsive therapy (ECT) is the treatment of choice when catatonic patients respond poorly or partially to high-dose benzodiazepines.28,29

Benzodiazepine reversal for ECT

Benzodiazepines have anticonvulsant properties that may interfere with the therapeutic efficacy of ECT.30 A multi-center study demonstrated that lorazepam (up to 4 mg/d as needed) in the 48 hours before the first ECT session was not associated with effects on seizure threshold or duration; however, larger lorazepam dosages were associated with briefer EEG seizure duration.31 Some patients may not tolerate withholding or tapering benzodiazepines in preparation for ECT. Studies investigating flumazenil for pre-ECT benzodiazepine reversal are lacking. One retrospective analysis showed that flumazenil administration immediately before and after ECT resulted in adequate seizures with no difference in clinical outcome compared with patients who were not receiving benzodiazepines or flumazenil.32

Tapering benzodiazepines

Slow discontinuation of benzodiazepines is recommended to avoid withdrawal symptoms, such as rebound anxiety, agitation, insomnia, or seizures, particularly when use exceeds 8 weeks. The onset of withdrawal symptoms varies, depending on the medication used. Withdrawal symptoms may appear in 1 to 2 days for agents with shorter half-lives, but may not appear until 3 to 7 days for agents with longer half-lives.33Table 2 lists recommended durations for tapering benzodiazepines.33,34 In general, decrease the total daily dose by 25% the first week, another 25% the second week, then 10% a week until discontinuation. When benzodiazepine use exceeds 1 year, a slower taper is recommended; for example, decrease 10% every 1 to 2 weeks. When 20% of the dosage remains, begin a 5% dose reduction every 2 to 4 weeks. Monitor patients for withdrawal symptoms or symptom exacerbation. If either occur, consider maintaining the current benzodiazepine dose or increasing the dose for 1 to 2 weeks or longer, if necessary, then continue to taper at a slower rate.34

Table 2

Recommendations for tapering benzodiazepines

Duration of useRecommended taper lengthComments
<6 to 8 weeksTaper may not be requiredDepending on clinical judgment and patient stability/preference, consider implementing a taper, particularly if using a high-dose benzodiazepine or an agent with a short or intermediate half-life, such as alprazolam or triazolam
8 weeks to 6 monthsSlowly over 2 to 3 weeksGo slower during latter half of taper. Tapering will reduce, not eliminate, withdrawal symptoms. Patients should avoid alcohol and stimulants during benzodiazepine withdrawal
6 months to 1 yearSlowly over 4 to 8 weeks
>1 yearSlowly over 2 to 4 months
Source: References 33,34
 

 

Risks of benzodiazepine use

For most indications, benzodiazepine therapy should be short-term.35 Use exceeding 2 to 4 weeks increases the risk for dependence and withdrawal. Tell patients to avoid alcohol while taking a benzodiazepine because this combination is potentially lethal. Benzodiazepines are commonly abused and abuse can lead to unintentional drug overdose. Benzodiazepines accounted for 37% of unintentional drug overdose deaths in West Virginia in 2006; in 46% of these cases, benzodiazepines were used for nonmedical purposes. Clinicians can help reduce the risk of diversion by limiting prescriptions to 30 days with no refills.36

Older patients taking benzodiazepines are at increased risk of falls and hip fractures.37 Lorazepam, oxazepam, and temazepam—agents with shorter half-lives that are not greatly affected by pharmacokinetic changes associated with aging—are preferred for these patients.34 Patients with dementia or other CNS-compromising conditions may become confused or delirious with regular benzodiazepine dosing. Educate patients to whom you prescribe benzodiazepines about the importance of gauging their level of sedation before driving or engaging in other tasks for which sedation could compromise their safety. Benzodiazepine use during pregnancy requires a careful discussion of risks and benefits (Box 2).38

Box 2

Using benzodiazepines during pregnancy

Benzodiazepine use during pregnancy has been associated with cleft palate and urogenital and neurologic malformations in the fetus.38 Although data are conflicting—particularly among recent meta-analyses that fail to demonstrate an association—some experts advise against benzodiazepine use in the first trimester. Participate in shared decision making with your patients and educate them about the potential risks and benefits of benzodiazepine use during the first trimester and throughout pregnancy. After delivery, newborns may develop “floppy baby syndrome”—which is associated with lethargy, difficulty eating, and respiratory depression—or withdrawal.38 To minimize this risk, consider tapering the benzodiazepine as the patient approaches delivery.

Related Resources

Drug Brand Names

  • Alprazolam • Xanax
  • Chlordiazepoxide • Librium, Limbitrol
  • Clonazepam • Klonopin
  • Clorazepate • Tranxene
  • Diazepam • Valium
  • Diphenhydramine • Benadryl, others
  • Estazolam • ProSom
  • Flumazenil • Romazicon
  • Flurazepam • Dalmane
  • Haloperidol • Haldol
  • Lithium • Lithobid
  • Lorazepam • Ativan
  • Olanzapine • Zyprexa
  • Oxazepam • Serax
  • Paroxetine • Paxil
  • Pregabalin • Lyrica
  • Propranolol • Inderal, InnoPran XL, others
  • Quazepam • Doral
  • Ramelteon • Rozerem
  • Sertraline • Zoloft
  • Temazepam • Restoril
  • Triazolam • Halcion
  • Valproic acid • Depakene, Stavzor, others

Disclosures

Drs. Bostwick and Yasugi report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Casher is a speaker for AstraZeneca and Sunovion Pharmaceuticals.

References

1. Mihic SJ, Harris RA. Hypnotics and sedatives. In: Brunton LL Chabner BA, Knollmann BC, eds. Goodman & Gilman’s the pharmacological basis of therapeutics. New York, NY: McGraw Hill and Company; 2011:457-480.

2. Facts and comparisons Web site. 2011 Wolters Kluwer Health Inc. http://online.factsandcomparisons.com. Accessed August 16, 2011.

3. DuPont RL, Greene W, Lydiard RB. Sedatives and hypnotics: pharmacology and epidemiology. In: Gold MS Hermann R, eds. UpToDate. http://www.uptodate.com/contents/sedatives-and-hypnotics-abuse-and-dependence-pharmacology-and-epidemiology. Accessed August 16, 2011.

4. U.S. Food and Drug Administration. Orange book: approved drug products with therapeutic equivalence evaluations. http://www.accessdata.fda.gov/scripts/cder/ob/default.cfm. Accessed August 16, 2011.

5. Chouinard G. Issues in the clinical use of benzodiazepines: potency withdrawal, and rebound. J Clin Psychiatry. 2004;65(suppl 5):7-12.

6. Shader RI, Greenblatt DJ. Can you provide a table of equivalencies for benzodiazepines and other marketed benzodiazepine receptor agonists? J Clin Psychopharmacol. 1997;17(4):331.-

7. Amato L, Minozzi S, Davoli M. Efficacy and safety of pharmacologic interventions for the treatment of the alcohol withdrawal syndrome. Cochrane Database Syst Rev. 2011;15(6):CD008537.-

8. Schutte-Rodin S, Broch L, Buysse D, et al. Clinical guideline for the evaluation and management of chronic insomnia in adults. J Clin Sleep Med. 2008;4(5):487-504.

9. Foral P, Dewan N, Malesker M. Insomnia: a therapeutic review for pharmacists. Consult Pharm. 2011;26(5):332-341.

10. Riemann D, Perlis ML. The treatments of chronic insomnia: a review of benzodiazepine receptor agonists and psychological and behavioral therapies. Sleep Med Rev. 2009;13(3):205-214.

11. Hidalgo RB, Tupler LA, Davidson JR. An effect-size analysis of pharmacologic treatments of generalized anxiety disorder. J Psychopharmacol. 2007;21(8):864-872.

12. Davidson JR, Zhang W, Connor KM, et al. A psychopharmacological treatment algorithm for generalised anxiety disorder (GAD). J Psychopharmacol. 2010;24(1):3-26.

13. Bandelow B, Zohar J, Hollander E, et al. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the pharmacological treatment of anxiety, obsessive-compulsive and post-traumatic stress disorders - first revision. World J Biol Psychiatry. 2008;9(4):248-312.

14. American Psychiatric Association. Practice guideline for the treatment of patients with panic disorder. 2nd ed. Arlington VA: American Psychiatric Publishing, Inc.; 2009.

15. Pollack MH, Simon NM, Worthington JJ, et al. Combined paroxetine and clonazepam treatment strategies compared to paroxetine monotherapy for panic disorder. J Psychopharmacol. 2003;17(3):276-282.

16. Watanabe N, Churchill R, Furukawa TA. Combined psychotherapy plus benzodiazepines for panic disorder. Cochrane Database Syst Rev. 2009;(1):CD005335.-

17. Otto MW, McHugh RK, Simon NM, et al. Efficacy of CBT for benzodiazepine discontinuation in patients with panic disorder: further evaluation. Behav Res Ther. 2010;48(8):720-727.

18. Davidson JR. Use of benzodiazepines in social anxiety disorder generalized anxiety disorder, and posttraumatic stress disorder. J Clin Psychiatry. 2004;65(suppl 5):29-33.

19. Argyropoulos SV, Sandford JJ, Nutt DJ. The psychobiology of anxiolytic drugs Part 2: pharmacological treatments of anxiety. Pharmacol Ther. 2000;88(3):213-227.

20. Connor KM, Davidson JR, Potts NL, et al. Discontinuation of clonazepam in the treatment of social phobia. J Clin Psychopharmacol. 1998;18(5):373-378.

21. Miller CH, Fleischhacker WW. Managing antipsychotic-induced acute and chronic akathisia. Drug Saf. 2000;22(1):73-81.

22. Rodnitzky RL. Drug-induced movement disorders. Clin Neuropharmacol. 2002;25(3):142-151.

23. Arbaizar B, Gómez-Acebo I, Llorca J. Postural induced tremor in psychiatry. Psychiatry Clin Neurosci. 2008;62(6):638-645.

24. Casher MI, Bess JD. Manual of inpatient psychiatry. New York NY: Cambridge University Press; 2010.

25. Battaglia J. Pharmacological management of acute agitation. Drugs. 2005;65(9):1207-1222.

26. Physicians’ desk reference. Montvale NJ: PDR Network, LLC; 2010.

27. Rosebush PI, Mazurek MF. Catatonia and its treatment. Schizophr Bull. 2010;36(2):239-242.

28. Ungvari GS, Kau LS, Wai-Kwong T, et al. The pharmacological treatment of catatonia: an overview. Eur Arch Psychiatry Clin Neurosci. 2001;251(suppl 1):I31-I34.

29. Fink M, Taylor MA. Catatonia: a clinician’s guide to diagnosis and treatment. New York NY: Cambridge University Press; 2003.

30. Naguib N, Koorn R. Interactions between psychotropics anaesthetics and electroconvulsive therapy: implications for drug choice and patient management. CNS Drugs. 2002;16(4):229-247.

31. Boylan LS, Haskett RF, Mulsant BH, et al. Determinants of seizure threshold in ECT: benzodiazepine use, anesthetic dosage, and other factors. J ECT. 2000;16(1):3-18.

32. Krystal AD, Watts BV, Weiner RD, et al. The use of flumazenil in the anxious and benzodiazepine-dependent ECT patient. J ECT. 1998;14(1):5-14.

33. Melton ST, Kirkwood CK. Anxiety disorders I: generalized anxiety panic, and social anxiety disorders. In: DiPiro JT, Talbert RL, Yee GC, et al, eds. Pharmacotherapy: a pathophysiologic approach. New York, NY: McGraw-Hill Companies; 2011:1209-1228.

34. Benzodiazepine toolkit. The Pharmacist’s Letter/Prescriber’s Letter. 2011;27(4):270406.-

35. Lader M. Benzodiazepines revisited – will we ever learn? Addiction. 2011;106(12):2086-2109.

36. Toblin RL, Paulozzi LJ, Logan JE, et al. Mental illness and psychotropic drug use among prescription drug overdose deaths: a medical examiner chart review. J Clin Psychiatry. 2010;71(4):491-496.

37. Ashton H. The diagnosis and management of benzodiazepine dependence. Curr Opin Psychiatry. 2005;18(3):249-255.

38. Menon SJ. Psychotropic medication during pregnancy and lactation. Arch Gynecol Obstet. 2008;277(1):1-13.

Article PDF
Author and Disclosure Information

Jolene R. Bostwick, PharmD, BCPS, BCPP
Clinical Assistant Professor of Pharmacy, University of Michigan College of Pharmacy, Clinical Pharmacist, University of Michigan Health System, Ann Arbor, MI
Michael I. Casher, MD
Clinical Assistant Professor, Department of Psychiatry, University of Michigan Medical School, Director of Inpatient Adult Psychiatry, University of Michigan Health System, Ann Arbor, MI
Shinji Yasugi, MD
First-Year Psychiatry Resident, University of Michigan Health System, Ann Arbor, MI

Audio / Podcast
Issue
Current Psychiatry - 11(04)
Publications
Topics
Page Number
54-64
Legacy Keywords
Benzodiazepines; alcohol withdrawal; insomnia; anxiety disorders; panic disorder; akathisia; catatonia; agitation
Sections
Audio / Podcast
Audio / Podcast
Author and Disclosure Information

Jolene R. Bostwick, PharmD, BCPS, BCPP
Clinical Assistant Professor of Pharmacy, University of Michigan College of Pharmacy, Clinical Pharmacist, University of Michigan Health System, Ann Arbor, MI
Michael I. Casher, MD
Clinical Assistant Professor, Department of Psychiatry, University of Michigan Medical School, Director of Inpatient Adult Psychiatry, University of Michigan Health System, Ann Arbor, MI
Shinji Yasugi, MD
First-Year Psychiatry Resident, University of Michigan Health System, Ann Arbor, MI

Author and Disclosure Information

Jolene R. Bostwick, PharmD, BCPS, BCPP
Clinical Assistant Professor of Pharmacy, University of Michigan College of Pharmacy, Clinical Pharmacist, University of Michigan Health System, Ann Arbor, MI
Michael I. Casher, MD
Clinical Assistant Professor, Department of Psychiatry, University of Michigan Medical School, Director of Inpatient Adult Psychiatry, University of Michigan Health System, Ann Arbor, MI
Shinji Yasugi, MD
First-Year Psychiatry Resident, University of Michigan Health System, Ann Arbor, MI

Article PDF
Article PDF

Since the discovery of chlordiazepoxide in the 1950s, benzodiazepines have revolutionized the treatment of anxiety and insomnia, largely because of their improved safety profile compared with barbiturates, formerly the preferred sedative-hypnotic.1 In addition to their anxiolytic and sedative-hypnotic effects, benzodiazepines exhibit anterograde amnesia, anticonvulsant, and muscle relaxant properties.1 Psychiatrists use benzodiazepines to treat anxiety and sleep disorders, acute agitation, alcohol withdrawal, catatonia, and psychotropic side effects such as akathisia. This article highlights the evidence for using benzodiazepines in anxiety and other disorders and why they generally should not be used for obsessive-compulsive disorder and posttraumatic stress disorder (Box 1).

Box 1

When not to use benzodiazepines: OCD and PTSD

Current evidence indicates little support for using benzodiazepines for obsessive-compulsive disorder (OCD). The American Psychiatric Association (APA) and the World Federation of Biological Psychiatry do not recommend benzodiazepines for treating OCD because of a lack of evidence for efficacy.a,b An earlier study suggested clonazepam monotherapy was effective for OCDc; however, a more recent study did not show a benefit on rate of response or degree of symptom improvement.d Augmentation strategies with benzodiazepines also do not appear to be beneficial for OCD management. A recent double-blind, placebo-controlled study failed to demonstrate faster symptom improvement by augmenting sertraline with clonazepam, although the study had a small sample size and high drop-out rate.e

Because benzodiazepines have negligible action on core posttraumatic stress disorder (PTSD) symptoms (re-experiencing, avoidance, and hyperarousal), selective serotonin reuptake inhibitors and other agents largely have supplanted them for PTSD treatment.f Use of benzodiazepines for PTSD is associated with withdrawal symptoms, more severe symptoms after discontinuation, and possible disinhibition, and may interfere with patients’ efforts to integrate trauma experiences. Although benzodiazepines may reduce distress associated with acute trauma, there is evidence—in clinical studies and animal models—that early benzodiazepine administration fails to prevent PTSD and may increase its incidence.g The International Consensus Group on Depression and Anxiety, the APA, and the British Association for Psychopharmacology all highlight the limited role, if any, for benzodiazepines in PTSD.h-j

References

  1. Bandelow B, Zohar J, Hollander E, et al. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the pharmacological treatment of anxiety, obsessive-compulsive and post-traumatic stress disorders - first revision. World J Biol Psychiatry. 2008;9(4):248-312.
  2. American Psychiatric Association. Practice guideline for the treatment of patients with obsessive-compulsive disorder. Arlington, VA: American Psychiatric Publishing, Inc.; 2007.
  3. Hewlett WA, Vinogradov S, Agras WS. Clomipramine, clonazepam, and clonidine treatment of obsessive compulsive disorder. J Clin Psychopharmacol. 1992;12(6):420-430.
  4. Hollander E, Kaplan A, Stahl SM. A double-blind, placebo-controlled trial of clonazepam in obsessive-compulsive disorder. World J Biol Psychiatry. 2003;4(1):30-34.
  5. Crockett BA, Churchill E, Davidson JR. A double-blind combination study of clonazepam and sertraline in OCD. Ann Clin Psychiatry. 2004;16(3):127-132.
  6. Argyropoulos SV, Sandford JJ, Nutt DJ. The psychobiology of anxiolytic drugs. Part 2: pharmacological treatments of anxiety. Pharmacol Ther. 2000;88(3):213-227.
  7. Matar MA, Zohar J, Kaplan Z, et al. Alprazolam treatment immediately after stress exposure interferes with the normal HPA-stress response and increases vulnerability to subsequent stress in an animal model of PTSD. Eur Neuropsychopharmacol. 2009;19(4):283-295.
  8. Ballenger JC, Davidson JR, Lecrubier Y, et al. Consensus statement update on posttraumatic stress disorder from the international consensus group on depression and anxiety. J Clin Psychiatry. 2004;65(suppl 1):55-62.
  9. Ursano RJ, Bell C, Eth S, et al. Practice guideline for the treatment of patients with acute stress disorder and posttraumatic stress disorder. Am J Psychiatry. 2004;161(11 suppl):3-31.
  10. Baldwin DS, Anderson IM, Nutt DJ, et al. Evidence-based guidelines for the pharmacological treatment of anxiety disorders: recommendations from the British Association for Psychopharmacology. J Psychopharmacol. 2005;19(6):567-596.

Pharmacokinetic properties

Most benzodiazepines are considered to have similar efficacy; therefore, selection is based on pharmacokinetic considerations. Table 1 compares the indication, onset, and half-life of 12 commonly used benzodiazepines.2-6 Although Table 1 lists approximate equivalent doses, studies report inconsistent data. These are approximations only and should not be used independently to make therapy decisions.

Table 1

Oral benzodiazepines: Indications, onset, half-life, and equivalent doses

DrugFDA-approved indication(s)Onset of actionApproximate half-life (hours) in healthy adultsApproximate equivalent dose (mg)aComments
AlprazolamAnxiety disorders, panic disorderIntermediate6.3 to 26.9 (IR), 10.7 to 15.8 (XR)0.5Increased risk for abuse because of greater lipid solubility
ChlordiazepoxideAnxiety disorders, acute alcohol withdrawal, preoperative apprehension and anxietyIntermediate24 to 4810Risk for accumulation because of long-acting metabolites (desmethyldiazepam, oxazepam)
ClonazepamSeizure disorders, panic disorderIntermediate18 to 500.25 to 0.5Use caution in patients with liver disease
ClorazepateAnxiety, seizures, acute alcohol withdrawalFast40 to 507.5Risk for accumulation because of long-acting metabolites (desmethyldiazepam, oxazepam)
DiazepamAnxiety disorders, acute alcohol withdrawal, muscle spasms, convulsive disordersVery fast20 to 1005Risk for accumulation because of long-acting metabolites (temazepam, desmethyldiazepam, oxazepam). Increased risk for abuse because of quick onset
EstazolamInsomniaIntermediate10 to 240.3 to 2None
FlurazepamInsomniaIntermediate47 to 10030Avoid in geriatric patients or patients with liver impairment
LorazepamAnxietyIntermediate10 to 201Preferred for patients with liver impairment and geriatric patients
OxazepamAnxiety, acute alcohol withdrawalSlow to intermediate5 to 2030Preferred for patients with liver impairment and geriatric patients
QuazepamInsomniaIntermediate39 to 735 to 15Risk for accumulation because of long-acting metabolites (desmethyldiazepam, oxazepam)
TemazepamInsomniaIntermediate3.5 to 18.430Preferred for patients with liver impairment and geriatric patients
TriazolamInsomniaFast1.5 to 5.50.25Lacks active metabolites
IR: immediate release; XR: extended release
aInterpret with caution, conflicting data exist
Source: References 2-6
 

 

A diverse range of indications

Alcohol withdrawal. Benzodiazepines are the treatment of choice for alcohol withdrawal syndrome, particularly to prevent seizures.7 Research supports symptom-triggered therapy using the revised Clinical Institute Withdrawal Assessment for Alcohol. Benzodiazepines reduce CNS sympathetic hyperactivity to mitigate withdrawal from alcohol by decreasing tachycardia, tremor, insomnia, agitation, and anxiety. Furthermore, these agents provide prophylaxis against serious sequelae such as seizures and delirium.

Insomnia. The American Academy of Sleep Medicine considers benzodiazepine receptor agonists (BzRAs, which include benzodiazepines and non-benzodiazepines) and ramelteon first-line pharmacotherapy for primary insomnia.8 However, pharmacologic treatment should be short-term. Agents with short to intermediate half-lives and rapid onset, such as triazolam, can aid sleep initiation. Those with longer half-lives, such as temazepam, could address sleep maintenance. If a patient does not respond to the initial agent, try another medication within the same class, because patients may respond differently. Use lower starting doses in geriatric patients.9 Closely monitor for adverse effects, rebound insomnia, and potential abuse or tolerance. Identify comorbid conditions and medications that may impair sleep, and address them accordingly.

Psychological and behavioral treatments given over 4 to 8 weeks can yield stable sleep improvements for up to 2 years. If available, these interventions may be considered first-line for treating insomnia because of their lasting effects compared with BzRAs.10

Generalized anxiety disorder (GAD). Benzodiazepines effectively treat GAD because they work quickly and are well tolerated. However, there are better first-line treatment options when considering efficacy studies and dependence and tolerance concerns. One effect-size comparison of 21 double-blind, placebo-controlled trials showed that the efficacy of selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), and pregabalin are comparable to benzodiazepines.11 Benzodiazepines can be used in the first 2 to 3 weeks after initiating antidepressants to alleviate and prevent worsening of anxiety that may occur at the start of antidepressant therapy. Recent treatment guidelines recommend benzodiazepines as a second-line treatment or for treatment-resistant GAD in patients who do not have a substance abuse history.12,13

Panic disorder. Efficacy of benzodiazepines for panic disorder is comparable to SSRIs, SNRIs, and tricyclic antidepressants (TCAs). SSRIs and SNRIs are considered first-line treatments for panic disorder because of their favorable side effect profile.14 In practice, benzodiazepines often are combined with SSRIs, SNRIs, or TCAs. A randomized controlled trial demonstrated that paroxetine and clonazepam (mean dose 1.6 mg/d at 5 weeks) resulted in a more rapid response compared with paroxetine alone, although this difference lasted only a few weeks.15 Furthermore, this study suggested that brief treatment with clonazepam followed by a taper is as effective as sustained treatment with paroxetine and clonazepam.15

There is a lack of high-quality data on combining cognitive-behavioral therapy (CBT) and benzodiazepines for panic disorder, although a Cochrane Review found that adding a benzodiazepine to CBT did not lead to a significant difference in response compared with psychotherapy alone.16 A recent randomized controlled trial demonstrated that tapering benzodiazepines combined with CBT was associated with successful discontinuation of the drug and prevented return of panic symptoms.17

Social anxiety. A meta-analysis found that for treating social anxiety, benzodiazepines have better efficacy than SSRIs, monoamine oxidase inhibitors, and anticonvulsants.18 Longer-acting benzodiazepines may be more effective than shorter-acting agents. One study of patients with social anxiety showed a 38% response rate for alprazolam vs 20% for placebo over 12 weeks, and a similar 10-week study demonstrated a 73% recovery rate with clonazepam vs 22% for placebo.19 In addition, studies have observed that patients can be maintained on clonazepam for up to 2 years without symptom relapse and will tolerate slow-taper discontinuation.18,20 Sedation and drowsiness can be lessened by limiting clonazepam doses to 2 to 3 mg/d.

Akathisia and tremor. Akathisia, a syndrome of motor restlessness and inner turmoil, is associated with antipsychotics but can occur with SSRIs. Reducing the dosage or switching to another, usually less potent agent often can relieve akathisia. When these remedies are not tenable, consider benzodiazepines along with other medications—including beta blockers and anticholinergic agents—with demonstrated efficacy in reducing akathisia symptoms. Lorazepam, diazepam, and clonazepam have demonstrated efficacy for relieving akathisia in comparison studies with placebo, propranolol, and diphenhydramine.21,22

Drug-induced postural tremor can occur with several psychotropics, including lithium, valproic acid, antidepressants, and antipsychotics. A tremor is considered mild if a patient can drink a glass of water with 1 hand without spilling and severe if holding a glass with 2 hands is difficult. Propranolol is most commonly prescribed for these tremors, but alprazolam and clonazepam have demonstrated efficacy, either as monotherapy or coadministered with a beta blocker.23

 

 

Acute agitation. Agitated patients often have acute psychosis and/or mania or dyscontrol secondary to axis II disorders.24 Patients may be paranoid, hostile, disruptive, and combative. Rapidly initiating medication can prevent the need for more restrictive measures, such as seclusion or restraint. Antipsychotics—especially high-potency agents such as haloperidol—and benzodiazepines, as monotherapy or in combination, are a mainstay treatment. Although treatment protocols favor atypical antipsychotics over typical antipsychotics, benzodiazepines are a viable option because of their anxiolytic and sedative effects. Advantages of benzodiazepine monotherapy include decreased extrapyramidal symptoms, greater patient acceptance/preference, and increased sedation compared with antipsychotics. Lorazepam, 1 to 2 mg intramuscularly (IM) or orally, is well tolerated because of its favorable drug-drug interaction profile and lack of significant cardiac side effects. Benzodiazepines can cause respiratory depression in patients with chronic lung disease and additive sedation secondary to opiates, other sedatives/hypnotics, or alcohol. Behavioral disinhibition is rare and is associated with preexisting CNS pathology or mental retardation.25 The IM olanzapine package insert warns against coadministering IM lorazepam because of additive cardiorespiratory depressive effects and excessive somnolence.26

Catatonia. The characteristic symptoms of catatonia are immobility, negativism, muteness, and failure to eat or drink. Benzodiazepines improve these symptoms in approximately 70% to 80% of catatonic patients with affective disorders. Response rates are lower in catatonia in patients with schizophrenia.27 If catatonia in a patient with psychosis is missed, giving antipsychotics before benzodiazepines may worsen catatonic symptoms or precipitate neuroleptic malignant syndrome in some cases. When you suspect a patient has catatonia, start with lorazepam, 1 to 2 mg IV or IM, and examine the patient for diminishing catatonic signs within 1 to 2 hours. If catatonia signs lessen, begin regularly scheduled lorazepam, with dosing varying by age—be more cautious in geriatric patients—and symptom severity. Titrate benzodiazepines for stuporous patients more slowly (eg, 1 mg 3 times a day as a starting dose) than for excited catatonic patients. Lorazepam can be increased gradually as tolerated; it is not unusual for patients to require up to 8 to 12 mg/d. Electroconvulsive therapy (ECT) is the treatment of choice when catatonic patients respond poorly or partially to high-dose benzodiazepines.28,29

Benzodiazepine reversal for ECT

Benzodiazepines have anticonvulsant properties that may interfere with the therapeutic efficacy of ECT.30 A multi-center study demonstrated that lorazepam (up to 4 mg/d as needed) in the 48 hours before the first ECT session was not associated with effects on seizure threshold or duration; however, larger lorazepam dosages were associated with briefer EEG seizure duration.31 Some patients may not tolerate withholding or tapering benzodiazepines in preparation for ECT. Studies investigating flumazenil for pre-ECT benzodiazepine reversal are lacking. One retrospective analysis showed that flumazenil administration immediately before and after ECT resulted in adequate seizures with no difference in clinical outcome compared with patients who were not receiving benzodiazepines or flumazenil.32

Tapering benzodiazepines

Slow discontinuation of benzodiazepines is recommended to avoid withdrawal symptoms, such as rebound anxiety, agitation, insomnia, or seizures, particularly when use exceeds 8 weeks. The onset of withdrawal symptoms varies, depending on the medication used. Withdrawal symptoms may appear in 1 to 2 days for agents with shorter half-lives, but may not appear until 3 to 7 days for agents with longer half-lives.33Table 2 lists recommended durations for tapering benzodiazepines.33,34 In general, decrease the total daily dose by 25% the first week, another 25% the second week, then 10% a week until discontinuation. When benzodiazepine use exceeds 1 year, a slower taper is recommended; for example, decrease 10% every 1 to 2 weeks. When 20% of the dosage remains, begin a 5% dose reduction every 2 to 4 weeks. Monitor patients for withdrawal symptoms or symptom exacerbation. If either occur, consider maintaining the current benzodiazepine dose or increasing the dose for 1 to 2 weeks or longer, if necessary, then continue to taper at a slower rate.34

Table 2

Recommendations for tapering benzodiazepines

Duration of useRecommended taper lengthComments
<6 to 8 weeksTaper may not be requiredDepending on clinical judgment and patient stability/preference, consider implementing a taper, particularly if using a high-dose benzodiazepine or an agent with a short or intermediate half-life, such as alprazolam or triazolam
8 weeks to 6 monthsSlowly over 2 to 3 weeksGo slower during latter half of taper. Tapering will reduce, not eliminate, withdrawal symptoms. Patients should avoid alcohol and stimulants during benzodiazepine withdrawal
6 months to 1 yearSlowly over 4 to 8 weeks
>1 yearSlowly over 2 to 4 months
Source: References 33,34
 

 

Risks of benzodiazepine use

For most indications, benzodiazepine therapy should be short-term.35 Use exceeding 2 to 4 weeks increases the risk for dependence and withdrawal. Tell patients to avoid alcohol while taking a benzodiazepine because this combination is potentially lethal. Benzodiazepines are commonly abused and abuse can lead to unintentional drug overdose. Benzodiazepines accounted for 37% of unintentional drug overdose deaths in West Virginia in 2006; in 46% of these cases, benzodiazepines were used for nonmedical purposes. Clinicians can help reduce the risk of diversion by limiting prescriptions to 30 days with no refills.36

Older patients taking benzodiazepines are at increased risk of falls and hip fractures.37 Lorazepam, oxazepam, and temazepam—agents with shorter half-lives that are not greatly affected by pharmacokinetic changes associated with aging—are preferred for these patients.34 Patients with dementia or other CNS-compromising conditions may become confused or delirious with regular benzodiazepine dosing. Educate patients to whom you prescribe benzodiazepines about the importance of gauging their level of sedation before driving or engaging in other tasks for which sedation could compromise their safety. Benzodiazepine use during pregnancy requires a careful discussion of risks and benefits (Box 2).38

Box 2

Using benzodiazepines during pregnancy

Benzodiazepine use during pregnancy has been associated with cleft palate and urogenital and neurologic malformations in the fetus.38 Although data are conflicting—particularly among recent meta-analyses that fail to demonstrate an association—some experts advise against benzodiazepine use in the first trimester. Participate in shared decision making with your patients and educate them about the potential risks and benefits of benzodiazepine use during the first trimester and throughout pregnancy. After delivery, newborns may develop “floppy baby syndrome”—which is associated with lethargy, difficulty eating, and respiratory depression—or withdrawal.38 To minimize this risk, consider tapering the benzodiazepine as the patient approaches delivery.

Related Resources

Drug Brand Names

  • Alprazolam • Xanax
  • Chlordiazepoxide • Librium, Limbitrol
  • Clonazepam • Klonopin
  • Clorazepate • Tranxene
  • Diazepam • Valium
  • Diphenhydramine • Benadryl, others
  • Estazolam • ProSom
  • Flumazenil • Romazicon
  • Flurazepam • Dalmane
  • Haloperidol • Haldol
  • Lithium • Lithobid
  • Lorazepam • Ativan
  • Olanzapine • Zyprexa
  • Oxazepam • Serax
  • Paroxetine • Paxil
  • Pregabalin • Lyrica
  • Propranolol • Inderal, InnoPran XL, others
  • Quazepam • Doral
  • Ramelteon • Rozerem
  • Sertraline • Zoloft
  • Temazepam • Restoril
  • Triazolam • Halcion
  • Valproic acid • Depakene, Stavzor, others

Disclosures

Drs. Bostwick and Yasugi report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Casher is a speaker for AstraZeneca and Sunovion Pharmaceuticals.

Since the discovery of chlordiazepoxide in the 1950s, benzodiazepines have revolutionized the treatment of anxiety and insomnia, largely because of their improved safety profile compared with barbiturates, formerly the preferred sedative-hypnotic.1 In addition to their anxiolytic and sedative-hypnotic effects, benzodiazepines exhibit anterograde amnesia, anticonvulsant, and muscle relaxant properties.1 Psychiatrists use benzodiazepines to treat anxiety and sleep disorders, acute agitation, alcohol withdrawal, catatonia, and psychotropic side effects such as akathisia. This article highlights the evidence for using benzodiazepines in anxiety and other disorders and why they generally should not be used for obsessive-compulsive disorder and posttraumatic stress disorder (Box 1).

Box 1

When not to use benzodiazepines: OCD and PTSD

Current evidence indicates little support for using benzodiazepines for obsessive-compulsive disorder (OCD). The American Psychiatric Association (APA) and the World Federation of Biological Psychiatry do not recommend benzodiazepines for treating OCD because of a lack of evidence for efficacy.a,b An earlier study suggested clonazepam monotherapy was effective for OCDc; however, a more recent study did not show a benefit on rate of response or degree of symptom improvement.d Augmentation strategies with benzodiazepines also do not appear to be beneficial for OCD management. A recent double-blind, placebo-controlled study failed to demonstrate faster symptom improvement by augmenting sertraline with clonazepam, although the study had a small sample size and high drop-out rate.e

Because benzodiazepines have negligible action on core posttraumatic stress disorder (PTSD) symptoms (re-experiencing, avoidance, and hyperarousal), selective serotonin reuptake inhibitors and other agents largely have supplanted them for PTSD treatment.f Use of benzodiazepines for PTSD is associated with withdrawal symptoms, more severe symptoms after discontinuation, and possible disinhibition, and may interfere with patients’ efforts to integrate trauma experiences. Although benzodiazepines may reduce distress associated with acute trauma, there is evidence—in clinical studies and animal models—that early benzodiazepine administration fails to prevent PTSD and may increase its incidence.g The International Consensus Group on Depression and Anxiety, the APA, and the British Association for Psychopharmacology all highlight the limited role, if any, for benzodiazepines in PTSD.h-j

References

  1. Bandelow B, Zohar J, Hollander E, et al. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the pharmacological treatment of anxiety, obsessive-compulsive and post-traumatic stress disorders - first revision. World J Biol Psychiatry. 2008;9(4):248-312.
  2. American Psychiatric Association. Practice guideline for the treatment of patients with obsessive-compulsive disorder. Arlington, VA: American Psychiatric Publishing, Inc.; 2007.
  3. Hewlett WA, Vinogradov S, Agras WS. Clomipramine, clonazepam, and clonidine treatment of obsessive compulsive disorder. J Clin Psychopharmacol. 1992;12(6):420-430.
  4. Hollander E, Kaplan A, Stahl SM. A double-blind, placebo-controlled trial of clonazepam in obsessive-compulsive disorder. World J Biol Psychiatry. 2003;4(1):30-34.
  5. Crockett BA, Churchill E, Davidson JR. A double-blind combination study of clonazepam and sertraline in OCD. Ann Clin Psychiatry. 2004;16(3):127-132.
  6. Argyropoulos SV, Sandford JJ, Nutt DJ. The psychobiology of anxiolytic drugs. Part 2: pharmacological treatments of anxiety. Pharmacol Ther. 2000;88(3):213-227.
  7. Matar MA, Zohar J, Kaplan Z, et al. Alprazolam treatment immediately after stress exposure interferes with the normal HPA-stress response and increases vulnerability to subsequent stress in an animal model of PTSD. Eur Neuropsychopharmacol. 2009;19(4):283-295.
  8. Ballenger JC, Davidson JR, Lecrubier Y, et al. Consensus statement update on posttraumatic stress disorder from the international consensus group on depression and anxiety. J Clin Psychiatry. 2004;65(suppl 1):55-62.
  9. Ursano RJ, Bell C, Eth S, et al. Practice guideline for the treatment of patients with acute stress disorder and posttraumatic stress disorder. Am J Psychiatry. 2004;161(11 suppl):3-31.
  10. Baldwin DS, Anderson IM, Nutt DJ, et al. Evidence-based guidelines for the pharmacological treatment of anxiety disorders: recommendations from the British Association for Psychopharmacology. J Psychopharmacol. 2005;19(6):567-596.

Pharmacokinetic properties

Most benzodiazepines are considered to have similar efficacy; therefore, selection is based on pharmacokinetic considerations. Table 1 compares the indication, onset, and half-life of 12 commonly used benzodiazepines.2-6 Although Table 1 lists approximate equivalent doses, studies report inconsistent data. These are approximations only and should not be used independently to make therapy decisions.

Table 1

Oral benzodiazepines: Indications, onset, half-life, and equivalent doses

DrugFDA-approved indication(s)Onset of actionApproximate half-life (hours) in healthy adultsApproximate equivalent dose (mg)aComments
AlprazolamAnxiety disorders, panic disorderIntermediate6.3 to 26.9 (IR), 10.7 to 15.8 (XR)0.5Increased risk for abuse because of greater lipid solubility
ChlordiazepoxideAnxiety disorders, acute alcohol withdrawal, preoperative apprehension and anxietyIntermediate24 to 4810Risk for accumulation because of long-acting metabolites (desmethyldiazepam, oxazepam)
ClonazepamSeizure disorders, panic disorderIntermediate18 to 500.25 to 0.5Use caution in patients with liver disease
ClorazepateAnxiety, seizures, acute alcohol withdrawalFast40 to 507.5Risk for accumulation because of long-acting metabolites (desmethyldiazepam, oxazepam)
DiazepamAnxiety disorders, acute alcohol withdrawal, muscle spasms, convulsive disordersVery fast20 to 1005Risk for accumulation because of long-acting metabolites (temazepam, desmethyldiazepam, oxazepam). Increased risk for abuse because of quick onset
EstazolamInsomniaIntermediate10 to 240.3 to 2None
FlurazepamInsomniaIntermediate47 to 10030Avoid in geriatric patients or patients with liver impairment
LorazepamAnxietyIntermediate10 to 201Preferred for patients with liver impairment and geriatric patients
OxazepamAnxiety, acute alcohol withdrawalSlow to intermediate5 to 2030Preferred for patients with liver impairment and geriatric patients
QuazepamInsomniaIntermediate39 to 735 to 15Risk for accumulation because of long-acting metabolites (desmethyldiazepam, oxazepam)
TemazepamInsomniaIntermediate3.5 to 18.430Preferred for patients with liver impairment and geriatric patients
TriazolamInsomniaFast1.5 to 5.50.25Lacks active metabolites
IR: immediate release; XR: extended release
aInterpret with caution, conflicting data exist
Source: References 2-6
 

 

A diverse range of indications

Alcohol withdrawal. Benzodiazepines are the treatment of choice for alcohol withdrawal syndrome, particularly to prevent seizures.7 Research supports symptom-triggered therapy using the revised Clinical Institute Withdrawal Assessment for Alcohol. Benzodiazepines reduce CNS sympathetic hyperactivity to mitigate withdrawal from alcohol by decreasing tachycardia, tremor, insomnia, agitation, and anxiety. Furthermore, these agents provide prophylaxis against serious sequelae such as seizures and delirium.

Insomnia. The American Academy of Sleep Medicine considers benzodiazepine receptor agonists (BzRAs, which include benzodiazepines and non-benzodiazepines) and ramelteon first-line pharmacotherapy for primary insomnia.8 However, pharmacologic treatment should be short-term. Agents with short to intermediate half-lives and rapid onset, such as triazolam, can aid sleep initiation. Those with longer half-lives, such as temazepam, could address sleep maintenance. If a patient does not respond to the initial agent, try another medication within the same class, because patients may respond differently. Use lower starting doses in geriatric patients.9 Closely monitor for adverse effects, rebound insomnia, and potential abuse or tolerance. Identify comorbid conditions and medications that may impair sleep, and address them accordingly.

Psychological and behavioral treatments given over 4 to 8 weeks can yield stable sleep improvements for up to 2 years. If available, these interventions may be considered first-line for treating insomnia because of their lasting effects compared with BzRAs.10

Generalized anxiety disorder (GAD). Benzodiazepines effectively treat GAD because they work quickly and are well tolerated. However, there are better first-line treatment options when considering efficacy studies and dependence and tolerance concerns. One effect-size comparison of 21 double-blind, placebo-controlled trials showed that the efficacy of selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), and pregabalin are comparable to benzodiazepines.11 Benzodiazepines can be used in the first 2 to 3 weeks after initiating antidepressants to alleviate and prevent worsening of anxiety that may occur at the start of antidepressant therapy. Recent treatment guidelines recommend benzodiazepines as a second-line treatment or for treatment-resistant GAD in patients who do not have a substance abuse history.12,13

Panic disorder. Efficacy of benzodiazepines for panic disorder is comparable to SSRIs, SNRIs, and tricyclic antidepressants (TCAs). SSRIs and SNRIs are considered first-line treatments for panic disorder because of their favorable side effect profile.14 In practice, benzodiazepines often are combined with SSRIs, SNRIs, or TCAs. A randomized controlled trial demonstrated that paroxetine and clonazepam (mean dose 1.6 mg/d at 5 weeks) resulted in a more rapid response compared with paroxetine alone, although this difference lasted only a few weeks.15 Furthermore, this study suggested that brief treatment with clonazepam followed by a taper is as effective as sustained treatment with paroxetine and clonazepam.15

There is a lack of high-quality data on combining cognitive-behavioral therapy (CBT) and benzodiazepines for panic disorder, although a Cochrane Review found that adding a benzodiazepine to CBT did not lead to a significant difference in response compared with psychotherapy alone.16 A recent randomized controlled trial demonstrated that tapering benzodiazepines combined with CBT was associated with successful discontinuation of the drug and prevented return of panic symptoms.17

Social anxiety. A meta-analysis found that for treating social anxiety, benzodiazepines have better efficacy than SSRIs, monoamine oxidase inhibitors, and anticonvulsants.18 Longer-acting benzodiazepines may be more effective than shorter-acting agents. One study of patients with social anxiety showed a 38% response rate for alprazolam vs 20% for placebo over 12 weeks, and a similar 10-week study demonstrated a 73% recovery rate with clonazepam vs 22% for placebo.19 In addition, studies have observed that patients can be maintained on clonazepam for up to 2 years without symptom relapse and will tolerate slow-taper discontinuation.18,20 Sedation and drowsiness can be lessened by limiting clonazepam doses to 2 to 3 mg/d.

Akathisia and tremor. Akathisia, a syndrome of motor restlessness and inner turmoil, is associated with antipsychotics but can occur with SSRIs. Reducing the dosage or switching to another, usually less potent agent often can relieve akathisia. When these remedies are not tenable, consider benzodiazepines along with other medications—including beta blockers and anticholinergic agents—with demonstrated efficacy in reducing akathisia symptoms. Lorazepam, diazepam, and clonazepam have demonstrated efficacy for relieving akathisia in comparison studies with placebo, propranolol, and diphenhydramine.21,22

Drug-induced postural tremor can occur with several psychotropics, including lithium, valproic acid, antidepressants, and antipsychotics. A tremor is considered mild if a patient can drink a glass of water with 1 hand without spilling and severe if holding a glass with 2 hands is difficult. Propranolol is most commonly prescribed for these tremors, but alprazolam and clonazepam have demonstrated efficacy, either as monotherapy or coadministered with a beta blocker.23

 

 

Acute agitation. Agitated patients often have acute psychosis and/or mania or dyscontrol secondary to axis II disorders.24 Patients may be paranoid, hostile, disruptive, and combative. Rapidly initiating medication can prevent the need for more restrictive measures, such as seclusion or restraint. Antipsychotics—especially high-potency agents such as haloperidol—and benzodiazepines, as monotherapy or in combination, are a mainstay treatment. Although treatment protocols favor atypical antipsychotics over typical antipsychotics, benzodiazepines are a viable option because of their anxiolytic and sedative effects. Advantages of benzodiazepine monotherapy include decreased extrapyramidal symptoms, greater patient acceptance/preference, and increased sedation compared with antipsychotics. Lorazepam, 1 to 2 mg intramuscularly (IM) or orally, is well tolerated because of its favorable drug-drug interaction profile and lack of significant cardiac side effects. Benzodiazepines can cause respiratory depression in patients with chronic lung disease and additive sedation secondary to opiates, other sedatives/hypnotics, or alcohol. Behavioral disinhibition is rare and is associated with preexisting CNS pathology or mental retardation.25 The IM olanzapine package insert warns against coadministering IM lorazepam because of additive cardiorespiratory depressive effects and excessive somnolence.26

Catatonia. The characteristic symptoms of catatonia are immobility, negativism, muteness, and failure to eat or drink. Benzodiazepines improve these symptoms in approximately 70% to 80% of catatonic patients with affective disorders. Response rates are lower in catatonia in patients with schizophrenia.27 If catatonia in a patient with psychosis is missed, giving antipsychotics before benzodiazepines may worsen catatonic symptoms or precipitate neuroleptic malignant syndrome in some cases. When you suspect a patient has catatonia, start with lorazepam, 1 to 2 mg IV or IM, and examine the patient for diminishing catatonic signs within 1 to 2 hours. If catatonia signs lessen, begin regularly scheduled lorazepam, with dosing varying by age—be more cautious in geriatric patients—and symptom severity. Titrate benzodiazepines for stuporous patients more slowly (eg, 1 mg 3 times a day as a starting dose) than for excited catatonic patients. Lorazepam can be increased gradually as tolerated; it is not unusual for patients to require up to 8 to 12 mg/d. Electroconvulsive therapy (ECT) is the treatment of choice when catatonic patients respond poorly or partially to high-dose benzodiazepines.28,29

Benzodiazepine reversal for ECT

Benzodiazepines have anticonvulsant properties that may interfere with the therapeutic efficacy of ECT.30 A multi-center study demonstrated that lorazepam (up to 4 mg/d as needed) in the 48 hours before the first ECT session was not associated with effects on seizure threshold or duration; however, larger lorazepam dosages were associated with briefer EEG seizure duration.31 Some patients may not tolerate withholding or tapering benzodiazepines in preparation for ECT. Studies investigating flumazenil for pre-ECT benzodiazepine reversal are lacking. One retrospective analysis showed that flumazenil administration immediately before and after ECT resulted in adequate seizures with no difference in clinical outcome compared with patients who were not receiving benzodiazepines or flumazenil.32

Tapering benzodiazepines

Slow discontinuation of benzodiazepines is recommended to avoid withdrawal symptoms, such as rebound anxiety, agitation, insomnia, or seizures, particularly when use exceeds 8 weeks. The onset of withdrawal symptoms varies, depending on the medication used. Withdrawal symptoms may appear in 1 to 2 days for agents with shorter half-lives, but may not appear until 3 to 7 days for agents with longer half-lives.33Table 2 lists recommended durations for tapering benzodiazepines.33,34 In general, decrease the total daily dose by 25% the first week, another 25% the second week, then 10% a week until discontinuation. When benzodiazepine use exceeds 1 year, a slower taper is recommended; for example, decrease 10% every 1 to 2 weeks. When 20% of the dosage remains, begin a 5% dose reduction every 2 to 4 weeks. Monitor patients for withdrawal symptoms or symptom exacerbation. If either occur, consider maintaining the current benzodiazepine dose or increasing the dose for 1 to 2 weeks or longer, if necessary, then continue to taper at a slower rate.34

Table 2

Recommendations for tapering benzodiazepines

Duration of useRecommended taper lengthComments
<6 to 8 weeksTaper may not be requiredDepending on clinical judgment and patient stability/preference, consider implementing a taper, particularly if using a high-dose benzodiazepine or an agent with a short or intermediate half-life, such as alprazolam or triazolam
8 weeks to 6 monthsSlowly over 2 to 3 weeksGo slower during latter half of taper. Tapering will reduce, not eliminate, withdrawal symptoms. Patients should avoid alcohol and stimulants during benzodiazepine withdrawal
6 months to 1 yearSlowly over 4 to 8 weeks
>1 yearSlowly over 2 to 4 months
Source: References 33,34
 

 

Risks of benzodiazepine use

For most indications, benzodiazepine therapy should be short-term.35 Use exceeding 2 to 4 weeks increases the risk for dependence and withdrawal. Tell patients to avoid alcohol while taking a benzodiazepine because this combination is potentially lethal. Benzodiazepines are commonly abused and abuse can lead to unintentional drug overdose. Benzodiazepines accounted for 37% of unintentional drug overdose deaths in West Virginia in 2006; in 46% of these cases, benzodiazepines were used for nonmedical purposes. Clinicians can help reduce the risk of diversion by limiting prescriptions to 30 days with no refills.36

Older patients taking benzodiazepines are at increased risk of falls and hip fractures.37 Lorazepam, oxazepam, and temazepam—agents with shorter half-lives that are not greatly affected by pharmacokinetic changes associated with aging—are preferred for these patients.34 Patients with dementia or other CNS-compromising conditions may become confused or delirious with regular benzodiazepine dosing. Educate patients to whom you prescribe benzodiazepines about the importance of gauging their level of sedation before driving or engaging in other tasks for which sedation could compromise their safety. Benzodiazepine use during pregnancy requires a careful discussion of risks and benefits (Box 2).38

Box 2

Using benzodiazepines during pregnancy

Benzodiazepine use during pregnancy has been associated with cleft palate and urogenital and neurologic malformations in the fetus.38 Although data are conflicting—particularly among recent meta-analyses that fail to demonstrate an association—some experts advise against benzodiazepine use in the first trimester. Participate in shared decision making with your patients and educate them about the potential risks and benefits of benzodiazepine use during the first trimester and throughout pregnancy. After delivery, newborns may develop “floppy baby syndrome”—which is associated with lethargy, difficulty eating, and respiratory depression—or withdrawal.38 To minimize this risk, consider tapering the benzodiazepine as the patient approaches delivery.

Related Resources

Drug Brand Names

  • Alprazolam • Xanax
  • Chlordiazepoxide • Librium, Limbitrol
  • Clonazepam • Klonopin
  • Clorazepate • Tranxene
  • Diazepam • Valium
  • Diphenhydramine • Benadryl, others
  • Estazolam • ProSom
  • Flumazenil • Romazicon
  • Flurazepam • Dalmane
  • Haloperidol • Haldol
  • Lithium • Lithobid
  • Lorazepam • Ativan
  • Olanzapine • Zyprexa
  • Oxazepam • Serax
  • Paroxetine • Paxil
  • Pregabalin • Lyrica
  • Propranolol • Inderal, InnoPran XL, others
  • Quazepam • Doral
  • Ramelteon • Rozerem
  • Sertraline • Zoloft
  • Temazepam • Restoril
  • Triazolam • Halcion
  • Valproic acid • Depakene, Stavzor, others

Disclosures

Drs. Bostwick and Yasugi report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Casher is a speaker for AstraZeneca and Sunovion Pharmaceuticals.

References

1. Mihic SJ, Harris RA. Hypnotics and sedatives. In: Brunton LL Chabner BA, Knollmann BC, eds. Goodman & Gilman’s the pharmacological basis of therapeutics. New York, NY: McGraw Hill and Company; 2011:457-480.

2. Facts and comparisons Web site. 2011 Wolters Kluwer Health Inc. http://online.factsandcomparisons.com. Accessed August 16, 2011.

3. DuPont RL, Greene W, Lydiard RB. Sedatives and hypnotics: pharmacology and epidemiology. In: Gold MS Hermann R, eds. UpToDate. http://www.uptodate.com/contents/sedatives-and-hypnotics-abuse-and-dependence-pharmacology-and-epidemiology. Accessed August 16, 2011.

4. U.S. Food and Drug Administration. Orange book: approved drug products with therapeutic equivalence evaluations. http://www.accessdata.fda.gov/scripts/cder/ob/default.cfm. Accessed August 16, 2011.

5. Chouinard G. Issues in the clinical use of benzodiazepines: potency withdrawal, and rebound. J Clin Psychiatry. 2004;65(suppl 5):7-12.

6. Shader RI, Greenblatt DJ. Can you provide a table of equivalencies for benzodiazepines and other marketed benzodiazepine receptor agonists? J Clin Psychopharmacol. 1997;17(4):331.-

7. Amato L, Minozzi S, Davoli M. Efficacy and safety of pharmacologic interventions for the treatment of the alcohol withdrawal syndrome. Cochrane Database Syst Rev. 2011;15(6):CD008537.-

8. Schutte-Rodin S, Broch L, Buysse D, et al. Clinical guideline for the evaluation and management of chronic insomnia in adults. J Clin Sleep Med. 2008;4(5):487-504.

9. Foral P, Dewan N, Malesker M. Insomnia: a therapeutic review for pharmacists. Consult Pharm. 2011;26(5):332-341.

10. Riemann D, Perlis ML. The treatments of chronic insomnia: a review of benzodiazepine receptor agonists and psychological and behavioral therapies. Sleep Med Rev. 2009;13(3):205-214.

11. Hidalgo RB, Tupler LA, Davidson JR. An effect-size analysis of pharmacologic treatments of generalized anxiety disorder. J Psychopharmacol. 2007;21(8):864-872.

12. Davidson JR, Zhang W, Connor KM, et al. A psychopharmacological treatment algorithm for generalised anxiety disorder (GAD). J Psychopharmacol. 2010;24(1):3-26.

13. Bandelow B, Zohar J, Hollander E, et al. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the pharmacological treatment of anxiety, obsessive-compulsive and post-traumatic stress disorders - first revision. World J Biol Psychiatry. 2008;9(4):248-312.

14. American Psychiatric Association. Practice guideline for the treatment of patients with panic disorder. 2nd ed. Arlington VA: American Psychiatric Publishing, Inc.; 2009.

15. Pollack MH, Simon NM, Worthington JJ, et al. Combined paroxetine and clonazepam treatment strategies compared to paroxetine monotherapy for panic disorder. J Psychopharmacol. 2003;17(3):276-282.

16. Watanabe N, Churchill R, Furukawa TA. Combined psychotherapy plus benzodiazepines for panic disorder. Cochrane Database Syst Rev. 2009;(1):CD005335.-

17. Otto MW, McHugh RK, Simon NM, et al. Efficacy of CBT for benzodiazepine discontinuation in patients with panic disorder: further evaluation. Behav Res Ther. 2010;48(8):720-727.

18. Davidson JR. Use of benzodiazepines in social anxiety disorder generalized anxiety disorder, and posttraumatic stress disorder. J Clin Psychiatry. 2004;65(suppl 5):29-33.

19. Argyropoulos SV, Sandford JJ, Nutt DJ. The psychobiology of anxiolytic drugs Part 2: pharmacological treatments of anxiety. Pharmacol Ther. 2000;88(3):213-227.

20. Connor KM, Davidson JR, Potts NL, et al. Discontinuation of clonazepam in the treatment of social phobia. J Clin Psychopharmacol. 1998;18(5):373-378.

21. Miller CH, Fleischhacker WW. Managing antipsychotic-induced acute and chronic akathisia. Drug Saf. 2000;22(1):73-81.

22. Rodnitzky RL. Drug-induced movement disorders. Clin Neuropharmacol. 2002;25(3):142-151.

23. Arbaizar B, Gómez-Acebo I, Llorca J. Postural induced tremor in psychiatry. Psychiatry Clin Neurosci. 2008;62(6):638-645.

24. Casher MI, Bess JD. Manual of inpatient psychiatry. New York NY: Cambridge University Press; 2010.

25. Battaglia J. Pharmacological management of acute agitation. Drugs. 2005;65(9):1207-1222.

26. Physicians’ desk reference. Montvale NJ: PDR Network, LLC; 2010.

27. Rosebush PI, Mazurek MF. Catatonia and its treatment. Schizophr Bull. 2010;36(2):239-242.

28. Ungvari GS, Kau LS, Wai-Kwong T, et al. The pharmacological treatment of catatonia: an overview. Eur Arch Psychiatry Clin Neurosci. 2001;251(suppl 1):I31-I34.

29. Fink M, Taylor MA. Catatonia: a clinician’s guide to diagnosis and treatment. New York NY: Cambridge University Press; 2003.

30. Naguib N, Koorn R. Interactions between psychotropics anaesthetics and electroconvulsive therapy: implications for drug choice and patient management. CNS Drugs. 2002;16(4):229-247.

31. Boylan LS, Haskett RF, Mulsant BH, et al. Determinants of seizure threshold in ECT: benzodiazepine use, anesthetic dosage, and other factors. J ECT. 2000;16(1):3-18.

32. Krystal AD, Watts BV, Weiner RD, et al. The use of flumazenil in the anxious and benzodiazepine-dependent ECT patient. J ECT. 1998;14(1):5-14.

33. Melton ST, Kirkwood CK. Anxiety disorders I: generalized anxiety panic, and social anxiety disorders. In: DiPiro JT, Talbert RL, Yee GC, et al, eds. Pharmacotherapy: a pathophysiologic approach. New York, NY: McGraw-Hill Companies; 2011:1209-1228.

34. Benzodiazepine toolkit. The Pharmacist’s Letter/Prescriber’s Letter. 2011;27(4):270406.-

35. Lader M. Benzodiazepines revisited – will we ever learn? Addiction. 2011;106(12):2086-2109.

36. Toblin RL, Paulozzi LJ, Logan JE, et al. Mental illness and psychotropic drug use among prescription drug overdose deaths: a medical examiner chart review. J Clin Psychiatry. 2010;71(4):491-496.

37. Ashton H. The diagnosis and management of benzodiazepine dependence. Curr Opin Psychiatry. 2005;18(3):249-255.

38. Menon SJ. Psychotropic medication during pregnancy and lactation. Arch Gynecol Obstet. 2008;277(1):1-13.

References

1. Mihic SJ, Harris RA. Hypnotics and sedatives. In: Brunton LL Chabner BA, Knollmann BC, eds. Goodman & Gilman’s the pharmacological basis of therapeutics. New York, NY: McGraw Hill and Company; 2011:457-480.

2. Facts and comparisons Web site. 2011 Wolters Kluwer Health Inc. http://online.factsandcomparisons.com. Accessed August 16, 2011.

3. DuPont RL, Greene W, Lydiard RB. Sedatives and hypnotics: pharmacology and epidemiology. In: Gold MS Hermann R, eds. UpToDate. http://www.uptodate.com/contents/sedatives-and-hypnotics-abuse-and-dependence-pharmacology-and-epidemiology. Accessed August 16, 2011.

4. U.S. Food and Drug Administration. Orange book: approved drug products with therapeutic equivalence evaluations. http://www.accessdata.fda.gov/scripts/cder/ob/default.cfm. Accessed August 16, 2011.

5. Chouinard G. Issues in the clinical use of benzodiazepines: potency withdrawal, and rebound. J Clin Psychiatry. 2004;65(suppl 5):7-12.

6. Shader RI, Greenblatt DJ. Can you provide a table of equivalencies for benzodiazepines and other marketed benzodiazepine receptor agonists? J Clin Psychopharmacol. 1997;17(4):331.-

7. Amato L, Minozzi S, Davoli M. Efficacy and safety of pharmacologic interventions for the treatment of the alcohol withdrawal syndrome. Cochrane Database Syst Rev. 2011;15(6):CD008537.-

8. Schutte-Rodin S, Broch L, Buysse D, et al. Clinical guideline for the evaluation and management of chronic insomnia in adults. J Clin Sleep Med. 2008;4(5):487-504.

9. Foral P, Dewan N, Malesker M. Insomnia: a therapeutic review for pharmacists. Consult Pharm. 2011;26(5):332-341.

10. Riemann D, Perlis ML. The treatments of chronic insomnia: a review of benzodiazepine receptor agonists and psychological and behavioral therapies. Sleep Med Rev. 2009;13(3):205-214.

11. Hidalgo RB, Tupler LA, Davidson JR. An effect-size analysis of pharmacologic treatments of generalized anxiety disorder. J Psychopharmacol. 2007;21(8):864-872.

12. Davidson JR, Zhang W, Connor KM, et al. A psychopharmacological treatment algorithm for generalised anxiety disorder (GAD). J Psychopharmacol. 2010;24(1):3-26.

13. Bandelow B, Zohar J, Hollander E, et al. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the pharmacological treatment of anxiety, obsessive-compulsive and post-traumatic stress disorders - first revision. World J Biol Psychiatry. 2008;9(4):248-312.

14. American Psychiatric Association. Practice guideline for the treatment of patients with panic disorder. 2nd ed. Arlington VA: American Psychiatric Publishing, Inc.; 2009.

15. Pollack MH, Simon NM, Worthington JJ, et al. Combined paroxetine and clonazepam treatment strategies compared to paroxetine monotherapy for panic disorder. J Psychopharmacol. 2003;17(3):276-282.

16. Watanabe N, Churchill R, Furukawa TA. Combined psychotherapy plus benzodiazepines for panic disorder. Cochrane Database Syst Rev. 2009;(1):CD005335.-

17. Otto MW, McHugh RK, Simon NM, et al. Efficacy of CBT for benzodiazepine discontinuation in patients with panic disorder: further evaluation. Behav Res Ther. 2010;48(8):720-727.

18. Davidson JR. Use of benzodiazepines in social anxiety disorder generalized anxiety disorder, and posttraumatic stress disorder. J Clin Psychiatry. 2004;65(suppl 5):29-33.

19. Argyropoulos SV, Sandford JJ, Nutt DJ. The psychobiology of anxiolytic drugs Part 2: pharmacological treatments of anxiety. Pharmacol Ther. 2000;88(3):213-227.

20. Connor KM, Davidson JR, Potts NL, et al. Discontinuation of clonazepam in the treatment of social phobia. J Clin Psychopharmacol. 1998;18(5):373-378.

21. Miller CH, Fleischhacker WW. Managing antipsychotic-induced acute and chronic akathisia. Drug Saf. 2000;22(1):73-81.

22. Rodnitzky RL. Drug-induced movement disorders. Clin Neuropharmacol. 2002;25(3):142-151.

23. Arbaizar B, Gómez-Acebo I, Llorca J. Postural induced tremor in psychiatry. Psychiatry Clin Neurosci. 2008;62(6):638-645.

24. Casher MI, Bess JD. Manual of inpatient psychiatry. New York NY: Cambridge University Press; 2010.

25. Battaglia J. Pharmacological management of acute agitation. Drugs. 2005;65(9):1207-1222.

26. Physicians’ desk reference. Montvale NJ: PDR Network, LLC; 2010.

27. Rosebush PI, Mazurek MF. Catatonia and its treatment. Schizophr Bull. 2010;36(2):239-242.

28. Ungvari GS, Kau LS, Wai-Kwong T, et al. The pharmacological treatment of catatonia: an overview. Eur Arch Psychiatry Clin Neurosci. 2001;251(suppl 1):I31-I34.

29. Fink M, Taylor MA. Catatonia: a clinician’s guide to diagnosis and treatment. New York NY: Cambridge University Press; 2003.

30. Naguib N, Koorn R. Interactions between psychotropics anaesthetics and electroconvulsive therapy: implications for drug choice and patient management. CNS Drugs. 2002;16(4):229-247.

31. Boylan LS, Haskett RF, Mulsant BH, et al. Determinants of seizure threshold in ECT: benzodiazepine use, anesthetic dosage, and other factors. J ECT. 2000;16(1):3-18.

32. Krystal AD, Watts BV, Weiner RD, et al. The use of flumazenil in the anxious and benzodiazepine-dependent ECT patient. J ECT. 1998;14(1):5-14.

33. Melton ST, Kirkwood CK. Anxiety disorders I: generalized anxiety panic, and social anxiety disorders. In: DiPiro JT, Talbert RL, Yee GC, et al, eds. Pharmacotherapy: a pathophysiologic approach. New York, NY: McGraw-Hill Companies; 2011:1209-1228.

34. Benzodiazepine toolkit. The Pharmacist’s Letter/Prescriber’s Letter. 2011;27(4):270406.-

35. Lader M. Benzodiazepines revisited – will we ever learn? Addiction. 2011;106(12):2086-2109.

36. Toblin RL, Paulozzi LJ, Logan JE, et al. Mental illness and psychotropic drug use among prescription drug overdose deaths: a medical examiner chart review. J Clin Psychiatry. 2010;71(4):491-496.

37. Ashton H. The diagnosis and management of benzodiazepine dependence. Curr Opin Psychiatry. 2005;18(3):249-255.

38. Menon SJ. Psychotropic medication during pregnancy and lactation. Arch Gynecol Obstet. 2008;277(1):1-13.

Issue
Current Psychiatry - 11(04)
Issue
Current Psychiatry - 11(04)
Page Number
54-64
Page Number
54-64
Publications
Publications
Topics
Article Type
Display Headline
Benzodiazepines: A versatile clinical tool
Display Headline
Benzodiazepines: A versatile clinical tool
Legacy Keywords
Benzodiazepines; alcohol withdrawal; insomnia; anxiety disorders; panic disorder; akathisia; catatonia; agitation
Legacy Keywords
Benzodiazepines; alcohol withdrawal; insomnia; anxiety disorders; panic disorder; akathisia; catatonia; agitation
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Nonsuicidal self-injury: How categorization guides treatment

Article Type
Changed
Tue, 12/11/2018 - 15:04
Display Headline
Nonsuicidal self-injury: How categorization guides treatment

Discuss this article at www.facebook.com/CurrentPsychiatry

Formerly called self-mutilation, self-injury, or self-harm, nonsuicidal self-injury (NSSI) is the deliberate and direct alteration or destruction of healthy body tissue without suicidal intent; these behaviors range from skin cutting or burning to eye enucleation or amputation of body parts. NSSI must be deliberate, as opposed to accidental or indirect behaviors—such as overdoses or ingesting harmful substances—that cause injury that is uncertain, ambiguous as to course, or invisible (the injuries do not disfigure observable body tissue).1 NSSI acts are done without an intent to die, although persons who self-harm may have suicidal ideation and passive thoughts of dying.2 Persons who repeatedly engage in NSSI and are demoralized over their inability to control it are at risk for suicide attempts.3

NSSI can be classified as nonpathological or pathological.4 Culturally sanctioned, nonpathological NSSI consists of body modification practices such as tattoos or piercing. Body modification practices may be a sublimation of pathological NSSI. For a description of nonpathological NSSI, see the Box.5 Pathological NSSI typically is a method of emotional regulation. Understanding why patients engage in pathological NSSI and how it is categorized can help guide assessment and treatment.

Box

Body modification: When self-injury is not pathological

Body modification practices and rituals are culturally sanctioned forms of nonsuicidal self-injury (NSSI). Body modification practices include tattooing and piercing earlobes, nipples, and other body parts to accommodate jewelry. Most practices are harmless but when carried to extremes, they may point to underlying neuroses. For some patients, a tattoo or piercing may be psychologically beneficial—eg, to reclaim one’s body after an attack or rape.5

Body modification rituals, such as head gashing by Sufi healers, penis cutting during aboriginal coming-of-age ceremonies, and Hindu body piercing to attain spiritual goals, are meaningful activities that reflect the tradition, symbolism, and beliefs of a society. These rituals serve an elemental purpose by correcting or preventing destabilizing conditions that threaten people and communities, such as mental and physical diseases; angry gods, spirits, or ancestors; failure of children to accept adult responsibilities; conflicts (eg, male-female, intergenerational, interclass, intertribal); loosening of clear social role distinctions; loss of group identity; immoral or sinful behaviors; and ecological disasters.

These rituals are effective because participants believe they promote healing, spirituality, and social order. Knowledge about body modification practices and rituals in which NSSI is perceived to be therapeutic opens the door to an understanding of pathological NSSI as a form of self-help behavior and allows clinicians to have a more empathic interaction with patients who self-injure.

Why people engage in NSSI

NSSI is best regarded as a pathological approach to emotional regulation and distress tolerance that provides rapid but temporary relief from disturbing thoughts, feelings, and emotions. For approximately 90% of patients, NSSI decreases symptoms, most commonly untenable anxiety (“It’s like popping a balloon”), depressed mood, racing thoughts, swirling emotions, anger, hallucinations, and flashbacks.6,7 In some instances, NSSI generates desired feelings and self-stimulation during periods of dissociation, depersonalization, grief, insecurity, loneliness, extreme boredom, self-pity, and alienation.8,9 NSSI also may signal distress to elicit a caring response from others or provide a means of escape from intolerable social situations.10Table 1 lists factors associated with NSSI.

Table 1

Factors associated with NSSI

High levels of negative and unpleasant thoughts and feelingsa
Poor communication skills and problem-solving abilitiesb
Abuse, maltreatment, hostility, and marked criticism during childhoodc,d
Under- or over-arousal responses to stressb
High valuation of NSSI to achieve a desired responsee
Need for self-punishmenta
Modeling behaviors based on exposure to NSSI among peers, on the Internet—ie, postings on YouTube—and in the mediaf
NSSI: nonsuicidal self-injury Source:
References
  1. Deliberto TL, Nock MK. An exploratory study of correlates, onset, and offset of non-suicidal self-injury. Arch Suicide Res. 2008;12(3):219-231.
  2. Nock MK, Mendes WB. Physiological arousal, distress tolerance, and social problem-solving deficits among adolescent self-injurers. J Consult Clin Psychol. 2008;76(1):28-38.
  3. Weierich MR, Nock MK. Posttraumatic stress symptoms mediate the relation between childhood sexual abuse and nonsuicidal self-injury. J Consult Clin Psychol. 2008;76(1):39-44.
  4. Yates TM. The developmental psychopathology of self-injurious behavior: compensatory regulation in posttraumatic adaptation. Clin Psychol Rev. 2004;24(1):35-74.
  5. Whitlock JL, Powers JL, Eckenrode J. The virtual cutting edge: the Internet and adolescent self-injury. Dev Psychol. 2006;42(3):407-417.
  6. Lewis SP, Heath NL, St Denis JM, et al. The scope of nonsuicidal self-injury on YouTube. Pediatrics. 2011;127(3):e552-e557.

The functional approach

One model of classifying NSSI focuses on the behavioral functions it serves.11,12 In this model, the most common function of NSSI is removal or escape from an aversive affective or cognitive state (automatic positive reinforcement). Automatic negative reinforcement explains using NSSI to generate feelings—eg, by patients with anhedonia or numbness. NSSI also may be used as a signal of distress to gain attention, access helpful environmental resources (social positive reinforcement), or remove distressing interpersonal demands (social negative reinforcement).

 

 

The functional model is key to providing thorough clinical evaluations that should include understanding the antecedent and consequent thoughts, feelings, situations, triggers, and vulnerabilities related to NSSI acts.

The medical approach

A descriptive, phenomenological model of NSSI classification uses concepts and terminology with which most psychiatrists are familiar, takes into account patients who have comorbid psychiatric disorders, is based on atheoretical, descriptive observations, and fits into what might be regarded as a “medical model.” In this classification, NSSI usually is regarded as a symptom or associated feature of a specific psychiatric disorder, although it may occur in persons who do not meet diagnostic criteria of a mental illness—eg, “copycat” cutting in high school students.13,14 NSSI may fall within 4 descriptive categories: major, stereotypic, compulsive, or impulsive. For psychiatric disorders associated with these types of pathological NSSI, see Table 2.

Major NSSI includes infrequent acts that destroy significant body tissue, such as eye enucleation and amputation of body parts. They are sudden, messy, and often bloody acts. Seventy-five percent occur during a psychotic state, mainly schizophrenia; of these, approximately one-half occur during a first psychotic episode.15 The reasons patients typically offer for such behavior often defy logical understanding—eg, to enhance general well-being—but most center on religion, such as a concrete interpretation of biblical texts about removing an offending eye or hand or becoming an eunuch,16,17 or on sexuality, such as controlling troubling hypersexuality or fear of giving in to homosexual urges.18

Stereotypic NSSI acts, most commonly associated with severe and profound mental retardation, include repetitive head banging; eye gouging; biting lips, the tongue, cheeks, or fingers; and face or head slapping. The behaviors may be monotonously repetitive, have a rhythmic pattern, and be performed without shame or guilt in the presence of onlookers.

Compulsive NSSI encompasses repetitive behaviors such as severe skin scratching and nail biting, hair pulling (trichotillomania), and skin digging (delusional parasitosis).

Impulsive NSSI consists of acts such as skin cutting, burning, and carving; sticking pins or other objects under the skin or into the chest or abdomen; interfering with wound healing; and smashing hand or foot bones. These behaviors usually are episodic and occur more frequently in females. The average age of onset in patients who engage in impulsive NSSI is 12 to 14, although it may occur throughout the life cycle.

One or 2 isolated instances of impulsive NSSI do not have much prognostic importance unless they are serious enough to warrant an emergency department visit. The real danger is when the behavior becomes repetitive and “addictive.” The crossover from episodic to repetitive usually varies from 5 to 10 episodes.

Persons who engage in repetitive NSSI may use multiple methods, but skin cutting predominates. Such persons often develop a self-identity as a “cutter,” are preoccupied with their NSSI, may carve words into their skin, and may perform acts of self-harm with other self-injurers. Some may cut themselves hundreds or even thousands of times, creating scars that result in social morbidity. They often seek professional help avidly, but may become so demoralized over their inability to stop their NSSI that they are at risk for suicide.3 In some repetitive self-injurers, other impulsive behaviors such as bulimia or substance abuse may alternate or coexist with NSSI. This pattern often runs its course in 5 to 15 years and may end abruptly, especially in patients with borderline personality disorder.

Table 2

Psychiatric disorders associated with pathological NSSI

Type of NSSIRelated psychiatric disorders
MajorAlcohol/drug intoxication, body integrity identity disordera
StereotypicAutism,b Tourette’s syndrome,c Lesch-Nyhan syndrome,d hereditary neuropathies,e mental retardation
CompulsiveTrichotillomania, delusional parasitosis
ImpulsiveAnxiety disorders (generalized, acute stress, posttraumatic stress, obsessive-compulsive, substance-inducedf-h); borderline, histrionic, and antisocial personality disordersi,j; somatoform and factitious disordersk,l; dissociative identity and depersonalization disordersm,n; anorexia and bulimia nervosao,p; depressive disordersq,r; bipolar disorders; schizophreniat,u; alcohol use disorderv; kleptomaniaw
NSSI: nonsuicidal self-injury Source:
References
  1. First MB. Desire for amputation of a limb: paraphilia, psychosis, or a new type of identity disorder. Psychol Med. 2005;35(6):919-928.
  2. Abrahams BS, Geschwind DH. Advances in autism genetics: on the threshold of a new neurobiology. Nat Rev Genet. 2008;9(5):341-355.
  3. Robertson MM, Trimble MR, Lees AJ. Self-injurious behaviour and the Gilles de la Tourette syndrome: a clinical study and review of the literature. Psychol Med. 1989;19(3):611-625.
  4. Baumeister AA, Frye GD. The biochemical basis of the behavioral disorder in the Lesch-Nyhan syndrome. Neurosci Biobehav Rev. 1985;9(2):169-178.
  5. Gadoth N, Mass E. Hereditary neuropathies with self-mutilation. J Pediatr Neurol. 2004;2(4):205-211.
  6. Nock MK, Prinstein MJ. A functional approach to the assessment of self-mutilative behavior. J Consult Clin Psychol. 2004;72(5):885-890.
  7. Pitman RK. Self-mutilation in combat-related PTSD. Am J Psychiatry. 1990;147(1):123-124.
  8. Primeau F, Fontaine R. Obsessive disorder with self-mutilation: a subgroup responsive to pharmacotherapy. Can J Psychiatry. 1987;32(8):699-701.
  9. Stone MH. Borderline personality disorder. Primary Psychiatry. 2006;13(5):36-39.
  10. Coid J, Wilkins J, Coid B, et al. Self-mutilation in female remanded prisoners II: a cluster analytic approach towards identification of a behavioral syndrome. Crim Behav Ment Health. 1992;2:1-14.
  11. Rogers T. Self-inflicted eye-injuries. Br J Psychiatry. 1987;151:691-693.
  12. Nielsen K, Jeppesen M, Simmelsgaard L, et al. Self-inflicted skin diseases. A retrospective analysis of 57 patients with dermatitis artefacta seen in a dermatology department. Acta Derm Venereol. 2005;85(6):512-515.
  13. Bliss EL. Multiple personalities. A report of 14 cases with implications for schizophrenia and hysteria. Arch Gen Psychiatry. 1980;37(12):1388-1397.
  14. Miller F, Bashkin EA. Depersonalization and self-mutilation. Psychoanal Q. 1974;43(4):638-649.
  15. Paul T, Schroeter K, Dahme B, et al. Self-injurious behavior in women with eating disorders. Am J Psychiatry. 2002;159(3):408-411.
  16. Favazza AR, DeRosear L, Conterio K. Self-mutilation and eating disorders. Suicide Life Threat Behav. 1989;19(4):352-361.
  17. Nixon MK, Cloutier PF, Aggarwal S. Affect regulation and addictive aspects of repetitive self-injury in hospitalized adolescents. J Am Acad Child Adolesc Psychiatry. 2002;41(11):1333-1341.
  18. Nock MK, Joiner TE Jr, Gordon KH, et al. Non-suicidal self-injury among adolescents: diagnostic correlates and relation to suicide attempts. Psychiatry Res. 2006;144(1):65-72.
  19. Esposito-Smythers C, Goldstein T, Birmaher B, et al. Clinical and psychosocial correlates of non-suicidal self-injury within a sample of children and adolescents with bipolar disorder. J Affect Disord. 2010;125(1-3):89-97.
  20. Nelson SH, Grunebaum H. A follow-up study of wrist slashers. Am J Psychiatry. 1971;127(10):1345-1349.
  21. Green AH. Self-mutilation in schizophrenic children. Arch Gen Psychiatry. 1967;17(2):234-244.
  22. Favazza AR, Conterio K. Female habitual self-mutilators. Acta Psychiatr Scand. 1989;79(3):283-289.
  23. Evans C, Lacey JH. Multiple self-damaging behaviour among alcoholic women. A prevalence study. Br J Psychiatry. 1992;161:643-647.
 

 

First-line treatment: Psychotherapy

Many studies have demonstrated the efficacy of psychotherapy as the primary treatment for NSSI.19-21 Except for patients with Lesch-Nyhan syndrome or other rare neurologic syndromes, the biologic causes of NSSI, including the role of endogenous opioids, are unclear. No medications are FDA-approved for NSSI. Pharmacotherapy may help NSSI patients, but such treatment recommendations are based on clinical experience, and polypharmacy is common.22 Studies have not demonstrated specific benefits or consistent efficacy of pharmacotherapy for NSSI.23

Major NSSI. Prevention is key to addressing major NSSI. Consider atypical antipsychotics for psychotic patients who are preoccupied with religion, the Bible, or sexuality, as well as those who dramatically and suddenly change their appearance by cutting off their hair, engaging in extreme body modification practices, or wearing bizarre clothes.24 In my clinical experience, agitated patients who have committed major NSSI are at high risk for a second episode and should receive pharmacotherapy based on treatment guidelines and hospitalized until the agitation is controlled.

Stereotypic NSSI. Patients with this form of NSSI often cannot articulate what is bothering them. With input from caretakers, assess the likelihood that a patient is reacting to pain. Analgesics may be effective. Also check for infections such as otitis media. Selecting a medication can be challenging. Start with a moderate dose of a selective serotonin reuptake inhibitor (SSRI), then slowly add an atypical antipsychotic, followed by a mood stabilizer, then clonidine, and then a beta blocker; a trial of naltrexone also is an option.23 Behavior therapy is the primary treatment.

Compulsive NSSI. Compulsive NSSI patients typically seek help from dermatologists or family physicians. Literature on psychiatric treatment is limited, but SSRIs, lithium, benzodiazepines, and atypical antipsychotics (for delusional parasitosis) may be effective. N-acetylcysteine, 600 mg twice a day, may relieve trichotillomania.25 Treatment should include psychotherapy.

Impulsive NSSI. Patients who engage in episodic impulsive NSSI should receive pharmacotherapy for underlying psychiatric illnesses such as generalized anxiety disorder, posttraumatic stress disorder, or depression. Do not automatically diagnose borderline personality disorder. Patients whose NSSI behavior is uncontrollable initially should receive high doses of SSRIs that can be lowered when impulsivity decreases, atypical antipsychotics, and a mood stabilizer such as lamotrigine. Psychotherapy is vital, especially dialectical behavior therapy. Cognitive-behavioral and interpersonal therapies also are effective, as is psychodynamic therapy.19-21

NSSI patients and their families may benefit from Web sites that provide information, advice, monitored blogs, and support groups (see Related Resources).

Related Resources

  • Favazza A. Bodies under siege: self-mutilation, nonsuicidal self-injury, and body modification in culture and psychiatry. 3rd ed. Baltimore, MD: Johns Hopkins University Press; 2011.
  • Nock MK. Understanding nonsuicidal self-injury: origins, assessment, and treatment. Washington, DC: American Psychological Association; 2009.
  • Cornell University Family Life Development Center. About self-injury. www.crpsib.com/whatissi.asp.

Drug Brand Names

  • Clonidine • Catapres, Kapvay
  • Lamotrigine • Lamictal
  • Lithium • Eskalith, Lithobid
  • Naltrexone • ReVia

Disclosure

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

References

1. Walsh BW, Rosen PM. Self-mutilation: theory research, and treatment. New York, NY: Guilford Press; 2008:32.

2. Nock MK, Favazza AR. Nonsuicidal self-injury: definition and classification. In: Nock MK ed. Understanding nonsuicidal self-injury: origins, assessment, and treatment. Washington, DC: American Psychological Association; 2009:9-18.

3. Favazza AR, Conterio K. Female habitual self-mutilators. Acta Psychiatr Scand. 1989;79(3):283-289.

4. Favazza A. Bodies under siege: self-mutilation nonsuicidal self-injury, and body modification in culture and psychiatry. 3rd ed. Baltimore, MD: Johns Hopkins University Press; 2011.

5. Gallina R. Tattoos and body piercing. In: Vale V Juno A, eds. Modern primitives. San Francisco, CA: Re/Search Publications; 1989:101-105.

6. Chapman AL, Gratz KL, Brown MZ. Solving the puzzle of deliberate self-harm: the experiential avoidance model. Behav Res Ther. 2006;44(3):371-394.

7. Nock MK, Prinstein MJ. Contextual features and behavioral functions of self-mutilation among adolescents. J Abnorm Psychol. 2005;114(1):140-146.

8. Miller F, Bashkin EA. Depersonalization and self-mutilation. Psychoanal Q. 1974;43(4):638-649.

9. Klonsky ED. The functions of deliberate self-injury: a review of the evidence. Clin Psychol Rev. 2007;27(2):226-239.

10. Nock MK. Actions speak louder than words: an elaborated theoretical model of the social functions of self-injury and other harmful behaviors. Appl Prev Psychol. 2008;12(4):159-168.

11. Nock MK, Prinstein MJ. A functional approach to the assessment of self-mutilative behavior. J Consult Clin Psychol. 2004;72(5):885-890.

12. Nock MK, Prinstein MJ. Contextual features and behavioral functions of self-mutilation among adolescents. J Abnorm Psychol. 2005;114(1):140-146.

13. Favazza AR, Rosenthal RJ. Diagnostic issues in self-mutilation. Hosp Community Psychiatry. 1993;44(2):134-140.

14. Rosen PM, Walsh BW. Patterns of contagion in self-mutilation epidemics. Am J Psychiatry. 1989;146(5):656-658.

15. Large M, Babidge N, Andrews D, et al. Major self-mutilation in the first episode of psychosis. Schizophr Bull. 2009;35(5):1012-1021.

16. Kushner AW. Two cases of auto-castration due to religious delusions. Br J Med Psychol. 1967;40(3):293-298.

17. Moskovitz RA, Byrd T. Rescuing the angel within: PCP-related self-enucleation. Psychosomatics. 1983;24(4):402-403,406.

18. Cleveland SE. Three cases of self-castration. J Nerv Ment Dis. 1956;123(4):386-391.

19. Linehan MM, Comtois KA, Murray AM, et al. Two-year randomized controlled trial and follow-up of dialectical behavior therapy vs therapy by experts for suicidal behaviors and borderline personality disorder. Arch Gen Psychiatry. 2006;63(7):757-766.

20. Kahng S, Iwata BA, Lewin AB. Behavioral treatment of self-injury 1964 to 2000. Am J Ment Retard. 2002;107(3):212-221.

21. Levy KN, Yeomans FE, Diamond D. Psychodynamic treatments of self-injury. J Clin Psychol. 2007;63(11):1105-1120.

22. Lott IT, McGregor M, Engelman L, et al. Longitudinal prescribing patterns for psychoactive medications in community-based individuals with developmental disabilities: utilization of pharmacy records. J Intellect Disabil Res. 2004;48(Pt 6):563-571.

23. Sandman CA. Psychopharmacologic treatment of nonsuicidal self-injury. In: Nock MK ed. Understanding nonsuicidal self-injury: origins, assessment, and treatment. Washington, DC: American Psychological Association; 2009:291-322.

24. Sweeny S, Zamecnik K. Predictors of self-mutilation in patients with schizophrenia. Am J Psychiatry. 1981;138(8):1086-1089.

25. Grant JE, Odlaug BL, Kim SW. N-acetylcysteine a glutamate modulator, in the treatment of trichotillomania: a double-blind, placebo-controlled study. Arch Gen Psychiatry. 2009;66(7):756-763.

Article PDF
Author and Disclosure Information

Armando R. Favazza, MD
Emeritus Professor of Psychiatry, University of Missouri-Columbia, Columbia, MO

Issue
Current Psychiatry - 11(03)
Publications
Page Number
21-26
Legacy Keywords
nonsuicidal self-injury; NSSI; self-harm; pathological NSSI; types of self-harm
Sections
Author and Disclosure Information

Armando R. Favazza, MD
Emeritus Professor of Psychiatry, University of Missouri-Columbia, Columbia, MO

Author and Disclosure Information

Armando R. Favazza, MD
Emeritus Professor of Psychiatry, University of Missouri-Columbia, Columbia, MO

Article PDF
Article PDF

Discuss this article at www.facebook.com/CurrentPsychiatry

Formerly called self-mutilation, self-injury, or self-harm, nonsuicidal self-injury (NSSI) is the deliberate and direct alteration or destruction of healthy body tissue without suicidal intent; these behaviors range from skin cutting or burning to eye enucleation or amputation of body parts. NSSI must be deliberate, as opposed to accidental or indirect behaviors—such as overdoses or ingesting harmful substances—that cause injury that is uncertain, ambiguous as to course, or invisible (the injuries do not disfigure observable body tissue).1 NSSI acts are done without an intent to die, although persons who self-harm may have suicidal ideation and passive thoughts of dying.2 Persons who repeatedly engage in NSSI and are demoralized over their inability to control it are at risk for suicide attempts.3

NSSI can be classified as nonpathological or pathological.4 Culturally sanctioned, nonpathological NSSI consists of body modification practices such as tattoos or piercing. Body modification practices may be a sublimation of pathological NSSI. For a description of nonpathological NSSI, see the Box.5 Pathological NSSI typically is a method of emotional regulation. Understanding why patients engage in pathological NSSI and how it is categorized can help guide assessment and treatment.

Box

Body modification: When self-injury is not pathological

Body modification practices and rituals are culturally sanctioned forms of nonsuicidal self-injury (NSSI). Body modification practices include tattooing and piercing earlobes, nipples, and other body parts to accommodate jewelry. Most practices are harmless but when carried to extremes, they may point to underlying neuroses. For some patients, a tattoo or piercing may be psychologically beneficial—eg, to reclaim one’s body after an attack or rape.5

Body modification rituals, such as head gashing by Sufi healers, penis cutting during aboriginal coming-of-age ceremonies, and Hindu body piercing to attain spiritual goals, are meaningful activities that reflect the tradition, symbolism, and beliefs of a society. These rituals serve an elemental purpose by correcting or preventing destabilizing conditions that threaten people and communities, such as mental and physical diseases; angry gods, spirits, or ancestors; failure of children to accept adult responsibilities; conflicts (eg, male-female, intergenerational, interclass, intertribal); loosening of clear social role distinctions; loss of group identity; immoral or sinful behaviors; and ecological disasters.

These rituals are effective because participants believe they promote healing, spirituality, and social order. Knowledge about body modification practices and rituals in which NSSI is perceived to be therapeutic opens the door to an understanding of pathological NSSI as a form of self-help behavior and allows clinicians to have a more empathic interaction with patients who self-injure.

Why people engage in NSSI

NSSI is best regarded as a pathological approach to emotional regulation and distress tolerance that provides rapid but temporary relief from disturbing thoughts, feelings, and emotions. For approximately 90% of patients, NSSI decreases symptoms, most commonly untenable anxiety (“It’s like popping a balloon”), depressed mood, racing thoughts, swirling emotions, anger, hallucinations, and flashbacks.6,7 In some instances, NSSI generates desired feelings and self-stimulation during periods of dissociation, depersonalization, grief, insecurity, loneliness, extreme boredom, self-pity, and alienation.8,9 NSSI also may signal distress to elicit a caring response from others or provide a means of escape from intolerable social situations.10Table 1 lists factors associated with NSSI.

Table 1

Factors associated with NSSI

High levels of negative and unpleasant thoughts and feelingsa
Poor communication skills and problem-solving abilitiesb
Abuse, maltreatment, hostility, and marked criticism during childhoodc,d
Under- or over-arousal responses to stressb
High valuation of NSSI to achieve a desired responsee
Need for self-punishmenta
Modeling behaviors based on exposure to NSSI among peers, on the Internet—ie, postings on YouTube—and in the mediaf
NSSI: nonsuicidal self-injury Source:
References
  1. Deliberto TL, Nock MK. An exploratory study of correlates, onset, and offset of non-suicidal self-injury. Arch Suicide Res. 2008;12(3):219-231.
  2. Nock MK, Mendes WB. Physiological arousal, distress tolerance, and social problem-solving deficits among adolescent self-injurers. J Consult Clin Psychol. 2008;76(1):28-38.
  3. Weierich MR, Nock MK. Posttraumatic stress symptoms mediate the relation between childhood sexual abuse and nonsuicidal self-injury. J Consult Clin Psychol. 2008;76(1):39-44.
  4. Yates TM. The developmental psychopathology of self-injurious behavior: compensatory regulation in posttraumatic adaptation. Clin Psychol Rev. 2004;24(1):35-74.
  5. Whitlock JL, Powers JL, Eckenrode J. The virtual cutting edge: the Internet and adolescent self-injury. Dev Psychol. 2006;42(3):407-417.
  6. Lewis SP, Heath NL, St Denis JM, et al. The scope of nonsuicidal self-injury on YouTube. Pediatrics. 2011;127(3):e552-e557.

The functional approach

One model of classifying NSSI focuses on the behavioral functions it serves.11,12 In this model, the most common function of NSSI is removal or escape from an aversive affective or cognitive state (automatic positive reinforcement). Automatic negative reinforcement explains using NSSI to generate feelings—eg, by patients with anhedonia or numbness. NSSI also may be used as a signal of distress to gain attention, access helpful environmental resources (social positive reinforcement), or remove distressing interpersonal demands (social negative reinforcement).

 

 

The functional model is key to providing thorough clinical evaluations that should include understanding the antecedent and consequent thoughts, feelings, situations, triggers, and vulnerabilities related to NSSI acts.

The medical approach

A descriptive, phenomenological model of NSSI classification uses concepts and terminology with which most psychiatrists are familiar, takes into account patients who have comorbid psychiatric disorders, is based on atheoretical, descriptive observations, and fits into what might be regarded as a “medical model.” In this classification, NSSI usually is regarded as a symptom or associated feature of a specific psychiatric disorder, although it may occur in persons who do not meet diagnostic criteria of a mental illness—eg, “copycat” cutting in high school students.13,14 NSSI may fall within 4 descriptive categories: major, stereotypic, compulsive, or impulsive. For psychiatric disorders associated with these types of pathological NSSI, see Table 2.

Major NSSI includes infrequent acts that destroy significant body tissue, such as eye enucleation and amputation of body parts. They are sudden, messy, and often bloody acts. Seventy-five percent occur during a psychotic state, mainly schizophrenia; of these, approximately one-half occur during a first psychotic episode.15 The reasons patients typically offer for such behavior often defy logical understanding—eg, to enhance general well-being—but most center on religion, such as a concrete interpretation of biblical texts about removing an offending eye or hand or becoming an eunuch,16,17 or on sexuality, such as controlling troubling hypersexuality or fear of giving in to homosexual urges.18

Stereotypic NSSI acts, most commonly associated with severe and profound mental retardation, include repetitive head banging; eye gouging; biting lips, the tongue, cheeks, or fingers; and face or head slapping. The behaviors may be monotonously repetitive, have a rhythmic pattern, and be performed without shame or guilt in the presence of onlookers.

Compulsive NSSI encompasses repetitive behaviors such as severe skin scratching and nail biting, hair pulling (trichotillomania), and skin digging (delusional parasitosis).

Impulsive NSSI consists of acts such as skin cutting, burning, and carving; sticking pins or other objects under the skin or into the chest or abdomen; interfering with wound healing; and smashing hand or foot bones. These behaviors usually are episodic and occur more frequently in females. The average age of onset in patients who engage in impulsive NSSI is 12 to 14, although it may occur throughout the life cycle.

One or 2 isolated instances of impulsive NSSI do not have much prognostic importance unless they are serious enough to warrant an emergency department visit. The real danger is when the behavior becomes repetitive and “addictive.” The crossover from episodic to repetitive usually varies from 5 to 10 episodes.

Persons who engage in repetitive NSSI may use multiple methods, but skin cutting predominates. Such persons often develop a self-identity as a “cutter,” are preoccupied with their NSSI, may carve words into their skin, and may perform acts of self-harm with other self-injurers. Some may cut themselves hundreds or even thousands of times, creating scars that result in social morbidity. They often seek professional help avidly, but may become so demoralized over their inability to stop their NSSI that they are at risk for suicide.3 In some repetitive self-injurers, other impulsive behaviors such as bulimia or substance abuse may alternate or coexist with NSSI. This pattern often runs its course in 5 to 15 years and may end abruptly, especially in patients with borderline personality disorder.

Table 2

Psychiatric disorders associated with pathological NSSI

Type of NSSIRelated psychiatric disorders
MajorAlcohol/drug intoxication, body integrity identity disordera
StereotypicAutism,b Tourette’s syndrome,c Lesch-Nyhan syndrome,d hereditary neuropathies,e mental retardation
CompulsiveTrichotillomania, delusional parasitosis
ImpulsiveAnxiety disorders (generalized, acute stress, posttraumatic stress, obsessive-compulsive, substance-inducedf-h); borderline, histrionic, and antisocial personality disordersi,j; somatoform and factitious disordersk,l; dissociative identity and depersonalization disordersm,n; anorexia and bulimia nervosao,p; depressive disordersq,r; bipolar disorders; schizophreniat,u; alcohol use disorderv; kleptomaniaw
NSSI: nonsuicidal self-injury Source:
References
  1. First MB. Desire for amputation of a limb: paraphilia, psychosis, or a new type of identity disorder. Psychol Med. 2005;35(6):919-928.
  2. Abrahams BS, Geschwind DH. Advances in autism genetics: on the threshold of a new neurobiology. Nat Rev Genet. 2008;9(5):341-355.
  3. Robertson MM, Trimble MR, Lees AJ. Self-injurious behaviour and the Gilles de la Tourette syndrome: a clinical study and review of the literature. Psychol Med. 1989;19(3):611-625.
  4. Baumeister AA, Frye GD. The biochemical basis of the behavioral disorder in the Lesch-Nyhan syndrome. Neurosci Biobehav Rev. 1985;9(2):169-178.
  5. Gadoth N, Mass E. Hereditary neuropathies with self-mutilation. J Pediatr Neurol. 2004;2(4):205-211.
  6. Nock MK, Prinstein MJ. A functional approach to the assessment of self-mutilative behavior. J Consult Clin Psychol. 2004;72(5):885-890.
  7. Pitman RK. Self-mutilation in combat-related PTSD. Am J Psychiatry. 1990;147(1):123-124.
  8. Primeau F, Fontaine R. Obsessive disorder with self-mutilation: a subgroup responsive to pharmacotherapy. Can J Psychiatry. 1987;32(8):699-701.
  9. Stone MH. Borderline personality disorder. Primary Psychiatry. 2006;13(5):36-39.
  10. Coid J, Wilkins J, Coid B, et al. Self-mutilation in female remanded prisoners II: a cluster analytic approach towards identification of a behavioral syndrome. Crim Behav Ment Health. 1992;2:1-14.
  11. Rogers T. Self-inflicted eye-injuries. Br J Psychiatry. 1987;151:691-693.
  12. Nielsen K, Jeppesen M, Simmelsgaard L, et al. Self-inflicted skin diseases. A retrospective analysis of 57 patients with dermatitis artefacta seen in a dermatology department. Acta Derm Venereol. 2005;85(6):512-515.
  13. Bliss EL. Multiple personalities. A report of 14 cases with implications for schizophrenia and hysteria. Arch Gen Psychiatry. 1980;37(12):1388-1397.
  14. Miller F, Bashkin EA. Depersonalization and self-mutilation. Psychoanal Q. 1974;43(4):638-649.
  15. Paul T, Schroeter K, Dahme B, et al. Self-injurious behavior in women with eating disorders. Am J Psychiatry. 2002;159(3):408-411.
  16. Favazza AR, DeRosear L, Conterio K. Self-mutilation and eating disorders. Suicide Life Threat Behav. 1989;19(4):352-361.
  17. Nixon MK, Cloutier PF, Aggarwal S. Affect regulation and addictive aspects of repetitive self-injury in hospitalized adolescents. J Am Acad Child Adolesc Psychiatry. 2002;41(11):1333-1341.
  18. Nock MK, Joiner TE Jr, Gordon KH, et al. Non-suicidal self-injury among adolescents: diagnostic correlates and relation to suicide attempts. Psychiatry Res. 2006;144(1):65-72.
  19. Esposito-Smythers C, Goldstein T, Birmaher B, et al. Clinical and psychosocial correlates of non-suicidal self-injury within a sample of children and adolescents with bipolar disorder. J Affect Disord. 2010;125(1-3):89-97.
  20. Nelson SH, Grunebaum H. A follow-up study of wrist slashers. Am J Psychiatry. 1971;127(10):1345-1349.
  21. Green AH. Self-mutilation in schizophrenic children. Arch Gen Psychiatry. 1967;17(2):234-244.
  22. Favazza AR, Conterio K. Female habitual self-mutilators. Acta Psychiatr Scand. 1989;79(3):283-289.
  23. Evans C, Lacey JH. Multiple self-damaging behaviour among alcoholic women. A prevalence study. Br J Psychiatry. 1992;161:643-647.
 

 

First-line treatment: Psychotherapy

Many studies have demonstrated the efficacy of psychotherapy as the primary treatment for NSSI.19-21 Except for patients with Lesch-Nyhan syndrome or other rare neurologic syndromes, the biologic causes of NSSI, including the role of endogenous opioids, are unclear. No medications are FDA-approved for NSSI. Pharmacotherapy may help NSSI patients, but such treatment recommendations are based on clinical experience, and polypharmacy is common.22 Studies have not demonstrated specific benefits or consistent efficacy of pharmacotherapy for NSSI.23

Major NSSI. Prevention is key to addressing major NSSI. Consider atypical antipsychotics for psychotic patients who are preoccupied with religion, the Bible, or sexuality, as well as those who dramatically and suddenly change their appearance by cutting off their hair, engaging in extreme body modification practices, or wearing bizarre clothes.24 In my clinical experience, agitated patients who have committed major NSSI are at high risk for a second episode and should receive pharmacotherapy based on treatment guidelines and hospitalized until the agitation is controlled.

Stereotypic NSSI. Patients with this form of NSSI often cannot articulate what is bothering them. With input from caretakers, assess the likelihood that a patient is reacting to pain. Analgesics may be effective. Also check for infections such as otitis media. Selecting a medication can be challenging. Start with a moderate dose of a selective serotonin reuptake inhibitor (SSRI), then slowly add an atypical antipsychotic, followed by a mood stabilizer, then clonidine, and then a beta blocker; a trial of naltrexone also is an option.23 Behavior therapy is the primary treatment.

Compulsive NSSI. Compulsive NSSI patients typically seek help from dermatologists or family physicians. Literature on psychiatric treatment is limited, but SSRIs, lithium, benzodiazepines, and atypical antipsychotics (for delusional parasitosis) may be effective. N-acetylcysteine, 600 mg twice a day, may relieve trichotillomania.25 Treatment should include psychotherapy.

Impulsive NSSI. Patients who engage in episodic impulsive NSSI should receive pharmacotherapy for underlying psychiatric illnesses such as generalized anxiety disorder, posttraumatic stress disorder, or depression. Do not automatically diagnose borderline personality disorder. Patients whose NSSI behavior is uncontrollable initially should receive high doses of SSRIs that can be lowered when impulsivity decreases, atypical antipsychotics, and a mood stabilizer such as lamotrigine. Psychotherapy is vital, especially dialectical behavior therapy. Cognitive-behavioral and interpersonal therapies also are effective, as is psychodynamic therapy.19-21

NSSI patients and their families may benefit from Web sites that provide information, advice, monitored blogs, and support groups (see Related Resources).

Related Resources

  • Favazza A. Bodies under siege: self-mutilation, nonsuicidal self-injury, and body modification in culture and psychiatry. 3rd ed. Baltimore, MD: Johns Hopkins University Press; 2011.
  • Nock MK. Understanding nonsuicidal self-injury: origins, assessment, and treatment. Washington, DC: American Psychological Association; 2009.
  • Cornell University Family Life Development Center. About self-injury. www.crpsib.com/whatissi.asp.

Drug Brand Names

  • Clonidine • Catapres, Kapvay
  • Lamotrigine • Lamictal
  • Lithium • Eskalith, Lithobid
  • Naltrexone • ReVia

Disclosure

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

Discuss this article at www.facebook.com/CurrentPsychiatry

Formerly called self-mutilation, self-injury, or self-harm, nonsuicidal self-injury (NSSI) is the deliberate and direct alteration or destruction of healthy body tissue without suicidal intent; these behaviors range from skin cutting or burning to eye enucleation or amputation of body parts. NSSI must be deliberate, as opposed to accidental or indirect behaviors—such as overdoses or ingesting harmful substances—that cause injury that is uncertain, ambiguous as to course, or invisible (the injuries do not disfigure observable body tissue).1 NSSI acts are done without an intent to die, although persons who self-harm may have suicidal ideation and passive thoughts of dying.2 Persons who repeatedly engage in NSSI and are demoralized over their inability to control it are at risk for suicide attempts.3

NSSI can be classified as nonpathological or pathological.4 Culturally sanctioned, nonpathological NSSI consists of body modification practices such as tattoos or piercing. Body modification practices may be a sublimation of pathological NSSI. For a description of nonpathological NSSI, see the Box.5 Pathological NSSI typically is a method of emotional regulation. Understanding why patients engage in pathological NSSI and how it is categorized can help guide assessment and treatment.

Box

Body modification: When self-injury is not pathological

Body modification practices and rituals are culturally sanctioned forms of nonsuicidal self-injury (NSSI). Body modification practices include tattooing and piercing earlobes, nipples, and other body parts to accommodate jewelry. Most practices are harmless but when carried to extremes, they may point to underlying neuroses. For some patients, a tattoo or piercing may be psychologically beneficial—eg, to reclaim one’s body after an attack or rape.5

Body modification rituals, such as head gashing by Sufi healers, penis cutting during aboriginal coming-of-age ceremonies, and Hindu body piercing to attain spiritual goals, are meaningful activities that reflect the tradition, symbolism, and beliefs of a society. These rituals serve an elemental purpose by correcting or preventing destabilizing conditions that threaten people and communities, such as mental and physical diseases; angry gods, spirits, or ancestors; failure of children to accept adult responsibilities; conflicts (eg, male-female, intergenerational, interclass, intertribal); loosening of clear social role distinctions; loss of group identity; immoral or sinful behaviors; and ecological disasters.

These rituals are effective because participants believe they promote healing, spirituality, and social order. Knowledge about body modification practices and rituals in which NSSI is perceived to be therapeutic opens the door to an understanding of pathological NSSI as a form of self-help behavior and allows clinicians to have a more empathic interaction with patients who self-injure.

Why people engage in NSSI

NSSI is best regarded as a pathological approach to emotional regulation and distress tolerance that provides rapid but temporary relief from disturbing thoughts, feelings, and emotions. For approximately 90% of patients, NSSI decreases symptoms, most commonly untenable anxiety (“It’s like popping a balloon”), depressed mood, racing thoughts, swirling emotions, anger, hallucinations, and flashbacks.6,7 In some instances, NSSI generates desired feelings and self-stimulation during periods of dissociation, depersonalization, grief, insecurity, loneliness, extreme boredom, self-pity, and alienation.8,9 NSSI also may signal distress to elicit a caring response from others or provide a means of escape from intolerable social situations.10Table 1 lists factors associated with NSSI.

Table 1

Factors associated with NSSI

High levels of negative and unpleasant thoughts and feelingsa
Poor communication skills and problem-solving abilitiesb
Abuse, maltreatment, hostility, and marked criticism during childhoodc,d
Under- or over-arousal responses to stressb
High valuation of NSSI to achieve a desired responsee
Need for self-punishmenta
Modeling behaviors based on exposure to NSSI among peers, on the Internet—ie, postings on YouTube—and in the mediaf
NSSI: nonsuicidal self-injury Source:
References
  1. Deliberto TL, Nock MK. An exploratory study of correlates, onset, and offset of non-suicidal self-injury. Arch Suicide Res. 2008;12(3):219-231.
  2. Nock MK, Mendes WB. Physiological arousal, distress tolerance, and social problem-solving deficits among adolescent self-injurers. J Consult Clin Psychol. 2008;76(1):28-38.
  3. Weierich MR, Nock MK. Posttraumatic stress symptoms mediate the relation between childhood sexual abuse and nonsuicidal self-injury. J Consult Clin Psychol. 2008;76(1):39-44.
  4. Yates TM. The developmental psychopathology of self-injurious behavior: compensatory regulation in posttraumatic adaptation. Clin Psychol Rev. 2004;24(1):35-74.
  5. Whitlock JL, Powers JL, Eckenrode J. The virtual cutting edge: the Internet and adolescent self-injury. Dev Psychol. 2006;42(3):407-417.
  6. Lewis SP, Heath NL, St Denis JM, et al. The scope of nonsuicidal self-injury on YouTube. Pediatrics. 2011;127(3):e552-e557.

The functional approach

One model of classifying NSSI focuses on the behavioral functions it serves.11,12 In this model, the most common function of NSSI is removal or escape from an aversive affective or cognitive state (automatic positive reinforcement). Automatic negative reinforcement explains using NSSI to generate feelings—eg, by patients with anhedonia or numbness. NSSI also may be used as a signal of distress to gain attention, access helpful environmental resources (social positive reinforcement), or remove distressing interpersonal demands (social negative reinforcement).

 

 

The functional model is key to providing thorough clinical evaluations that should include understanding the antecedent and consequent thoughts, feelings, situations, triggers, and vulnerabilities related to NSSI acts.

The medical approach

A descriptive, phenomenological model of NSSI classification uses concepts and terminology with which most psychiatrists are familiar, takes into account patients who have comorbid psychiatric disorders, is based on atheoretical, descriptive observations, and fits into what might be regarded as a “medical model.” In this classification, NSSI usually is regarded as a symptom or associated feature of a specific psychiatric disorder, although it may occur in persons who do not meet diagnostic criteria of a mental illness—eg, “copycat” cutting in high school students.13,14 NSSI may fall within 4 descriptive categories: major, stereotypic, compulsive, or impulsive. For psychiatric disorders associated with these types of pathological NSSI, see Table 2.

Major NSSI includes infrequent acts that destroy significant body tissue, such as eye enucleation and amputation of body parts. They are sudden, messy, and often bloody acts. Seventy-five percent occur during a psychotic state, mainly schizophrenia; of these, approximately one-half occur during a first psychotic episode.15 The reasons patients typically offer for such behavior often defy logical understanding—eg, to enhance general well-being—but most center on religion, such as a concrete interpretation of biblical texts about removing an offending eye or hand or becoming an eunuch,16,17 or on sexuality, such as controlling troubling hypersexuality or fear of giving in to homosexual urges.18

Stereotypic NSSI acts, most commonly associated with severe and profound mental retardation, include repetitive head banging; eye gouging; biting lips, the tongue, cheeks, or fingers; and face or head slapping. The behaviors may be monotonously repetitive, have a rhythmic pattern, and be performed without shame or guilt in the presence of onlookers.

Compulsive NSSI encompasses repetitive behaviors such as severe skin scratching and nail biting, hair pulling (trichotillomania), and skin digging (delusional parasitosis).

Impulsive NSSI consists of acts such as skin cutting, burning, and carving; sticking pins or other objects under the skin or into the chest or abdomen; interfering with wound healing; and smashing hand or foot bones. These behaviors usually are episodic and occur more frequently in females. The average age of onset in patients who engage in impulsive NSSI is 12 to 14, although it may occur throughout the life cycle.

One or 2 isolated instances of impulsive NSSI do not have much prognostic importance unless they are serious enough to warrant an emergency department visit. The real danger is when the behavior becomes repetitive and “addictive.” The crossover from episodic to repetitive usually varies from 5 to 10 episodes.

Persons who engage in repetitive NSSI may use multiple methods, but skin cutting predominates. Such persons often develop a self-identity as a “cutter,” are preoccupied with their NSSI, may carve words into their skin, and may perform acts of self-harm with other self-injurers. Some may cut themselves hundreds or even thousands of times, creating scars that result in social morbidity. They often seek professional help avidly, but may become so demoralized over their inability to stop their NSSI that they are at risk for suicide.3 In some repetitive self-injurers, other impulsive behaviors such as bulimia or substance abuse may alternate or coexist with NSSI. This pattern often runs its course in 5 to 15 years and may end abruptly, especially in patients with borderline personality disorder.

Table 2

Psychiatric disorders associated with pathological NSSI

Type of NSSIRelated psychiatric disorders
MajorAlcohol/drug intoxication, body integrity identity disordera
StereotypicAutism,b Tourette’s syndrome,c Lesch-Nyhan syndrome,d hereditary neuropathies,e mental retardation
CompulsiveTrichotillomania, delusional parasitosis
ImpulsiveAnxiety disorders (generalized, acute stress, posttraumatic stress, obsessive-compulsive, substance-inducedf-h); borderline, histrionic, and antisocial personality disordersi,j; somatoform and factitious disordersk,l; dissociative identity and depersonalization disordersm,n; anorexia and bulimia nervosao,p; depressive disordersq,r; bipolar disorders; schizophreniat,u; alcohol use disorderv; kleptomaniaw
NSSI: nonsuicidal self-injury Source:
References
  1. First MB. Desire for amputation of a limb: paraphilia, psychosis, or a new type of identity disorder. Psychol Med. 2005;35(6):919-928.
  2. Abrahams BS, Geschwind DH. Advances in autism genetics: on the threshold of a new neurobiology. Nat Rev Genet. 2008;9(5):341-355.
  3. Robertson MM, Trimble MR, Lees AJ. Self-injurious behaviour and the Gilles de la Tourette syndrome: a clinical study and review of the literature. Psychol Med. 1989;19(3):611-625.
  4. Baumeister AA, Frye GD. The biochemical basis of the behavioral disorder in the Lesch-Nyhan syndrome. Neurosci Biobehav Rev. 1985;9(2):169-178.
  5. Gadoth N, Mass E. Hereditary neuropathies with self-mutilation. J Pediatr Neurol. 2004;2(4):205-211.
  6. Nock MK, Prinstein MJ. A functional approach to the assessment of self-mutilative behavior. J Consult Clin Psychol. 2004;72(5):885-890.
  7. Pitman RK. Self-mutilation in combat-related PTSD. Am J Psychiatry. 1990;147(1):123-124.
  8. Primeau F, Fontaine R. Obsessive disorder with self-mutilation: a subgroup responsive to pharmacotherapy. Can J Psychiatry. 1987;32(8):699-701.
  9. Stone MH. Borderline personality disorder. Primary Psychiatry. 2006;13(5):36-39.
  10. Coid J, Wilkins J, Coid B, et al. Self-mutilation in female remanded prisoners II: a cluster analytic approach towards identification of a behavioral syndrome. Crim Behav Ment Health. 1992;2:1-14.
  11. Rogers T. Self-inflicted eye-injuries. Br J Psychiatry. 1987;151:691-693.
  12. Nielsen K, Jeppesen M, Simmelsgaard L, et al. Self-inflicted skin diseases. A retrospective analysis of 57 patients with dermatitis artefacta seen in a dermatology department. Acta Derm Venereol. 2005;85(6):512-515.
  13. Bliss EL. Multiple personalities. A report of 14 cases with implications for schizophrenia and hysteria. Arch Gen Psychiatry. 1980;37(12):1388-1397.
  14. Miller F, Bashkin EA. Depersonalization and self-mutilation. Psychoanal Q. 1974;43(4):638-649.
  15. Paul T, Schroeter K, Dahme B, et al. Self-injurious behavior in women with eating disorders. Am J Psychiatry. 2002;159(3):408-411.
  16. Favazza AR, DeRosear L, Conterio K. Self-mutilation and eating disorders. Suicide Life Threat Behav. 1989;19(4):352-361.
  17. Nixon MK, Cloutier PF, Aggarwal S. Affect regulation and addictive aspects of repetitive self-injury in hospitalized adolescents. J Am Acad Child Adolesc Psychiatry. 2002;41(11):1333-1341.
  18. Nock MK, Joiner TE Jr, Gordon KH, et al. Non-suicidal self-injury among adolescents: diagnostic correlates and relation to suicide attempts. Psychiatry Res. 2006;144(1):65-72.
  19. Esposito-Smythers C, Goldstein T, Birmaher B, et al. Clinical and psychosocial correlates of non-suicidal self-injury within a sample of children and adolescents with bipolar disorder. J Affect Disord. 2010;125(1-3):89-97.
  20. Nelson SH, Grunebaum H. A follow-up study of wrist slashers. Am J Psychiatry. 1971;127(10):1345-1349.
  21. Green AH. Self-mutilation in schizophrenic children. Arch Gen Psychiatry. 1967;17(2):234-244.
  22. Favazza AR, Conterio K. Female habitual self-mutilators. Acta Psychiatr Scand. 1989;79(3):283-289.
  23. Evans C, Lacey JH. Multiple self-damaging behaviour among alcoholic women. A prevalence study. Br J Psychiatry. 1992;161:643-647.
 

 

First-line treatment: Psychotherapy

Many studies have demonstrated the efficacy of psychotherapy as the primary treatment for NSSI.19-21 Except for patients with Lesch-Nyhan syndrome or other rare neurologic syndromes, the biologic causes of NSSI, including the role of endogenous opioids, are unclear. No medications are FDA-approved for NSSI. Pharmacotherapy may help NSSI patients, but such treatment recommendations are based on clinical experience, and polypharmacy is common.22 Studies have not demonstrated specific benefits or consistent efficacy of pharmacotherapy for NSSI.23

Major NSSI. Prevention is key to addressing major NSSI. Consider atypical antipsychotics for psychotic patients who are preoccupied with religion, the Bible, or sexuality, as well as those who dramatically and suddenly change their appearance by cutting off their hair, engaging in extreme body modification practices, or wearing bizarre clothes.24 In my clinical experience, agitated patients who have committed major NSSI are at high risk for a second episode and should receive pharmacotherapy based on treatment guidelines and hospitalized until the agitation is controlled.

Stereotypic NSSI. Patients with this form of NSSI often cannot articulate what is bothering them. With input from caretakers, assess the likelihood that a patient is reacting to pain. Analgesics may be effective. Also check for infections such as otitis media. Selecting a medication can be challenging. Start with a moderate dose of a selective serotonin reuptake inhibitor (SSRI), then slowly add an atypical antipsychotic, followed by a mood stabilizer, then clonidine, and then a beta blocker; a trial of naltrexone also is an option.23 Behavior therapy is the primary treatment.

Compulsive NSSI. Compulsive NSSI patients typically seek help from dermatologists or family physicians. Literature on psychiatric treatment is limited, but SSRIs, lithium, benzodiazepines, and atypical antipsychotics (for delusional parasitosis) may be effective. N-acetylcysteine, 600 mg twice a day, may relieve trichotillomania.25 Treatment should include psychotherapy.

Impulsive NSSI. Patients who engage in episodic impulsive NSSI should receive pharmacotherapy for underlying psychiatric illnesses such as generalized anxiety disorder, posttraumatic stress disorder, or depression. Do not automatically diagnose borderline personality disorder. Patients whose NSSI behavior is uncontrollable initially should receive high doses of SSRIs that can be lowered when impulsivity decreases, atypical antipsychotics, and a mood stabilizer such as lamotrigine. Psychotherapy is vital, especially dialectical behavior therapy. Cognitive-behavioral and interpersonal therapies also are effective, as is psychodynamic therapy.19-21

NSSI patients and their families may benefit from Web sites that provide information, advice, monitored blogs, and support groups (see Related Resources).

Related Resources

  • Favazza A. Bodies under siege: self-mutilation, nonsuicidal self-injury, and body modification in culture and psychiatry. 3rd ed. Baltimore, MD: Johns Hopkins University Press; 2011.
  • Nock MK. Understanding nonsuicidal self-injury: origins, assessment, and treatment. Washington, DC: American Psychological Association; 2009.
  • Cornell University Family Life Development Center. About self-injury. www.crpsib.com/whatissi.asp.

Drug Brand Names

  • Clonidine • Catapres, Kapvay
  • Lamotrigine • Lamictal
  • Lithium • Eskalith, Lithobid
  • Naltrexone • ReVia

Disclosure

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

References

1. Walsh BW, Rosen PM. Self-mutilation: theory research, and treatment. New York, NY: Guilford Press; 2008:32.

2. Nock MK, Favazza AR. Nonsuicidal self-injury: definition and classification. In: Nock MK ed. Understanding nonsuicidal self-injury: origins, assessment, and treatment. Washington, DC: American Psychological Association; 2009:9-18.

3. Favazza AR, Conterio K. Female habitual self-mutilators. Acta Psychiatr Scand. 1989;79(3):283-289.

4. Favazza A. Bodies under siege: self-mutilation nonsuicidal self-injury, and body modification in culture and psychiatry. 3rd ed. Baltimore, MD: Johns Hopkins University Press; 2011.

5. Gallina R. Tattoos and body piercing. In: Vale V Juno A, eds. Modern primitives. San Francisco, CA: Re/Search Publications; 1989:101-105.

6. Chapman AL, Gratz KL, Brown MZ. Solving the puzzle of deliberate self-harm: the experiential avoidance model. Behav Res Ther. 2006;44(3):371-394.

7. Nock MK, Prinstein MJ. Contextual features and behavioral functions of self-mutilation among adolescents. J Abnorm Psychol. 2005;114(1):140-146.

8. Miller F, Bashkin EA. Depersonalization and self-mutilation. Psychoanal Q. 1974;43(4):638-649.

9. Klonsky ED. The functions of deliberate self-injury: a review of the evidence. Clin Psychol Rev. 2007;27(2):226-239.

10. Nock MK. Actions speak louder than words: an elaborated theoretical model of the social functions of self-injury and other harmful behaviors. Appl Prev Psychol. 2008;12(4):159-168.

11. Nock MK, Prinstein MJ. A functional approach to the assessment of self-mutilative behavior. J Consult Clin Psychol. 2004;72(5):885-890.

12. Nock MK, Prinstein MJ. Contextual features and behavioral functions of self-mutilation among adolescents. J Abnorm Psychol. 2005;114(1):140-146.

13. Favazza AR, Rosenthal RJ. Diagnostic issues in self-mutilation. Hosp Community Psychiatry. 1993;44(2):134-140.

14. Rosen PM, Walsh BW. Patterns of contagion in self-mutilation epidemics. Am J Psychiatry. 1989;146(5):656-658.

15. Large M, Babidge N, Andrews D, et al. Major self-mutilation in the first episode of psychosis. Schizophr Bull. 2009;35(5):1012-1021.

16. Kushner AW. Two cases of auto-castration due to religious delusions. Br J Med Psychol. 1967;40(3):293-298.

17. Moskovitz RA, Byrd T. Rescuing the angel within: PCP-related self-enucleation. Psychosomatics. 1983;24(4):402-403,406.

18. Cleveland SE. Three cases of self-castration. J Nerv Ment Dis. 1956;123(4):386-391.

19. Linehan MM, Comtois KA, Murray AM, et al. Two-year randomized controlled trial and follow-up of dialectical behavior therapy vs therapy by experts for suicidal behaviors and borderline personality disorder. Arch Gen Psychiatry. 2006;63(7):757-766.

20. Kahng S, Iwata BA, Lewin AB. Behavioral treatment of self-injury 1964 to 2000. Am J Ment Retard. 2002;107(3):212-221.

21. Levy KN, Yeomans FE, Diamond D. Psychodynamic treatments of self-injury. J Clin Psychol. 2007;63(11):1105-1120.

22. Lott IT, McGregor M, Engelman L, et al. Longitudinal prescribing patterns for psychoactive medications in community-based individuals with developmental disabilities: utilization of pharmacy records. J Intellect Disabil Res. 2004;48(Pt 6):563-571.

23. Sandman CA. Psychopharmacologic treatment of nonsuicidal self-injury. In: Nock MK ed. Understanding nonsuicidal self-injury: origins, assessment, and treatment. Washington, DC: American Psychological Association; 2009:291-322.

24. Sweeny S, Zamecnik K. Predictors of self-mutilation in patients with schizophrenia. Am J Psychiatry. 1981;138(8):1086-1089.

25. Grant JE, Odlaug BL, Kim SW. N-acetylcysteine a glutamate modulator, in the treatment of trichotillomania: a double-blind, placebo-controlled study. Arch Gen Psychiatry. 2009;66(7):756-763.

References

1. Walsh BW, Rosen PM. Self-mutilation: theory research, and treatment. New York, NY: Guilford Press; 2008:32.

2. Nock MK, Favazza AR. Nonsuicidal self-injury: definition and classification. In: Nock MK ed. Understanding nonsuicidal self-injury: origins, assessment, and treatment. Washington, DC: American Psychological Association; 2009:9-18.

3. Favazza AR, Conterio K. Female habitual self-mutilators. Acta Psychiatr Scand. 1989;79(3):283-289.

4. Favazza A. Bodies under siege: self-mutilation nonsuicidal self-injury, and body modification in culture and psychiatry. 3rd ed. Baltimore, MD: Johns Hopkins University Press; 2011.

5. Gallina R. Tattoos and body piercing. In: Vale V Juno A, eds. Modern primitives. San Francisco, CA: Re/Search Publications; 1989:101-105.

6. Chapman AL, Gratz KL, Brown MZ. Solving the puzzle of deliberate self-harm: the experiential avoidance model. Behav Res Ther. 2006;44(3):371-394.

7. Nock MK, Prinstein MJ. Contextual features and behavioral functions of self-mutilation among adolescents. J Abnorm Psychol. 2005;114(1):140-146.

8. Miller F, Bashkin EA. Depersonalization and self-mutilation. Psychoanal Q. 1974;43(4):638-649.

9. Klonsky ED. The functions of deliberate self-injury: a review of the evidence. Clin Psychol Rev. 2007;27(2):226-239.

10. Nock MK. Actions speak louder than words: an elaborated theoretical model of the social functions of self-injury and other harmful behaviors. Appl Prev Psychol. 2008;12(4):159-168.

11. Nock MK, Prinstein MJ. A functional approach to the assessment of self-mutilative behavior. J Consult Clin Psychol. 2004;72(5):885-890.

12. Nock MK, Prinstein MJ. Contextual features and behavioral functions of self-mutilation among adolescents. J Abnorm Psychol. 2005;114(1):140-146.

13. Favazza AR, Rosenthal RJ. Diagnostic issues in self-mutilation. Hosp Community Psychiatry. 1993;44(2):134-140.

14. Rosen PM, Walsh BW. Patterns of contagion in self-mutilation epidemics. Am J Psychiatry. 1989;146(5):656-658.

15. Large M, Babidge N, Andrews D, et al. Major self-mutilation in the first episode of psychosis. Schizophr Bull. 2009;35(5):1012-1021.

16. Kushner AW. Two cases of auto-castration due to religious delusions. Br J Med Psychol. 1967;40(3):293-298.

17. Moskovitz RA, Byrd T. Rescuing the angel within: PCP-related self-enucleation. Psychosomatics. 1983;24(4):402-403,406.

18. Cleveland SE. Three cases of self-castration. J Nerv Ment Dis. 1956;123(4):386-391.

19. Linehan MM, Comtois KA, Murray AM, et al. Two-year randomized controlled trial and follow-up of dialectical behavior therapy vs therapy by experts for suicidal behaviors and borderline personality disorder. Arch Gen Psychiatry. 2006;63(7):757-766.

20. Kahng S, Iwata BA, Lewin AB. Behavioral treatment of self-injury 1964 to 2000. Am J Ment Retard. 2002;107(3):212-221.

21. Levy KN, Yeomans FE, Diamond D. Psychodynamic treatments of self-injury. J Clin Psychol. 2007;63(11):1105-1120.

22. Lott IT, McGregor M, Engelman L, et al. Longitudinal prescribing patterns for psychoactive medications in community-based individuals with developmental disabilities: utilization of pharmacy records. J Intellect Disabil Res. 2004;48(Pt 6):563-571.

23. Sandman CA. Psychopharmacologic treatment of nonsuicidal self-injury. In: Nock MK ed. Understanding nonsuicidal self-injury: origins, assessment, and treatment. Washington, DC: American Psychological Association; 2009:291-322.

24. Sweeny S, Zamecnik K. Predictors of self-mutilation in patients with schizophrenia. Am J Psychiatry. 1981;138(8):1086-1089.

25. Grant JE, Odlaug BL, Kim SW. N-acetylcysteine a glutamate modulator, in the treatment of trichotillomania: a double-blind, placebo-controlled study. Arch Gen Psychiatry. 2009;66(7):756-763.

Issue
Current Psychiatry - 11(03)
Issue
Current Psychiatry - 11(03)
Page Number
21-26
Page Number
21-26
Publications
Publications
Article Type
Display Headline
Nonsuicidal self-injury: How categorization guides treatment
Display Headline
Nonsuicidal self-injury: How categorization guides treatment
Legacy Keywords
nonsuicidal self-injury; NSSI; self-harm; pathological NSSI; types of self-harm
Legacy Keywords
nonsuicidal self-injury; NSSI; self-harm; pathological NSSI; types of self-harm
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media