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

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

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

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

Psychological first aid: Emergency care for terrorism and disaster survivors

Article Type
Changed
Tue, 12/11/2018 - 15:10
Display Headline
Psychological first aid: Emergency care for terrorism and disaster survivors

The night started like any other for Dr. Z. Kids in bed (too late) by 10:30, dog out by 11, fell asleep reading journal by 11:15. Sirens jolt her out of a solid stage 4. Her eyes widen, pulse quickens, mouth dries as she follows the glow of the TV into the living room. On TV, fire frames shots of tearful faces, body bags, and firefighters in protective suits. She catches the announcer’s voice: “…explosion at City Power and Light nuclear facility at 3:10 AM today. Fifty-five are known dead, and thousands are being told to evacuate. The blast’s cause is unknown, but terrorism is suspected.”

As her numbness slowly ebbs, Dr. Z’s questions rise insistently. How can I help the survivors? My patients? My children? What if the media call me? How could I have been better prepared for this?

Disaster shakes us to our human and biological core. More than any other clinical encounter, it reminds us that psychiatrists share the vulnerabilities of those we seek to help. Yet it also reminds us that even simple concepts and interventions can mobilize the healing process.

Are you ready to provide emergency psychiatric care following a disaster in your community—be it a nuclear accident, tornado, airplane crash, or terrorist act? Here is evidence—some counterintuitive—that can help you prepare.

Table 1

‘Psychological first aid’ for disaster survivors

Re-create sense of safety
  • Provide for basic needs (food, clothing, medical care)
  • Ensure that survivors are safe and protected from reminders of the event
  • Protect them from onlookers and the media
  • Help them establish a “personal space” and preserve privacy and modesty
Encourage social support
  • Help survivors connect with family and friends (most urgently, children with parents)
  • Educate family and friends about survivors’ normal reactions and how they can help
Re-establish sense of efficacy
  • Give survivors accurate, simple information about plans and events
  • Allow survivors to discuss events and feelings, but do not probe
  • Encourage them to re-establish normal routines and roles when possible
  • Help resolve practical problems, such as getting transportation or relief vouchers
  • Discuss self-care and strategies to reduce anxiety, such as grounding and relaxation techniques
  • Encourage survivors to support and assist others

Disaster’s psychobiology

Human reactions to disaster are often conceptualized as the mammalian survival response: flight, fight, and fright (freezing). In most cases, these reactions are adaptive and dissipate after safety is restored. Posttraumatic stress disorder (PTSD) develops in about 5% of natural disaster victims and 33% of mass shooting victims.1

Responses that do go awry appear to be associated with abnormally low cortisol and persistent adrenergic activation, leading to sensitization of the fear response.2 Reminders of trauma or persistent stressors—such as pain, problems with finances or housing, or bereavement—may exacerbate sensitization. On the other hand, preclinical studies suggest that social support3,4 and active coping5 mitigate physiologic stress responses, confirming numerous clinical observations that associate lack of social support and avoidant coping with eventual PTSD development.

Three basics. Just as our emergency medicine colleagues must often revert to life-support basics, we must remind ourselves of biology’s three basics for psychic resilience:

  • safety (including—as much as possible—protection from reminders of trauma and ongoing stress)
  • meaningful social connection
  • re-establishing a sense of efficacy to overcome helplessness (Table 1).

Like the stress response, these protective factors seem hard-wired into our biological make-up. They form the foundation for all phases of disaster psychiatry interventions, from planning to immediate interventions through longterm follow-up.

Disaster planning

As a mental health professional, plan to operate within established disaster plans and agencies, not only for the sake of efficiency but also because structure and support are paramount in disaster situations. Check with the American Psychiatric Association’s local branch to determine if a disaster mental health plan exists. If not, explore how to work directly with local American Red Cross chapters and hospitals, which recruit personnel for the Disaster Medical Assistance Teams mobilized by the public health service.

Table 2

Normal reactions to disaster for adults and children

All ages
EmotionalShock, fear, grief, anger, guilt, shame, helplessness, hopelessness, numbness, emptiness Decreased ability to feel interest, pleasure, love
CognitiveConfusion, disorientation, indecisiveness, worry, shortened attention span, poor concentration, memory difficulties, unwanted memories, self-blame
PhysicalTension, fatigue, edginess, insomnia, generalized aches and pains, startling easily, rapid heartbeat, nausea, decreased appetite and sex drive
InterpersonalDifficulties being intimate, being over-controlling, feeling rejected or abandoned
Children’s age-specific disaster responses
PreschoolSeparation fears, regression, fussiness, temper tantrums, somatization Sleep disturbances including nightmares, somnambulism, night terrors
School-ageMay still have the above, as well as excessive guilt and worries about others’ safety, poor concentration and loss of school performance, repetitious re-telling or play related to trauma
AdolescentDepression, acting out, wish for revenge, sleeping and eating disturbances, altered view of the future
 

 

Immediate disaster mental health plans vary in detail according to local needs and resources but should at least address:

  • providing on-site interventions
  • disseminating information about responses to trauma
  • identifying and publicizing local mental health resources.

Immediate interventions

Immediately following a disaster, psychiatrists are frequently asked to assist with on-site crisis and medical interventions, evaluate survivors with unusual or intense reactions, and provide public education about psychological reactions to disaster.

On-site response. All responders, regardless of discipline, should provide disaster survivors with “psychological first aid,” which is directed at reestablishing safety, connection, and efficacy. Basic crisis intervention principles are useful when support and reassurance are not enough.6

For example, relaxation exercises can reduce anxiety and improve sleep. Use focused, structured relaxation tools—such as progressive muscle relaxation and breathing training—as unstructured exercises can exacerbate dissociation and re-experiencing. Grounding techniques, by which survivors learn to focus all senses on immediate surroundings, often alleviate dissociation and flashbacks.

Care for children. Because children’s reactions to disaster greatly depend on their caregivers’ responses (social referencing), focus acute interventions for children on:

  • re-connecting them with their families
  • reducing caregivers’ distress
  • educating caregivers about providing age appropriate information and support (see Related resources).

Medical care. As physicians, psychiatrists may be called upon to intervene medically. Although it is generally advisable to stay within our usual practice, medical personnel may be in short supply. Fortunately, Good Samaritan laws exist in every state, and the potential for a successful malpractice suit against a physician responding in a disaster is almost zero, unless the physician’s performance is grossly negligent (such as moving the neck of a patient with obvious head or neck injuries).7

Principles regarding informed consent and right to refuse treatment—along with the usual exceptions—apply during disasters.

Evaluating survivors in shelters and hospitals requires knowing the normal and abnormal responses to disaster, being able to differentially diagnose changes in mental status, and understanding risk factors for trauma’s psychiatric sequelae. Aside from PTSD, trauma’s long-term effects include other anxiety disorders, depression, substance abuse, and eating disorders. In addition to the usual components of a psychiatric evaluation, assessments must address event-related factors such as proximity to the disaster, loss of significant others or property, physical injuries, immediate needs, and social support.8

Normal stress reactions. Frequently described as “a normal response to an abnormal situation,” the normal stress reaction is multidimensional and depends on the person’s developmental level (Table 2). About 10% to 25% of survivors experience intense affect and dissociation, whereas a similar number may appear unusually calm.

Interventions beyond the“first aid” described above are not usually needed unless individuals:

  • are a danger to themselves or others
  • are psychotic
  • have no social supports
  • cannot perform tasks necessary for self-care and to begin the recovery process.

Always re-assess when there is any question about a survivor’s immediate reaction to trauma.

In DSM-IV-TR’s trauma-related diagnoses, the symptom clusters often do not capture many disaster survivors’ subjective experience: the shattering of fundamental beliefs regarding themselves (invulnerability), the world (predictability, safety), and others (trust, benevolence).9 By empathizing with these responses, you can help survivors feel less isolated and estranged.

Differential diagnosis. Survivors’ mental status changes may be manifestations of the stress response, but they also may represent:

  • exacerbations of pre-existing psychiatric or general medical conditions
  • hypoxemia, hypovolemia, or CNS trauma from physical injury
  • responses to medications used for resuscitation or pain control, such as atropine, epinephrine, lidocaine, or morphine.

Effects of bioterrorism agents must also be considered. For example, organophosphorus compounds such as the nerve agents sarin and soman can cause impaired concentration, depression, and anxiety. Anthrax can cause rapidly progressing meningitis.10 Delirium must be differentiated from dissociation; patients with dissociation can be re-oriented, and changes will resolve with time rather than fluctuate.11

Psychopathology risk factors. Multiple studies have addressed risk factors for post-disaster psychiatric sequelae (usually PTSD). In general, risk increases with repeated trauma exposure (including TV viewing), prior trauma, lack of social support, injury, pre-existing psychiatric problems, traumatic bereavement (having witnessed the violent death of a loved one), avoidant coping, and having strong negative beliefs about the meanings of normal stress reactions such as tearfulness, anxiety, and insomnia.

Because a recent meta-analysis supports these observations,12 follow-up evaluation for signs of PTSD is recommended for:

  • survivors with one or more of the risk factors discussed above
  • vulnerable groups such as rescue workers, children, and dependent individuals
  • survivors whose symptoms persist after 2 months.13

Decompensation immediately after a disaster seems to be the exception for psychiatric patients, despite their longer-term vulnerability. One psychiatrist who in 1989 survived Hurricane Hugo—the most intense storm to strike the Mid-Atlantic region in 100 years—noted that demand for inpatient psychiatric services did not increase in the storm’s aftermath. The only patient calls she received were inquiries about her own physical safety.14

 

 

Caregivers and rescue workers—including psychi-atrists—are also disaster survivors, and you need to tend to your needs for safety and support. Consult frequently with colleagues within and outside the disaster area, as much for support as for information and guidance.15 Remember also that rescue workers are occasionally targets for victims’ rage at their circumstances. Anticipating and explaining this displacement reduces its toxicity.

Using medications

Uses psychotropics judiciously in the first 48 hours of trauma. Medication effects may interfere with neurologic assessment of the injured, and monitoring and follow-up may not be possible.

However, drug therapy should start quickly when survivors are acutely psychotic or their behavior endangers themselves, others, or the milieu. Medications usually include a fast-acting benzodiazepine and/or an antipsychotic, as described in guidelines for managing agitation.16 Always provide structure and supervision for medicated patients.

No guidelines exist for using medications to manage distressing—but less-severe—acute stress-related symptoms. Some experts advocate using adrenergic antagonists such as clonidine, guanfacine, and beta blockers to reduce excessive arousal. These drugs have not been adequately studied in this setting, however, and may harm those with cardiovascular instability from preexisting conditions or injuries.

Table 3

Psychoeducation: Simple messages for workers and trauma survivors

Get adequate rest, food, sleep
Avoid exposure to trauma cues, including TV images
Seek support from loved ones and peers
Talk about events and feelings only if this feels comfortable and helpful
Return to normal routine as much as possible
Take action to rebuild, but at a reasonable pace
Reach out to others who may need assistance
Get help:
  • immediately for abnormal reactions (psychosis, suicidality, risky behavior including substance abuse)
  • if normal reactions (insomnia, anxiety, mild dissociation) persist beyond 2 months
  • if at high risk for persistent reaction (bereaved, injured, prior trauma or psychiatric disorder, no social support)

Short-term (<1 week) benzodiazepine use for panic symptoms and severe insomnia is acceptable, but longer-term use may increase PTSD risk.17 A selective serotonin reuptake inhibitor may help individuals with pre-existing PTSD or depression, if you can arrange follow-up.

In the aftermath

‘Debriefing.’ Critical incident stress debriefing (CISD) is a structured, one-session group intervention in which survivors’ experiences and emotional reactions are discussed and education and follow-up recommendations are provided. Developed by Mitchell in 1983,18 CISD was widely used until systematic evaluations revealed that it did not alleviate psychological distress or prevent PTSD.19

Table 4

How to interact with news media during and after a disaster

For organizations
Identify a spokesperson with media experience beforehand
Ensure that the spokesperson is well-informed about all aspects of the disaster
For spokespersons
Always
  • Reply immediately to news media requests; find out the reporter’s deadline and refer the caller if you cannot meet it
  • Clearly identify yourself and the organization you represent
  • Understand what the reporter needs—what’s “behind the story”
  • Use simple, declarative sentences; avoid medical jargon
  • Emphasize one or two points
  • Be accurate and honest but use a positive, hopeful frame
  • On TV, look at the interviewer, not the camera
Never
  • Speak off the record
  • Discuss individual cases
  • Speculate on diagnosis or treatment for someone you have not examined (such as a terrorist leader)

Thus, although survivor meetings may provide information, education, screening, and support, avoid detailed discussions of events and emotions. Any meetings should be conducted by mental health clinicians and should not be mandatory. Reserve the term “debriefing” for operational reviews by rescue personnel.20

Public education. Educate survivors, rescue workers, health care providers, teachers, and relief agency workers. Provide concise, simple messages as suggested in Table 3. News media provide our most effective means of reaching out to survivors, which is why having a pre-existing relationship is so important. Some guidelines for working with the media are presented in Table 4.

Outreach. Numerous educational resources are available for survivors and their caregivers (see Related resources). Other potentially useful outreach tools include:

  • meetings with teachers’ organizations
  • continuing medical education activities for primary care providers
  • telephone hot lines.

Legal and ethical issues

Disaster scenes are chaotic and informal, and professionals must be flexible, often providing general support and information rather than specific clinical interventions. However, it is important in each encounter to decide whether a patient-physician relationship has begun.

As a general rule, a physician-patient relationship is established whenever diagnosis or treatment is discussed. Once that happens, briefly document:

  • signs and symptoms
  • working diagnosis
  • suicide or violence potential
  • treatment and/or follow-up plans.

Confidentiality may be difficult to preserve in chaotic situations involving workers from many agencies. Even in disasters, however, you must obtain permission before sharing information unless the individual’s situation is emergent.

Table 5

 

 

Keys to effective disaster psychiatry

Be prepared
  • Contact your local American Psychiatric Association district branch or American Red Cross chapter to learn about existing mental health disaster plans
  • Remember the three basics of psychic resiliency: safety, support, and efficacy
  • Know your place, ask for help, and use existing resources
Care for survivors
  • Your presence alone may be healing
  • Assess thoroughly; symptoms may be related to medical issues or pre-existing conditions, not just stress response
  • Do no harm; avoid psychological debriefing interventions and unnecessary medications
  • Follow up with survivors who exhibit risk factors or symptoms persisting >2 months
  • Consider trauma-focused CBT for survivors with acute stress disorder
Care for yourself
  • As a survivor, tend to your own safety and self-care needs
  • Consult frequently with colleagues within and outside the disaster area for support, information, and guidance

Long-term interventions

Longer-term disaster interventions include continued outreach and education and needed follow-up services. Existing structures may provide effective follow-up, but additional resources are often needed.

Federal programs. Following a presidentialdeclared disaster, the Federal Emergency Management Agency (FEMA) provides funding for crisis counseling. Programs are typically funded for 9 to 15 months and administered through the emergency services and disaster relief branch of the Substance Abuse and Mental Health Services Administration (SAMHSA) and community mental health organizations. Examples include Project Heartland following the 1995 Oklahoma City federal building bombing and Project COPE following California’s 1989 Loma Prieta earthquake.

Cognitive-behavioral therapy. For adult survivors with acute stress disorder, specific cognitive-behavioral therapy (CBT) provided by trained therapists may prevent PTSD and other trauma sequelae, such as depression.21 CBT interventions may begin as early as 2 weeks after trauma and focus sequentially on anxiety management, cognitive restructuring, imaginal exposure followed by in vivo exposure, and relapse prevention.

Three controlled trials found 6-month PTSD rates of 14% to 20% among acute stress disorder patients treated with CBT, compared with 58% to 67% with supportive counseling.22-24 Although studies of interventions immediately following trauma are lacking, trauma-focused CBT is also recommended for children.25 Evidence-based treatments for PTSD are discussed in detail elsewhere.26

Not unprepared after all

With some reflection, Dr. Z realized she had the tools to help her community. Her feelings of helplessness receded as she envisioned how she could help survivors understand their experiences, re-create a sense of safety, restore important connections to loved ones, and begin to rebuild their lives (Table 5).

Related resources

For clinicians

  • Young BH, Ford JD, Ruzek JI, et al. Disaster mental health: a guidebook for clinicians and administrators. Washington, DC: National Center for Post-Traumatic Stress Disorder, 1998. http://ncptsd.org/publications/disaster/index.html
  • Hillman JL. Crisis intervention and trauma: new approaches to evidence-based practice. New York: Kluwer Academic/Plenum Publishers, 2002.
  • Office of the Surgeon General Web site on medical aspects of nuclear, biological, and chemical warfare. http://www.nbc-med.org

For survivors and clinicians

Drug brand names

  • Clonidine • Catapres
  • Guanfacine • Tenex

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. Effects of traumatic stress in a disaster situation. Washington, DC: National Center for Post-Traumatic Stress Disorder. Department of Veterans Affairs, September 12, 2001 (Accessed March 31, 2004 at http://www.ncptsd.org/facts/disasters/fs_effects_disaster.html).

2. Yehuda R, McFarlane AC, Shalev AY. Predicting the development of post-traumatic stress disorder from the acute response to a traumatic event. Biol Psychiatry 1998;44:1305-13.

3. Levine S, Lyons DM, Schatzberg AF. Psychobiological consequences of social relationships. Ann NY Acad Sci 1997;807:210-18.

4. Uchino BN, Garvey TS. The availability of social support reduces cardiovascular reactivity to acute psychological stress. J Behav Med 1997;20:15-27.

5. LeDoux JE, Gorman JM. A call to action: overcoming anxiety through active coping. Am J Psychiatry 2001;158:1953-5.

6. Hillman JL. Crisis intervention and trauma: new approaches to evidence-based practice. New York: Kluwer Academic/Plenum Publishers, 2002.

7. Daniels G. Good Samaritan acts. Emerg Med Clin North Am 1999;17:491-504.

8. Mental-health intervention for disasters. Washington, DC: National Center for Post-Traumatic Stress Disorder, Department of Veterans Affairs, Sept. 12, 2001. (Accessed March 31, 2004 at http://www.ncptsd.org/facts/disasters/fs_treatment_disaster.html.)

9. Difede J, Apfeldorf WJ, Cloitre M, et al. Acute psychiatric responses to the explosion at the World Trade Center: a case series. J Nerv Ment Dis 1997;185:519-22.

10. DiGiovanni C. Domestic terrorism with chemical or biological agents: psychiatric aspects. Am J Psychiatry 1999;156:1500-5.

11. Rundle JR. Psychiatric issues in medical-surgical disaster casualties: consultation-liaison approach. Psychiatr Q 2000;71:245-58.

12. Brewin CR, Andrews B, Valentine JD. Meta-analysis of risk factors for post-traumatic stress disorder in trauma-exposed adults. J Consult Clin Psychol 2000;68:748-66.

13. Mental health and mass violence: evidence-based early psychological intervention for victims/survivors of mass violence. Washington, DC: National Institute of Mental Health, Nov. 1, 2002 (Accessed Oct. 29, 2003 at http://www.nimh.nih.gov/research/massviolence.pdf.)

14. Austin LS. Organizing a disaster response program in one’s home community. In: Austin LS (ed). Responding to disaster: a guide for mental health professionals. Washington, DC: American Psychiatric Publishing, 1992;53-68.

15. Rousseau AW. Notes from Oklahoma City’s recovery. In: Hall RCW, Norwood AE (eds). Disaster psychiatry handbook. Washington, DC: American Psychiatric Association Committee on Psychiatric Dimensions of Disaster, undated:3-11. (Accessed Oct. 29, 2003 at http://www.psych.org/disasterpsych/pdfs/apadisasterhandbk.pdf.)

16. Yildiz A, Sachs GS, Turgay A. Pharmacological management of agitation in emergency settings. Emerg Med J 2003;20:339-46.

17. Gelpin E, Bonne O, Peri T, et al. Treatment of recent trauma survivors with benzodiazepines: a prospective study. J Clin Psychiatry 1996;57:390-4.

18. Mitchell JT. When disaster strikes. J Em Med Serv 1983;8:36-9.

19. Rose S, Bisson J, Wessely S. Psychological debriefing for preventing posttraumatic stress disorder (PTSD). Cochrane Database Syst Rev 2002;(2):CD000560.-

20. Black J. Forming the libidinal cocoon: the Dallas airplane crashes, the Guadalupe river drownings, and Hurricane Hugo in the Virgin Islands. In: Austin LS (ed). Responding to disaster: a guide for mental health professionals. Washington DC: American Psychiatric Publishing, 1992;169:84.-

21. Ehlers A, Clark DM. Early psychological interventions for adult survivors of trauma: a review. Biol Psychiatry 2003;53:817-26.

22. Bryant RA, Moulds M, Guthrie R, Nixon RD. Treating acute stress disorder following mild traumatic brain injury. Am J Psychiatry 2003;160:585-7.

23. Bryant RA, Sackville T, Dang ST, et al. Treating acute stress disorder: an evaluation of cognitive behavior therapy and supportive counseling techniques. Am J Psychiatry 1999;156:1780-6.

24. Bryant RA, Harvey AG, Dang ST, et al. Treatment of acute stress disorder: a comparison of cognitive-behavioral therapy and supportive counseling. J Consult Clin Psychol 1998;66:862-6.

25. Cohen JA. Treating acute posttraumatic reactions in children and adolescents. Biol Psychiatry 2003;53:827-33.

26. Foa EB, Keane TM, Friedman MJ. Effective treatments for PTSD. New York: Guilford Press, 2000.

Article PDF
Author and Disclosure Information

Nancy L. Cloak, MD
Staff psychiatrist Portland State University Portland, OR

Pamela Edwards, MD
Assistant professor, Department of Psychiatry Oregon Health and Science University Portland

Issue
Current Psychiatry - 03(05)
Publications
Topics
Page Number
12-23
Sections
Author and Disclosure Information

Nancy L. Cloak, MD
Staff psychiatrist Portland State University Portland, OR

Pamela Edwards, MD
Assistant professor, Department of Psychiatry Oregon Health and Science University Portland

Author and Disclosure Information

Nancy L. Cloak, MD
Staff psychiatrist Portland State University Portland, OR

Pamela Edwards, MD
Assistant professor, Department of Psychiatry Oregon Health and Science University Portland

Article PDF
Article PDF

The night started like any other for Dr. Z. Kids in bed (too late) by 10:30, dog out by 11, fell asleep reading journal by 11:15. Sirens jolt her out of a solid stage 4. Her eyes widen, pulse quickens, mouth dries as she follows the glow of the TV into the living room. On TV, fire frames shots of tearful faces, body bags, and firefighters in protective suits. She catches the announcer’s voice: “…explosion at City Power and Light nuclear facility at 3:10 AM today. Fifty-five are known dead, and thousands are being told to evacuate. The blast’s cause is unknown, but terrorism is suspected.”

As her numbness slowly ebbs, Dr. Z’s questions rise insistently. How can I help the survivors? My patients? My children? What if the media call me? How could I have been better prepared for this?

Disaster shakes us to our human and biological core. More than any other clinical encounter, it reminds us that psychiatrists share the vulnerabilities of those we seek to help. Yet it also reminds us that even simple concepts and interventions can mobilize the healing process.

Are you ready to provide emergency psychiatric care following a disaster in your community—be it a nuclear accident, tornado, airplane crash, or terrorist act? Here is evidence—some counterintuitive—that can help you prepare.

Table 1

‘Psychological first aid’ for disaster survivors

Re-create sense of safety
  • Provide for basic needs (food, clothing, medical care)
  • Ensure that survivors are safe and protected from reminders of the event
  • Protect them from onlookers and the media
  • Help them establish a “personal space” and preserve privacy and modesty
Encourage social support
  • Help survivors connect with family and friends (most urgently, children with parents)
  • Educate family and friends about survivors’ normal reactions and how they can help
Re-establish sense of efficacy
  • Give survivors accurate, simple information about plans and events
  • Allow survivors to discuss events and feelings, but do not probe
  • Encourage them to re-establish normal routines and roles when possible
  • Help resolve practical problems, such as getting transportation or relief vouchers
  • Discuss self-care and strategies to reduce anxiety, such as grounding and relaxation techniques
  • Encourage survivors to support and assist others

Disaster’s psychobiology

Human reactions to disaster are often conceptualized as the mammalian survival response: flight, fight, and fright (freezing). In most cases, these reactions are adaptive and dissipate after safety is restored. Posttraumatic stress disorder (PTSD) develops in about 5% of natural disaster victims and 33% of mass shooting victims.1

Responses that do go awry appear to be associated with abnormally low cortisol and persistent adrenergic activation, leading to sensitization of the fear response.2 Reminders of trauma or persistent stressors—such as pain, problems with finances or housing, or bereavement—may exacerbate sensitization. On the other hand, preclinical studies suggest that social support3,4 and active coping5 mitigate physiologic stress responses, confirming numerous clinical observations that associate lack of social support and avoidant coping with eventual PTSD development.

Three basics. Just as our emergency medicine colleagues must often revert to life-support basics, we must remind ourselves of biology’s three basics for psychic resilience:

  • safety (including—as much as possible—protection from reminders of trauma and ongoing stress)
  • meaningful social connection
  • re-establishing a sense of efficacy to overcome helplessness (Table 1).

Like the stress response, these protective factors seem hard-wired into our biological make-up. They form the foundation for all phases of disaster psychiatry interventions, from planning to immediate interventions through longterm follow-up.

Disaster planning

As a mental health professional, plan to operate within established disaster plans and agencies, not only for the sake of efficiency but also because structure and support are paramount in disaster situations. Check with the American Psychiatric Association’s local branch to determine if a disaster mental health plan exists. If not, explore how to work directly with local American Red Cross chapters and hospitals, which recruit personnel for the Disaster Medical Assistance Teams mobilized by the public health service.

Table 2

Normal reactions to disaster for adults and children

All ages
EmotionalShock, fear, grief, anger, guilt, shame, helplessness, hopelessness, numbness, emptiness Decreased ability to feel interest, pleasure, love
CognitiveConfusion, disorientation, indecisiveness, worry, shortened attention span, poor concentration, memory difficulties, unwanted memories, self-blame
PhysicalTension, fatigue, edginess, insomnia, generalized aches and pains, startling easily, rapid heartbeat, nausea, decreased appetite and sex drive
InterpersonalDifficulties being intimate, being over-controlling, feeling rejected or abandoned
Children’s age-specific disaster responses
PreschoolSeparation fears, regression, fussiness, temper tantrums, somatization Sleep disturbances including nightmares, somnambulism, night terrors
School-ageMay still have the above, as well as excessive guilt and worries about others’ safety, poor concentration and loss of school performance, repetitious re-telling or play related to trauma
AdolescentDepression, acting out, wish for revenge, sleeping and eating disturbances, altered view of the future
 

 

Immediate disaster mental health plans vary in detail according to local needs and resources but should at least address:

  • providing on-site interventions
  • disseminating information about responses to trauma
  • identifying and publicizing local mental health resources.

Immediate interventions

Immediately following a disaster, psychiatrists are frequently asked to assist with on-site crisis and medical interventions, evaluate survivors with unusual or intense reactions, and provide public education about psychological reactions to disaster.

On-site response. All responders, regardless of discipline, should provide disaster survivors with “psychological first aid,” which is directed at reestablishing safety, connection, and efficacy. Basic crisis intervention principles are useful when support and reassurance are not enough.6

For example, relaxation exercises can reduce anxiety and improve sleep. Use focused, structured relaxation tools—such as progressive muscle relaxation and breathing training—as unstructured exercises can exacerbate dissociation and re-experiencing. Grounding techniques, by which survivors learn to focus all senses on immediate surroundings, often alleviate dissociation and flashbacks.

Care for children. Because children’s reactions to disaster greatly depend on their caregivers’ responses (social referencing), focus acute interventions for children on:

  • re-connecting them with their families
  • reducing caregivers’ distress
  • educating caregivers about providing age appropriate information and support (see Related resources).

Medical care. As physicians, psychiatrists may be called upon to intervene medically. Although it is generally advisable to stay within our usual practice, medical personnel may be in short supply. Fortunately, Good Samaritan laws exist in every state, and the potential for a successful malpractice suit against a physician responding in a disaster is almost zero, unless the physician’s performance is grossly negligent (such as moving the neck of a patient with obvious head or neck injuries).7

Principles regarding informed consent and right to refuse treatment—along with the usual exceptions—apply during disasters.

Evaluating survivors in shelters and hospitals requires knowing the normal and abnormal responses to disaster, being able to differentially diagnose changes in mental status, and understanding risk factors for trauma’s psychiatric sequelae. Aside from PTSD, trauma’s long-term effects include other anxiety disorders, depression, substance abuse, and eating disorders. In addition to the usual components of a psychiatric evaluation, assessments must address event-related factors such as proximity to the disaster, loss of significant others or property, physical injuries, immediate needs, and social support.8

Normal stress reactions. Frequently described as “a normal response to an abnormal situation,” the normal stress reaction is multidimensional and depends on the person’s developmental level (Table 2). About 10% to 25% of survivors experience intense affect and dissociation, whereas a similar number may appear unusually calm.

Interventions beyond the“first aid” described above are not usually needed unless individuals:

  • are a danger to themselves or others
  • are psychotic
  • have no social supports
  • cannot perform tasks necessary for self-care and to begin the recovery process.

Always re-assess when there is any question about a survivor’s immediate reaction to trauma.

In DSM-IV-TR’s trauma-related diagnoses, the symptom clusters often do not capture many disaster survivors’ subjective experience: the shattering of fundamental beliefs regarding themselves (invulnerability), the world (predictability, safety), and others (trust, benevolence).9 By empathizing with these responses, you can help survivors feel less isolated and estranged.

Differential diagnosis. Survivors’ mental status changes may be manifestations of the stress response, but they also may represent:

  • exacerbations of pre-existing psychiatric or general medical conditions
  • hypoxemia, hypovolemia, or CNS trauma from physical injury
  • responses to medications used for resuscitation or pain control, such as atropine, epinephrine, lidocaine, or morphine.

Effects of bioterrorism agents must also be considered. For example, organophosphorus compounds such as the nerve agents sarin and soman can cause impaired concentration, depression, and anxiety. Anthrax can cause rapidly progressing meningitis.10 Delirium must be differentiated from dissociation; patients with dissociation can be re-oriented, and changes will resolve with time rather than fluctuate.11

Psychopathology risk factors. Multiple studies have addressed risk factors for post-disaster psychiatric sequelae (usually PTSD). In general, risk increases with repeated trauma exposure (including TV viewing), prior trauma, lack of social support, injury, pre-existing psychiatric problems, traumatic bereavement (having witnessed the violent death of a loved one), avoidant coping, and having strong negative beliefs about the meanings of normal stress reactions such as tearfulness, anxiety, and insomnia.

Because a recent meta-analysis supports these observations,12 follow-up evaluation for signs of PTSD is recommended for:

  • survivors with one or more of the risk factors discussed above
  • vulnerable groups such as rescue workers, children, and dependent individuals
  • survivors whose symptoms persist after 2 months.13

Decompensation immediately after a disaster seems to be the exception for psychiatric patients, despite their longer-term vulnerability. One psychiatrist who in 1989 survived Hurricane Hugo—the most intense storm to strike the Mid-Atlantic region in 100 years—noted that demand for inpatient psychiatric services did not increase in the storm’s aftermath. The only patient calls she received were inquiries about her own physical safety.14

 

 

Caregivers and rescue workers—including psychi-atrists—are also disaster survivors, and you need to tend to your needs for safety and support. Consult frequently with colleagues within and outside the disaster area, as much for support as for information and guidance.15 Remember also that rescue workers are occasionally targets for victims’ rage at their circumstances. Anticipating and explaining this displacement reduces its toxicity.

Using medications

Uses psychotropics judiciously in the first 48 hours of trauma. Medication effects may interfere with neurologic assessment of the injured, and monitoring and follow-up may not be possible.

However, drug therapy should start quickly when survivors are acutely psychotic or their behavior endangers themselves, others, or the milieu. Medications usually include a fast-acting benzodiazepine and/or an antipsychotic, as described in guidelines for managing agitation.16 Always provide structure and supervision for medicated patients.

No guidelines exist for using medications to manage distressing—but less-severe—acute stress-related symptoms. Some experts advocate using adrenergic antagonists such as clonidine, guanfacine, and beta blockers to reduce excessive arousal. These drugs have not been adequately studied in this setting, however, and may harm those with cardiovascular instability from preexisting conditions or injuries.

Table 3

Psychoeducation: Simple messages for workers and trauma survivors

Get adequate rest, food, sleep
Avoid exposure to trauma cues, including TV images
Seek support from loved ones and peers
Talk about events and feelings only if this feels comfortable and helpful
Return to normal routine as much as possible
Take action to rebuild, but at a reasonable pace
Reach out to others who may need assistance
Get help:
  • immediately for abnormal reactions (psychosis, suicidality, risky behavior including substance abuse)
  • if normal reactions (insomnia, anxiety, mild dissociation) persist beyond 2 months
  • if at high risk for persistent reaction (bereaved, injured, prior trauma or psychiatric disorder, no social support)

Short-term (<1 week) benzodiazepine use for panic symptoms and severe insomnia is acceptable, but longer-term use may increase PTSD risk.17 A selective serotonin reuptake inhibitor may help individuals with pre-existing PTSD or depression, if you can arrange follow-up.

In the aftermath

‘Debriefing.’ Critical incident stress debriefing (CISD) is a structured, one-session group intervention in which survivors’ experiences and emotional reactions are discussed and education and follow-up recommendations are provided. Developed by Mitchell in 1983,18 CISD was widely used until systematic evaluations revealed that it did not alleviate psychological distress or prevent PTSD.19

Table 4

How to interact with news media during and after a disaster

For organizations
Identify a spokesperson with media experience beforehand
Ensure that the spokesperson is well-informed about all aspects of the disaster
For spokespersons
Always
  • Reply immediately to news media requests; find out the reporter’s deadline and refer the caller if you cannot meet it
  • Clearly identify yourself and the organization you represent
  • Understand what the reporter needs—what’s “behind the story”
  • Use simple, declarative sentences; avoid medical jargon
  • Emphasize one or two points
  • Be accurate and honest but use a positive, hopeful frame
  • On TV, look at the interviewer, not the camera
Never
  • Speak off the record
  • Discuss individual cases
  • Speculate on diagnosis or treatment for someone you have not examined (such as a terrorist leader)

Thus, although survivor meetings may provide information, education, screening, and support, avoid detailed discussions of events and emotions. Any meetings should be conducted by mental health clinicians and should not be mandatory. Reserve the term “debriefing” for operational reviews by rescue personnel.20

Public education. Educate survivors, rescue workers, health care providers, teachers, and relief agency workers. Provide concise, simple messages as suggested in Table 3. News media provide our most effective means of reaching out to survivors, which is why having a pre-existing relationship is so important. Some guidelines for working with the media are presented in Table 4.

Outreach. Numerous educational resources are available for survivors and their caregivers (see Related resources). Other potentially useful outreach tools include:

  • meetings with teachers’ organizations
  • continuing medical education activities for primary care providers
  • telephone hot lines.

Legal and ethical issues

Disaster scenes are chaotic and informal, and professionals must be flexible, often providing general support and information rather than specific clinical interventions. However, it is important in each encounter to decide whether a patient-physician relationship has begun.

As a general rule, a physician-patient relationship is established whenever diagnosis or treatment is discussed. Once that happens, briefly document:

  • signs and symptoms
  • working diagnosis
  • suicide or violence potential
  • treatment and/or follow-up plans.

Confidentiality may be difficult to preserve in chaotic situations involving workers from many agencies. Even in disasters, however, you must obtain permission before sharing information unless the individual’s situation is emergent.

Table 5

 

 

Keys to effective disaster psychiatry

Be prepared
  • Contact your local American Psychiatric Association district branch or American Red Cross chapter to learn about existing mental health disaster plans
  • Remember the three basics of psychic resiliency: safety, support, and efficacy
  • Know your place, ask for help, and use existing resources
Care for survivors
  • Your presence alone may be healing
  • Assess thoroughly; symptoms may be related to medical issues or pre-existing conditions, not just stress response
  • Do no harm; avoid psychological debriefing interventions and unnecessary medications
  • Follow up with survivors who exhibit risk factors or symptoms persisting >2 months
  • Consider trauma-focused CBT for survivors with acute stress disorder
Care for yourself
  • As a survivor, tend to your own safety and self-care needs
  • Consult frequently with colleagues within and outside the disaster area for support, information, and guidance

Long-term interventions

Longer-term disaster interventions include continued outreach and education and needed follow-up services. Existing structures may provide effective follow-up, but additional resources are often needed.

Federal programs. Following a presidentialdeclared disaster, the Federal Emergency Management Agency (FEMA) provides funding for crisis counseling. Programs are typically funded for 9 to 15 months and administered through the emergency services and disaster relief branch of the Substance Abuse and Mental Health Services Administration (SAMHSA) and community mental health organizations. Examples include Project Heartland following the 1995 Oklahoma City federal building bombing and Project COPE following California’s 1989 Loma Prieta earthquake.

Cognitive-behavioral therapy. For adult survivors with acute stress disorder, specific cognitive-behavioral therapy (CBT) provided by trained therapists may prevent PTSD and other trauma sequelae, such as depression.21 CBT interventions may begin as early as 2 weeks after trauma and focus sequentially on anxiety management, cognitive restructuring, imaginal exposure followed by in vivo exposure, and relapse prevention.

Three controlled trials found 6-month PTSD rates of 14% to 20% among acute stress disorder patients treated with CBT, compared with 58% to 67% with supportive counseling.22-24 Although studies of interventions immediately following trauma are lacking, trauma-focused CBT is also recommended for children.25 Evidence-based treatments for PTSD are discussed in detail elsewhere.26

Not unprepared after all

With some reflection, Dr. Z realized she had the tools to help her community. Her feelings of helplessness receded as she envisioned how she could help survivors understand their experiences, re-create a sense of safety, restore important connections to loved ones, and begin to rebuild their lives (Table 5).

Related resources

For clinicians

  • Young BH, Ford JD, Ruzek JI, et al. Disaster mental health: a guidebook for clinicians and administrators. Washington, DC: National Center for Post-Traumatic Stress Disorder, 1998. http://ncptsd.org/publications/disaster/index.html
  • Hillman JL. Crisis intervention and trauma: new approaches to evidence-based practice. New York: Kluwer Academic/Plenum Publishers, 2002.
  • Office of the Surgeon General Web site on medical aspects of nuclear, biological, and chemical warfare. http://www.nbc-med.org

For survivors and clinicians

Drug brand names

  • Clonidine • Catapres
  • Guanfacine • Tenex

Disclosure

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

The night started like any other for Dr. Z. Kids in bed (too late) by 10:30, dog out by 11, fell asleep reading journal by 11:15. Sirens jolt her out of a solid stage 4. Her eyes widen, pulse quickens, mouth dries as she follows the glow of the TV into the living room. On TV, fire frames shots of tearful faces, body bags, and firefighters in protective suits. She catches the announcer’s voice: “…explosion at City Power and Light nuclear facility at 3:10 AM today. Fifty-five are known dead, and thousands are being told to evacuate. The blast’s cause is unknown, but terrorism is suspected.”

As her numbness slowly ebbs, Dr. Z’s questions rise insistently. How can I help the survivors? My patients? My children? What if the media call me? How could I have been better prepared for this?

Disaster shakes us to our human and biological core. More than any other clinical encounter, it reminds us that psychiatrists share the vulnerabilities of those we seek to help. Yet it also reminds us that even simple concepts and interventions can mobilize the healing process.

Are you ready to provide emergency psychiatric care following a disaster in your community—be it a nuclear accident, tornado, airplane crash, or terrorist act? Here is evidence—some counterintuitive—that can help you prepare.

Table 1

‘Psychological first aid’ for disaster survivors

Re-create sense of safety
  • Provide for basic needs (food, clothing, medical care)
  • Ensure that survivors are safe and protected from reminders of the event
  • Protect them from onlookers and the media
  • Help them establish a “personal space” and preserve privacy and modesty
Encourage social support
  • Help survivors connect with family and friends (most urgently, children with parents)
  • Educate family and friends about survivors’ normal reactions and how they can help
Re-establish sense of efficacy
  • Give survivors accurate, simple information about plans and events
  • Allow survivors to discuss events and feelings, but do not probe
  • Encourage them to re-establish normal routines and roles when possible
  • Help resolve practical problems, such as getting transportation or relief vouchers
  • Discuss self-care and strategies to reduce anxiety, such as grounding and relaxation techniques
  • Encourage survivors to support and assist others

Disaster’s psychobiology

Human reactions to disaster are often conceptualized as the mammalian survival response: flight, fight, and fright (freezing). In most cases, these reactions are adaptive and dissipate after safety is restored. Posttraumatic stress disorder (PTSD) develops in about 5% of natural disaster victims and 33% of mass shooting victims.1

Responses that do go awry appear to be associated with abnormally low cortisol and persistent adrenergic activation, leading to sensitization of the fear response.2 Reminders of trauma or persistent stressors—such as pain, problems with finances or housing, or bereavement—may exacerbate sensitization. On the other hand, preclinical studies suggest that social support3,4 and active coping5 mitigate physiologic stress responses, confirming numerous clinical observations that associate lack of social support and avoidant coping with eventual PTSD development.

Three basics. Just as our emergency medicine colleagues must often revert to life-support basics, we must remind ourselves of biology’s three basics for psychic resilience:

  • safety (including—as much as possible—protection from reminders of trauma and ongoing stress)
  • meaningful social connection
  • re-establishing a sense of efficacy to overcome helplessness (Table 1).

Like the stress response, these protective factors seem hard-wired into our biological make-up. They form the foundation for all phases of disaster psychiatry interventions, from planning to immediate interventions through longterm follow-up.

Disaster planning

As a mental health professional, plan to operate within established disaster plans and agencies, not only for the sake of efficiency but also because structure and support are paramount in disaster situations. Check with the American Psychiatric Association’s local branch to determine if a disaster mental health plan exists. If not, explore how to work directly with local American Red Cross chapters and hospitals, which recruit personnel for the Disaster Medical Assistance Teams mobilized by the public health service.

Table 2

Normal reactions to disaster for adults and children

All ages
EmotionalShock, fear, grief, anger, guilt, shame, helplessness, hopelessness, numbness, emptiness Decreased ability to feel interest, pleasure, love
CognitiveConfusion, disorientation, indecisiveness, worry, shortened attention span, poor concentration, memory difficulties, unwanted memories, self-blame
PhysicalTension, fatigue, edginess, insomnia, generalized aches and pains, startling easily, rapid heartbeat, nausea, decreased appetite and sex drive
InterpersonalDifficulties being intimate, being over-controlling, feeling rejected or abandoned
Children’s age-specific disaster responses
PreschoolSeparation fears, regression, fussiness, temper tantrums, somatization Sleep disturbances including nightmares, somnambulism, night terrors
School-ageMay still have the above, as well as excessive guilt and worries about others’ safety, poor concentration and loss of school performance, repetitious re-telling or play related to trauma
AdolescentDepression, acting out, wish for revenge, sleeping and eating disturbances, altered view of the future
 

 

Immediate disaster mental health plans vary in detail according to local needs and resources but should at least address:

  • providing on-site interventions
  • disseminating information about responses to trauma
  • identifying and publicizing local mental health resources.

Immediate interventions

Immediately following a disaster, psychiatrists are frequently asked to assist with on-site crisis and medical interventions, evaluate survivors with unusual or intense reactions, and provide public education about psychological reactions to disaster.

On-site response. All responders, regardless of discipline, should provide disaster survivors with “psychological first aid,” which is directed at reestablishing safety, connection, and efficacy. Basic crisis intervention principles are useful when support and reassurance are not enough.6

For example, relaxation exercises can reduce anxiety and improve sleep. Use focused, structured relaxation tools—such as progressive muscle relaxation and breathing training—as unstructured exercises can exacerbate dissociation and re-experiencing. Grounding techniques, by which survivors learn to focus all senses on immediate surroundings, often alleviate dissociation and flashbacks.

Care for children. Because children’s reactions to disaster greatly depend on their caregivers’ responses (social referencing), focus acute interventions for children on:

  • re-connecting them with their families
  • reducing caregivers’ distress
  • educating caregivers about providing age appropriate information and support (see Related resources).

Medical care. As physicians, psychiatrists may be called upon to intervene medically. Although it is generally advisable to stay within our usual practice, medical personnel may be in short supply. Fortunately, Good Samaritan laws exist in every state, and the potential for a successful malpractice suit against a physician responding in a disaster is almost zero, unless the physician’s performance is grossly negligent (such as moving the neck of a patient with obvious head or neck injuries).7

Principles regarding informed consent and right to refuse treatment—along with the usual exceptions—apply during disasters.

Evaluating survivors in shelters and hospitals requires knowing the normal and abnormal responses to disaster, being able to differentially diagnose changes in mental status, and understanding risk factors for trauma’s psychiatric sequelae. Aside from PTSD, trauma’s long-term effects include other anxiety disorders, depression, substance abuse, and eating disorders. In addition to the usual components of a psychiatric evaluation, assessments must address event-related factors such as proximity to the disaster, loss of significant others or property, physical injuries, immediate needs, and social support.8

Normal stress reactions. Frequently described as “a normal response to an abnormal situation,” the normal stress reaction is multidimensional and depends on the person’s developmental level (Table 2). About 10% to 25% of survivors experience intense affect and dissociation, whereas a similar number may appear unusually calm.

Interventions beyond the“first aid” described above are not usually needed unless individuals:

  • are a danger to themselves or others
  • are psychotic
  • have no social supports
  • cannot perform tasks necessary for self-care and to begin the recovery process.

Always re-assess when there is any question about a survivor’s immediate reaction to trauma.

In DSM-IV-TR’s trauma-related diagnoses, the symptom clusters often do not capture many disaster survivors’ subjective experience: the shattering of fundamental beliefs regarding themselves (invulnerability), the world (predictability, safety), and others (trust, benevolence).9 By empathizing with these responses, you can help survivors feel less isolated and estranged.

Differential diagnosis. Survivors’ mental status changes may be manifestations of the stress response, but they also may represent:

  • exacerbations of pre-existing psychiatric or general medical conditions
  • hypoxemia, hypovolemia, or CNS trauma from physical injury
  • responses to medications used for resuscitation or pain control, such as atropine, epinephrine, lidocaine, or morphine.

Effects of bioterrorism agents must also be considered. For example, organophosphorus compounds such as the nerve agents sarin and soman can cause impaired concentration, depression, and anxiety. Anthrax can cause rapidly progressing meningitis.10 Delirium must be differentiated from dissociation; patients with dissociation can be re-oriented, and changes will resolve with time rather than fluctuate.11

Psychopathology risk factors. Multiple studies have addressed risk factors for post-disaster psychiatric sequelae (usually PTSD). In general, risk increases with repeated trauma exposure (including TV viewing), prior trauma, lack of social support, injury, pre-existing psychiatric problems, traumatic bereavement (having witnessed the violent death of a loved one), avoidant coping, and having strong negative beliefs about the meanings of normal stress reactions such as tearfulness, anxiety, and insomnia.

Because a recent meta-analysis supports these observations,12 follow-up evaluation for signs of PTSD is recommended for:

  • survivors with one or more of the risk factors discussed above
  • vulnerable groups such as rescue workers, children, and dependent individuals
  • survivors whose symptoms persist after 2 months.13

Decompensation immediately after a disaster seems to be the exception for psychiatric patients, despite their longer-term vulnerability. One psychiatrist who in 1989 survived Hurricane Hugo—the most intense storm to strike the Mid-Atlantic region in 100 years—noted that demand for inpatient psychiatric services did not increase in the storm’s aftermath. The only patient calls she received were inquiries about her own physical safety.14

 

 

Caregivers and rescue workers—including psychi-atrists—are also disaster survivors, and you need to tend to your needs for safety and support. Consult frequently with colleagues within and outside the disaster area, as much for support as for information and guidance.15 Remember also that rescue workers are occasionally targets for victims’ rage at their circumstances. Anticipating and explaining this displacement reduces its toxicity.

Using medications

Uses psychotropics judiciously in the first 48 hours of trauma. Medication effects may interfere with neurologic assessment of the injured, and monitoring and follow-up may not be possible.

However, drug therapy should start quickly when survivors are acutely psychotic or their behavior endangers themselves, others, or the milieu. Medications usually include a fast-acting benzodiazepine and/or an antipsychotic, as described in guidelines for managing agitation.16 Always provide structure and supervision for medicated patients.

No guidelines exist for using medications to manage distressing—but less-severe—acute stress-related symptoms. Some experts advocate using adrenergic antagonists such as clonidine, guanfacine, and beta blockers to reduce excessive arousal. These drugs have not been adequately studied in this setting, however, and may harm those with cardiovascular instability from preexisting conditions or injuries.

Table 3

Psychoeducation: Simple messages for workers and trauma survivors

Get adequate rest, food, sleep
Avoid exposure to trauma cues, including TV images
Seek support from loved ones and peers
Talk about events and feelings only if this feels comfortable and helpful
Return to normal routine as much as possible
Take action to rebuild, but at a reasonable pace
Reach out to others who may need assistance
Get help:
  • immediately for abnormal reactions (psychosis, suicidality, risky behavior including substance abuse)
  • if normal reactions (insomnia, anxiety, mild dissociation) persist beyond 2 months
  • if at high risk for persistent reaction (bereaved, injured, prior trauma or psychiatric disorder, no social support)

Short-term (<1 week) benzodiazepine use for panic symptoms and severe insomnia is acceptable, but longer-term use may increase PTSD risk.17 A selective serotonin reuptake inhibitor may help individuals with pre-existing PTSD or depression, if you can arrange follow-up.

In the aftermath

‘Debriefing.’ Critical incident stress debriefing (CISD) is a structured, one-session group intervention in which survivors’ experiences and emotional reactions are discussed and education and follow-up recommendations are provided. Developed by Mitchell in 1983,18 CISD was widely used until systematic evaluations revealed that it did not alleviate psychological distress or prevent PTSD.19

Table 4

How to interact with news media during and after a disaster

For organizations
Identify a spokesperson with media experience beforehand
Ensure that the spokesperson is well-informed about all aspects of the disaster
For spokespersons
Always
  • Reply immediately to news media requests; find out the reporter’s deadline and refer the caller if you cannot meet it
  • Clearly identify yourself and the organization you represent
  • Understand what the reporter needs—what’s “behind the story”
  • Use simple, declarative sentences; avoid medical jargon
  • Emphasize one or two points
  • Be accurate and honest but use a positive, hopeful frame
  • On TV, look at the interviewer, not the camera
Never
  • Speak off the record
  • Discuss individual cases
  • Speculate on diagnosis or treatment for someone you have not examined (such as a terrorist leader)

Thus, although survivor meetings may provide information, education, screening, and support, avoid detailed discussions of events and emotions. Any meetings should be conducted by mental health clinicians and should not be mandatory. Reserve the term “debriefing” for operational reviews by rescue personnel.20

Public education. Educate survivors, rescue workers, health care providers, teachers, and relief agency workers. Provide concise, simple messages as suggested in Table 3. News media provide our most effective means of reaching out to survivors, which is why having a pre-existing relationship is so important. Some guidelines for working with the media are presented in Table 4.

Outreach. Numerous educational resources are available for survivors and their caregivers (see Related resources). Other potentially useful outreach tools include:

  • meetings with teachers’ organizations
  • continuing medical education activities for primary care providers
  • telephone hot lines.

Legal and ethical issues

Disaster scenes are chaotic and informal, and professionals must be flexible, often providing general support and information rather than specific clinical interventions. However, it is important in each encounter to decide whether a patient-physician relationship has begun.

As a general rule, a physician-patient relationship is established whenever diagnosis or treatment is discussed. Once that happens, briefly document:

  • signs and symptoms
  • working diagnosis
  • suicide or violence potential
  • treatment and/or follow-up plans.

Confidentiality may be difficult to preserve in chaotic situations involving workers from many agencies. Even in disasters, however, you must obtain permission before sharing information unless the individual’s situation is emergent.

Table 5

 

 

Keys to effective disaster psychiatry

Be prepared
  • Contact your local American Psychiatric Association district branch or American Red Cross chapter to learn about existing mental health disaster plans
  • Remember the three basics of psychic resiliency: safety, support, and efficacy
  • Know your place, ask for help, and use existing resources
Care for survivors
  • Your presence alone may be healing
  • Assess thoroughly; symptoms may be related to medical issues or pre-existing conditions, not just stress response
  • Do no harm; avoid psychological debriefing interventions and unnecessary medications
  • Follow up with survivors who exhibit risk factors or symptoms persisting >2 months
  • Consider trauma-focused CBT for survivors with acute stress disorder
Care for yourself
  • As a survivor, tend to your own safety and self-care needs
  • Consult frequently with colleagues within and outside the disaster area for support, information, and guidance

Long-term interventions

Longer-term disaster interventions include continued outreach and education and needed follow-up services. Existing structures may provide effective follow-up, but additional resources are often needed.

Federal programs. Following a presidentialdeclared disaster, the Federal Emergency Management Agency (FEMA) provides funding for crisis counseling. Programs are typically funded for 9 to 15 months and administered through the emergency services and disaster relief branch of the Substance Abuse and Mental Health Services Administration (SAMHSA) and community mental health organizations. Examples include Project Heartland following the 1995 Oklahoma City federal building bombing and Project COPE following California’s 1989 Loma Prieta earthquake.

Cognitive-behavioral therapy. For adult survivors with acute stress disorder, specific cognitive-behavioral therapy (CBT) provided by trained therapists may prevent PTSD and other trauma sequelae, such as depression.21 CBT interventions may begin as early as 2 weeks after trauma and focus sequentially on anxiety management, cognitive restructuring, imaginal exposure followed by in vivo exposure, and relapse prevention.

Three controlled trials found 6-month PTSD rates of 14% to 20% among acute stress disorder patients treated with CBT, compared with 58% to 67% with supportive counseling.22-24 Although studies of interventions immediately following trauma are lacking, trauma-focused CBT is also recommended for children.25 Evidence-based treatments for PTSD are discussed in detail elsewhere.26

Not unprepared after all

With some reflection, Dr. Z realized she had the tools to help her community. Her feelings of helplessness receded as she envisioned how she could help survivors understand their experiences, re-create a sense of safety, restore important connections to loved ones, and begin to rebuild their lives (Table 5).

Related resources

For clinicians

  • Young BH, Ford JD, Ruzek JI, et al. Disaster mental health: a guidebook for clinicians and administrators. Washington, DC: National Center for Post-Traumatic Stress Disorder, 1998. http://ncptsd.org/publications/disaster/index.html
  • Hillman JL. Crisis intervention and trauma: new approaches to evidence-based practice. New York: Kluwer Academic/Plenum Publishers, 2002.
  • Office of the Surgeon General Web site on medical aspects of nuclear, biological, and chemical warfare. http://www.nbc-med.org

For survivors and clinicians

Drug brand names

  • Clonidine • Catapres
  • Guanfacine • Tenex

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. Effects of traumatic stress in a disaster situation. Washington, DC: National Center for Post-Traumatic Stress Disorder. Department of Veterans Affairs, September 12, 2001 (Accessed March 31, 2004 at http://www.ncptsd.org/facts/disasters/fs_effects_disaster.html).

2. Yehuda R, McFarlane AC, Shalev AY. Predicting the development of post-traumatic stress disorder from the acute response to a traumatic event. Biol Psychiatry 1998;44:1305-13.

3. Levine S, Lyons DM, Schatzberg AF. Psychobiological consequences of social relationships. Ann NY Acad Sci 1997;807:210-18.

4. Uchino BN, Garvey TS. The availability of social support reduces cardiovascular reactivity to acute psychological stress. J Behav Med 1997;20:15-27.

5. LeDoux JE, Gorman JM. A call to action: overcoming anxiety through active coping. Am J Psychiatry 2001;158:1953-5.

6. Hillman JL. Crisis intervention and trauma: new approaches to evidence-based practice. New York: Kluwer Academic/Plenum Publishers, 2002.

7. Daniels G. Good Samaritan acts. Emerg Med Clin North Am 1999;17:491-504.

8. Mental-health intervention for disasters. Washington, DC: National Center for Post-Traumatic Stress Disorder, Department of Veterans Affairs, Sept. 12, 2001. (Accessed March 31, 2004 at http://www.ncptsd.org/facts/disasters/fs_treatment_disaster.html.)

9. Difede J, Apfeldorf WJ, Cloitre M, et al. Acute psychiatric responses to the explosion at the World Trade Center: a case series. J Nerv Ment Dis 1997;185:519-22.

10. DiGiovanni C. Domestic terrorism with chemical or biological agents: psychiatric aspects. Am J Psychiatry 1999;156:1500-5.

11. Rundle JR. Psychiatric issues in medical-surgical disaster casualties: consultation-liaison approach. Psychiatr Q 2000;71:245-58.

12. Brewin CR, Andrews B, Valentine JD. Meta-analysis of risk factors for post-traumatic stress disorder in trauma-exposed adults. J Consult Clin Psychol 2000;68:748-66.

13. Mental health and mass violence: evidence-based early psychological intervention for victims/survivors of mass violence. Washington, DC: National Institute of Mental Health, Nov. 1, 2002 (Accessed Oct. 29, 2003 at http://www.nimh.nih.gov/research/massviolence.pdf.)

14. Austin LS. Organizing a disaster response program in one’s home community. In: Austin LS (ed). Responding to disaster: a guide for mental health professionals. Washington, DC: American Psychiatric Publishing, 1992;53-68.

15. Rousseau AW. Notes from Oklahoma City’s recovery. In: Hall RCW, Norwood AE (eds). Disaster psychiatry handbook. Washington, DC: American Psychiatric Association Committee on Psychiatric Dimensions of Disaster, undated:3-11. (Accessed Oct. 29, 2003 at http://www.psych.org/disasterpsych/pdfs/apadisasterhandbk.pdf.)

16. Yildiz A, Sachs GS, Turgay A. Pharmacological management of agitation in emergency settings. Emerg Med J 2003;20:339-46.

17. Gelpin E, Bonne O, Peri T, et al. Treatment of recent trauma survivors with benzodiazepines: a prospective study. J Clin Psychiatry 1996;57:390-4.

18. Mitchell JT. When disaster strikes. J Em Med Serv 1983;8:36-9.

19. Rose S, Bisson J, Wessely S. Psychological debriefing for preventing posttraumatic stress disorder (PTSD). Cochrane Database Syst Rev 2002;(2):CD000560.-

20. Black J. Forming the libidinal cocoon: the Dallas airplane crashes, the Guadalupe river drownings, and Hurricane Hugo in the Virgin Islands. In: Austin LS (ed). Responding to disaster: a guide for mental health professionals. Washington DC: American Psychiatric Publishing, 1992;169:84.-

21. Ehlers A, Clark DM. Early psychological interventions for adult survivors of trauma: a review. Biol Psychiatry 2003;53:817-26.

22. Bryant RA, Moulds M, Guthrie R, Nixon RD. Treating acute stress disorder following mild traumatic brain injury. Am J Psychiatry 2003;160:585-7.

23. Bryant RA, Sackville T, Dang ST, et al. Treating acute stress disorder: an evaluation of cognitive behavior therapy and supportive counseling techniques. Am J Psychiatry 1999;156:1780-6.

24. Bryant RA, Harvey AG, Dang ST, et al. Treatment of acute stress disorder: a comparison of cognitive-behavioral therapy and supportive counseling. J Consult Clin Psychol 1998;66:862-6.

25. Cohen JA. Treating acute posttraumatic reactions in children and adolescents. Biol Psychiatry 2003;53:827-33.

26. Foa EB, Keane TM, Friedman MJ. Effective treatments for PTSD. New York: Guilford Press, 2000.

References

1. Effects of traumatic stress in a disaster situation. Washington, DC: National Center for Post-Traumatic Stress Disorder. Department of Veterans Affairs, September 12, 2001 (Accessed March 31, 2004 at http://www.ncptsd.org/facts/disasters/fs_effects_disaster.html).

2. Yehuda R, McFarlane AC, Shalev AY. Predicting the development of post-traumatic stress disorder from the acute response to a traumatic event. Biol Psychiatry 1998;44:1305-13.

3. Levine S, Lyons DM, Schatzberg AF. Psychobiological consequences of social relationships. Ann NY Acad Sci 1997;807:210-18.

4. Uchino BN, Garvey TS. The availability of social support reduces cardiovascular reactivity to acute psychological stress. J Behav Med 1997;20:15-27.

5. LeDoux JE, Gorman JM. A call to action: overcoming anxiety through active coping. Am J Psychiatry 2001;158:1953-5.

6. Hillman JL. Crisis intervention and trauma: new approaches to evidence-based practice. New York: Kluwer Academic/Plenum Publishers, 2002.

7. Daniels G. Good Samaritan acts. Emerg Med Clin North Am 1999;17:491-504.

8. Mental-health intervention for disasters. Washington, DC: National Center for Post-Traumatic Stress Disorder, Department of Veterans Affairs, Sept. 12, 2001. (Accessed March 31, 2004 at http://www.ncptsd.org/facts/disasters/fs_treatment_disaster.html.)

9. Difede J, Apfeldorf WJ, Cloitre M, et al. Acute psychiatric responses to the explosion at the World Trade Center: a case series. J Nerv Ment Dis 1997;185:519-22.

10. DiGiovanni C. Domestic terrorism with chemical or biological agents: psychiatric aspects. Am J Psychiatry 1999;156:1500-5.

11. Rundle JR. Psychiatric issues in medical-surgical disaster casualties: consultation-liaison approach. Psychiatr Q 2000;71:245-58.

12. Brewin CR, Andrews B, Valentine JD. Meta-analysis of risk factors for post-traumatic stress disorder in trauma-exposed adults. J Consult Clin Psychol 2000;68:748-66.

13. Mental health and mass violence: evidence-based early psychological intervention for victims/survivors of mass violence. Washington, DC: National Institute of Mental Health, Nov. 1, 2002 (Accessed Oct. 29, 2003 at http://www.nimh.nih.gov/research/massviolence.pdf.)

14. Austin LS. Organizing a disaster response program in one’s home community. In: Austin LS (ed). Responding to disaster: a guide for mental health professionals. Washington, DC: American Psychiatric Publishing, 1992;53-68.

15. Rousseau AW. Notes from Oklahoma City’s recovery. In: Hall RCW, Norwood AE (eds). Disaster psychiatry handbook. Washington, DC: American Psychiatric Association Committee on Psychiatric Dimensions of Disaster, undated:3-11. (Accessed Oct. 29, 2003 at http://www.psych.org/disasterpsych/pdfs/apadisasterhandbk.pdf.)

16. Yildiz A, Sachs GS, Turgay A. Pharmacological management of agitation in emergency settings. Emerg Med J 2003;20:339-46.

17. Gelpin E, Bonne O, Peri T, et al. Treatment of recent trauma survivors with benzodiazepines: a prospective study. J Clin Psychiatry 1996;57:390-4.

18. Mitchell JT. When disaster strikes. J Em Med Serv 1983;8:36-9.

19. Rose S, Bisson J, Wessely S. Psychological debriefing for preventing posttraumatic stress disorder (PTSD). Cochrane Database Syst Rev 2002;(2):CD000560.-

20. Black J. Forming the libidinal cocoon: the Dallas airplane crashes, the Guadalupe river drownings, and Hurricane Hugo in the Virgin Islands. In: Austin LS (ed). Responding to disaster: a guide for mental health professionals. Washington DC: American Psychiatric Publishing, 1992;169:84.-

21. Ehlers A, Clark DM. Early psychological interventions for adult survivors of trauma: a review. Biol Psychiatry 2003;53:817-26.

22. Bryant RA, Moulds M, Guthrie R, Nixon RD. Treating acute stress disorder following mild traumatic brain injury. Am J Psychiatry 2003;160:585-7.

23. Bryant RA, Sackville T, Dang ST, et al. Treating acute stress disorder: an evaluation of cognitive behavior therapy and supportive counseling techniques. Am J Psychiatry 1999;156:1780-6.

24. Bryant RA, Harvey AG, Dang ST, et al. Treatment of acute stress disorder: a comparison of cognitive-behavioral therapy and supportive counseling. J Consult Clin Psychol 1998;66:862-6.

25. Cohen JA. Treating acute posttraumatic reactions in children and adolescents. Biol Psychiatry 2003;53:827-33.

26. Foa EB, Keane TM, Friedman MJ. Effective treatments for PTSD. New York: Guilford Press, 2000.

Issue
Current Psychiatry - 03(05)
Issue
Current Psychiatry - 03(05)
Page Number
12-23
Page Number
12-23
Publications
Publications
Topics
Article Type
Display Headline
Psychological first aid: Emergency care for terrorism and disaster survivors
Display Headline
Psychological first aid: Emergency care for terrorism and disaster survivors
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Trichotillomania: A heads-up on severe cases

Article Type
Changed
Tue, 12/11/2018 - 15:31
Display Headline
Trichotillomania: A heads-up on severe cases

History: Bald at age 9

Ms. D, age 41, began compulsively pulling out and eating her hair at age 8. When she didn’t get her way at home or was nervous about school, she would sit for hours, pulling and eating a strand or two at a time, ultimately ingesting a clump of hair.

By age 9, Ms. D was bald. In grade school, she often wore hats and scarves to class to avoid teasing from other children about her baldness. In high school, she kept to herself and frequently wore wigs.

Ms. D stops pulling for brief periods and her hair grows out, but she invariably resumes pulling when psychosocial stressors mount. Many of life’s normal anxieties—job interviews, work-related stress, social rejection—trigger episodes.

When she is bald, Ms. D pulls and eats hair off her wig. Over the years, she has spent thousands of dollars on custom-made wigs that mask her baldness while feeding her habit.

Ms. D’s episodes are increasingly interfering with her life. She has been steadily employed as an office assistant, but does not socialize with coworkers. She has not dated in years, and during an exacerbation leaves home only to go to work. She also pulls her eyelashes and eyebrows and picks her nails and cuticles.

Ms. D first presented in 1994 after seeing a television segment I did on trichotillomania. At intake, she was wearing a wig and exhibited anxious mood. She also has Crohn’s disease; a gastroenterologist monitors her closely.

Ms. D reports compulsive counting and checking but denies other similar behaviors. No immediate family members have exhibited obsessive-compulsive or hair-pulling behaviors. Her father abused alcohol and a sister has a stuttering problem, although Ms. D denies that these have affected her psychologically.

Ms. D’s hair-pulling behavior suggests:

  • a pica disorder
  • an impulse control disorder
  • or an obsessive-compulsive disorder?

Box

DSM-IV-TR diagnostic criteria for trichotillomania

  1. Recurrent pulling out of one’s own hair, resulting in noticeable hair loss.
  2. Increasing tension immediately before pulling out the hair or when attempting to resist the behavior.
  3. Pleasure, gratification, or relief while pulling out the hair.
  4. The disturbance is not better explained as another mental disorder and is not caused by a dermatological or other general medical condition.
  5. The disturbance causes clinically significant distress or impairment in social, occupational, or other important areas of functioning.

Source: Reprinted with permission from the Diagnostic and statistical manual of mental disorders, 4th ed., text revision. Copyright 2000. American Psychiatric Association.

Dr. Lundt’s observations

Trichotillomania, defined as compulsive pulling of hair, usually begins in childhood or adolescence. Scalp hair is most commonly pulled, but some patients also pull their eyelashes, pubic hair, and other body hair. Some, especially children, have reportedly pulled their pets’ hair.

Mansueto et al estimate that trichotillomania afflicts approximately 1.5% of males and 3.5% of females.1 These estimates, however, do not include persons with the disorder who are too embarrassed to seek treatment.

DSM-IV-TR classifies trichotillomania as an impulse control disorder (Box). Although comorbid anxiety and depressive disorders are common, Ms. D did not meet criteria for any other psychiatric disorder.

Trichotillomania often is episodic. Months or years of abstinence is common after periods of exacerbation, usually caused by stress (Figure).

Many clinicians mistakenly consider trichotillomania a benign disorder with few consequences beyond alopecia.2 Some patients, however, progress into trichophagia—ingestion of pulled hair. Trichophagia is a form of pica disorder, typically defined as persistent eating of non-nutritive substances. Patients often harbor tremendous shame over their hair-eating behavior and resist psychiatric or medical treatment.2

The undigested hair can form sometimes massive clumps called trichobezoars, which are most common among children and the developmentally disabled.4 Persons with trichophagia face a 37.5% risk of forming a trichobezoar.5 The mass can cause abdominal pain, nausea, vomiting, and weight loss; complications include GI obstruction, ulceration, perforation, and peritonitis.6 An untreated trichobezoar can be fatal,7 although such deaths are rare among patients being treated for trichotillomania.

Patients with trichotillomania often respond to medications used to treat obsessive-compulsive disorder, such as clomipramine. Some clinicians believe this agent is more effective than selective serotonin reuptake inhibitors (SSRIs) but more difficult to tolerate. For Ms. D, I started with both.

Treatment: ‘I Don’t need medication’

Initial treatments—including fluoxetine, 20 mg/d for 6 months; hypnotherapy; and clomipramine, 25 mg/d—were unsuccessful. Ms. D was only marginally compliant, believing that she did not need medication.

I referred Ms. D to an out-of-state residential behavioral program specializing in trichotillomania, but she refused to go even as her hair-pulling intensified. Clomipramine was gradually increased to 75 mg nightly, briefly decreasing her pulling, then to 100 mg nightly when symptoms re-emerged. Clomipramine blood levels were monitored with each dosage change to guard against CNS and cardiac toxicity and other side effects (GI complaints, dizziness, cardiac arrhythmias, somnolence).

 

 

After watching for seasonal patterns, I found that Ms. D’s hair-pulling worsened during the winter, although no seasonal change in mood was detected. Phototherapy produced initial success, but Ms. D continued to pull her eyelashes.

Eight weeks later, however, Ms. D’s symptoms escalated. Clomipramine was increased to 150 mg nightly, resulting in blood levels of 99 ng/mL for clomipramine and 204 ng/mL for the metabolite N-desmethylclomipramine—both within the therapeutic range. As she continued pulling, I added buspirone, 10 mg/d.

At this point, would you:

  • continue clomipramine and increase the dosage?
  • discontinue clomipramine and start another psychotropic?
  • or maintain clomipramine at the same dosage and add another psychotropic?

Dr. Lundt’s observations

Drug treatment of trichotillomania has not been studied extensively or long-term, and no consensus exists. Psychoanalysis, cognitive-behavioral therapy (CBT), and hypnotherapy are usually administered with psychotropics.8 Patients often respond to treatment at first, then reach a plateau and resume pulling their hair.9

Numerous psychotropics are used off-label to treat trichotillomania. Several agents have been shown in clinical trials and case reports to reduce hair pulling/eating behaviors (Table), but these findings are limited by small sample size, lack of control groups, and lack of a standard symptom rating scale.

Figure Trichotillomania: Excessive grooming, habit, or vicious circle?



Most clinicians begin with SSRIs because they are generally well tolerated, even at high dosages. Monotherapy often is not adequate for trichotillomania, however. Medication augmentation is common, although little empiric data support this practice.

When clomipramine did not work initally, I explored serotonergic combination strategies.

Table

Medications shown to benefit patients with trichotillomania

DrugEvidenceDosage range (mg/d)Potential side effects
First-line
Tricyclic
 ClomipramineSwedo et al10 50 to 300Sedation, weight gain, cardiac arrythmias
SSRIs
 Fluoxetinevan Minnen et al11 20 to 60GI symptoms, insomnia
 FluvoxamineGabriel12 50 to 250Sexual dysfunction, weight gain
 ParoxetineRavindran et al13 20 to 60Sexual dysfunction, weight gain
 SertralineBradford and Gratzer14 50 to 200Sexual dysfunction, GI symptoms, insomnia
Second-line
Antipsychotics
 HaloperidolVan Ameringen et al15 0.25 to 2Sedation, EPS
 OlanzapineGupta and Gupta16 2.5 to 10Sedation, weight gain
 PimozideStein and Hollander17 25 to 200Restlessness, EPS
 QuetiapineKhouzam et al8 25 to 200Sedation
 RisperidoneGabriel12 Senturk and Tanriverdi18 0.5 to 4.0Sedation, hyperprolactinemia
Mood stabilizer
 LithiumChristenson et al19 900 to 1,500Increased thirst, weight gain, tremor
EPS: extrapyramidal symptoms

Further treatment: Relapse, resection

Ms. D was lost to follow-up for 1 year. She returned in 1996, just after undergoing a laparotomy for removal of a trichobezoar large enough to fill two 2-inch-by-6-inch bags. She also had been treated for pneumonia and a pulmonary embolus.

Riddled with shame and embarrassment, Ms. D had stopped pulling for 10 months, during which time she was off medication. Her pulling behaviors re-emerged, however, and clomipramine was restarted and titrated to 250 mg nightly.

One year later, a second trichobezoar was resected. Her clomipramine/N-desmethylclomipramine level reached 1,535 ng/mL, although an ECG reading was normal. Subsequent clomipramine/N-desmethylclomipramine blood levels were within the therapeutic range. Fluvoxamine, 25 mg/d titrated across 6 weeks to 150 mg/d, was added.

Again, Ms. D stopped taking her medications and was lost to follow-up. Her gastroenterologist began managing her care and started sertraline, dosage unknown, to address her depressed mood. A third trichobezoar was removed.

When Ms. D returned to my practice, I resumed CBT and increased sertraline over 1 month from 100 to 300 mg/d. Adding olanzapine, 2.5 mg/d, diminished her anxiety and markedly decreased her hair pulling.

Months later, her hair-pulling/eating behaviors again intensified, resulting in a small-bowel obstruction and a fourth trichobezoar removal. Olanzapine was increased to 5 mg nightly without significant benefit and with sedating effects.

Clomipramine, 125 mg/d, was reintroduced and her symptoms improved dramatically. On a regimen of sertraline, clomipramine and olanzapine, Ms. D remained stable for 2 years.

Last year, however, a fifth trichobezoar measuring 20 x 15 cm was removed. Subsequent trials of methylphenidate, titrated to 72 mg every morning, and tramadol, titrated to 100 mg/d, were unsuccessful.

After 10 years of medication and psychotherapy with three different providers, Ms. D’s hair-pulling/eating behaviors persist. She is taking ziprasidone, 120 mg bid, and naltrexone, 100 mg bid, to help her impulse control, as well as sertraline, 300 mg/d, and clomipramine, 125 mg/d. Another trichobezoar removal—her sixth in 8 years—is scheduled.

What strategies exist for minimizing Ms. D’s hair-pulling behavior and keeping her in therapy?

Dr. Lundt’s observations

Trichotillomania’s waxing and waning course—and its destructive effects on a patient’s self-esteem—pose a clinical challenge. The disorder’s severity can range from cosmetically annoying to life-threatening, as in Ms. D’s case. Patients embarrassed by their behavior often prematurely leave treatment, desperate to cut off all social contact—including medical appointments.

It is crucial to maintain a nonjudgmental, inviting demeanor to alleviate the patient’s fears and facilitate a return to treatment. Support groups, especially online, can help decrease patients’ isolation and provide a reliable information network (see Related resources).

 

 

I have had excellent results with other trichotillomania patients—especially children and adolescents. Simply naming their condition and demystifying the problem can be therapeutic. Many patients have responded to SSRIs combined with CBT.

Not long ago, trichotillomania patients were met with ignorance and disbelief within the medical community as the disorder was poorly understood. We need to break this cycle of shame and continue investigating treatment strategies.

Related resources

  • StopPulling.com, an interactive behavioral program for persons with trichotillomania. www.stoppulling.com.
  • Penzel F. The hair pulling problem: a complete guide to trichotillomania. New York: Oxford University Press, 2003.
  • Keuthen NJ, Stein DJ, Christenson GA. Help for hair pullers: understanding and coping with trichotillomania. Oakland, CA: New Harbinger Publications, 2001.

Drug brand names

  • Amitriptyline • Elavil
  • Buspirone • BuSpar
  • Clomipramine • Anafranil
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Haloperidol • Haldol
  • Lithium • Eskalith, others
  • Methylphenidate • Concerta, Ritalin
  • Naltrexone • ReVia
  • Olanzapine • Zyprexa
  • Paroxetine • Paxil
  • Pimozide • Orap
  • Quetiapine • Seroquel
  • Risperidone • Risperdal
  • Sertraline • Zoloft
  • Tramadol • Ultram
  • Ziprasidone • Geodon

Disclosure

Dr. Lundt receives research grants from and/or is a speaker for Eli Lilly and Co., Pfizer Inc., GlaxoSmithKline, and Bristol-Myers Squibb Co.

References

1. Mansueto C, Ninan PT, Rothbaum B. Trichotillomania and its treatment in adults: a guide for clinicians. Available at: (http://www.trich.org/articles/view_default.asp?filename="0305_Cases" aid=22&yd= researchers_intro). Accessed April 13, 2004

2. Bouwer C, Stein DJ. Trichobezoars in trichotillomania. Psychosom Med 1998;60:658-60.

3. Philippopoulos GS. A case of trichotillomania (hair pulling). Doc Geigy Acta Psychosom 1961;9:304-12.

4. Lee J. Bezoars and foreign bodies in the stomach. Gastrointest Endosc 1996;6:605-19.

5. Christenson GA, Crow SJ. The characterization and treatment of trichotillomania. Clin Psychiatry 1996;57(suppl 8):42-7.

6. Lal MM, Dhall JC. Trichobezoar: A collective analysis of 39 cases from India with a case report. Indian J Pediatr 1975;12:351-3.

7. DeBakey M, Ochsner W. Bezoars and concretions: A comprehensive review of the literature with an analysis of 303 collected cases and a presentation of 8 additional cases. Surgery 1939;4:934-63.

8. Khouzam HR, Battista MA, Byers PE. An overview of trichotillomania and its response to treatment with quetiapine. Psychiatry 2002;65:261-70.

9. Keuthen NJ, Fraim C, Deckersbach T, et al. Longitudinal follow-up of naturalistic treatment outcome in patients with trichotillomania. J Clin Psychiatry 2001;62:101-7.

10. Swedo SE, Leonard HL, Rapoport JL, et al. A double-blind comparison of clomipramine and desipramine in the treatment of trichotillomania (hair pulling). N Engl J Med 1989;8:497-501.

11. van Minnen A, Hoogduin KA, Keijsers GP, et al. Treatment of trichotillomania with behavioral therapy or fluoxetine: a randomized, waiting-list controlled study. Arch Gen Psychiatry 2003;60:517-22.

12. Gabriel A. A case of resistant trichotillomania treated with risperidone-augmented fluvoxamine. Can J Psychiatry 2001;46:285-6.

13. Ravindran AV, Lapierre YD, Anisman H. Obsessive-compulsive spectrum disorders: Effective treatment with paroxetine. Can J Psychiatry 1999;44:805-7.

14. Bradford JM, Gratzer TG. A treatment for impulse control disorders and paraphilia: a case report. Can J Psychiatry 1995;40:4-5.

15. Van Ameringen M, Mancini C, Oakman JM, Farvolden p. The potential role of haloperidol in the treatment of trichotillomania. J Affect Disord 1999;56:219-26.

16. Gupta MA, Gupta AK. Olanzapine is effective in the management of some self-induced dermatoses: Three case reports. Cutis 2000;66:143-6.

17. Stein DJ, Hollander E. Low-dose pimozide augmentation of serotonin reuptake blockers in the treatment of trichotillomania. J Clin Psych 1992;53:123-6.

18. Senturk V, Tanriverdi N. Resistant trichotillomania and risperidone. Psychosomatics 2002;43:429-30

19. Christenson GA, Popkin MK, Mackenzie TB, Realmuto GM. Lithium treatment of chronic hair pulling. J Clin Psychiatry 1991;52:116-20.

Article PDF
Author and Disclosure Information

Leslie P. Lundt, MD
Affiliate faculty member Idaho State University, Pocatello, ID Practicing psychiatrist, Boise, ID

Issue
Current Psychiatry - 03(05)
Publications
Topics
Page Number
89-105
Sections
Author and Disclosure Information

Leslie P. Lundt, MD
Affiliate faculty member Idaho State University, Pocatello, ID Practicing psychiatrist, Boise, ID

Author and Disclosure Information

Leslie P. Lundt, MD
Affiliate faculty member Idaho State University, Pocatello, ID Practicing psychiatrist, Boise, ID

Article PDF
Article PDF

History: Bald at age 9

Ms. D, age 41, began compulsively pulling out and eating her hair at age 8. When she didn’t get her way at home or was nervous about school, she would sit for hours, pulling and eating a strand or two at a time, ultimately ingesting a clump of hair.

By age 9, Ms. D was bald. In grade school, she often wore hats and scarves to class to avoid teasing from other children about her baldness. In high school, she kept to herself and frequently wore wigs.

Ms. D stops pulling for brief periods and her hair grows out, but she invariably resumes pulling when psychosocial stressors mount. Many of life’s normal anxieties—job interviews, work-related stress, social rejection—trigger episodes.

When she is bald, Ms. D pulls and eats hair off her wig. Over the years, she has spent thousands of dollars on custom-made wigs that mask her baldness while feeding her habit.

Ms. D’s episodes are increasingly interfering with her life. She has been steadily employed as an office assistant, but does not socialize with coworkers. She has not dated in years, and during an exacerbation leaves home only to go to work. She also pulls her eyelashes and eyebrows and picks her nails and cuticles.

Ms. D first presented in 1994 after seeing a television segment I did on trichotillomania. At intake, she was wearing a wig and exhibited anxious mood. She also has Crohn’s disease; a gastroenterologist monitors her closely.

Ms. D reports compulsive counting and checking but denies other similar behaviors. No immediate family members have exhibited obsessive-compulsive or hair-pulling behaviors. Her father abused alcohol and a sister has a stuttering problem, although Ms. D denies that these have affected her psychologically.

Ms. D’s hair-pulling behavior suggests:

  • a pica disorder
  • an impulse control disorder
  • or an obsessive-compulsive disorder?

Box

DSM-IV-TR diagnostic criteria for trichotillomania

  1. Recurrent pulling out of one’s own hair, resulting in noticeable hair loss.
  2. Increasing tension immediately before pulling out the hair or when attempting to resist the behavior.
  3. Pleasure, gratification, or relief while pulling out the hair.
  4. The disturbance is not better explained as another mental disorder and is not caused by a dermatological or other general medical condition.
  5. The disturbance causes clinically significant distress or impairment in social, occupational, or other important areas of functioning.

Source: Reprinted with permission from the Diagnostic and statistical manual of mental disorders, 4th ed., text revision. Copyright 2000. American Psychiatric Association.

Dr. Lundt’s observations

Trichotillomania, defined as compulsive pulling of hair, usually begins in childhood or adolescence. Scalp hair is most commonly pulled, but some patients also pull their eyelashes, pubic hair, and other body hair. Some, especially children, have reportedly pulled their pets’ hair.

Mansueto et al estimate that trichotillomania afflicts approximately 1.5% of males and 3.5% of females.1 These estimates, however, do not include persons with the disorder who are too embarrassed to seek treatment.

DSM-IV-TR classifies trichotillomania as an impulse control disorder (Box). Although comorbid anxiety and depressive disorders are common, Ms. D did not meet criteria for any other psychiatric disorder.

Trichotillomania often is episodic. Months or years of abstinence is common after periods of exacerbation, usually caused by stress (Figure).

Many clinicians mistakenly consider trichotillomania a benign disorder with few consequences beyond alopecia.2 Some patients, however, progress into trichophagia—ingestion of pulled hair. Trichophagia is a form of pica disorder, typically defined as persistent eating of non-nutritive substances. Patients often harbor tremendous shame over their hair-eating behavior and resist psychiatric or medical treatment.2

The undigested hair can form sometimes massive clumps called trichobezoars, which are most common among children and the developmentally disabled.4 Persons with trichophagia face a 37.5% risk of forming a trichobezoar.5 The mass can cause abdominal pain, nausea, vomiting, and weight loss; complications include GI obstruction, ulceration, perforation, and peritonitis.6 An untreated trichobezoar can be fatal,7 although such deaths are rare among patients being treated for trichotillomania.

Patients with trichotillomania often respond to medications used to treat obsessive-compulsive disorder, such as clomipramine. Some clinicians believe this agent is more effective than selective serotonin reuptake inhibitors (SSRIs) but more difficult to tolerate. For Ms. D, I started with both.

Treatment: ‘I Don’t need medication’

Initial treatments—including fluoxetine, 20 mg/d for 6 months; hypnotherapy; and clomipramine, 25 mg/d—were unsuccessful. Ms. D was only marginally compliant, believing that she did not need medication.

I referred Ms. D to an out-of-state residential behavioral program specializing in trichotillomania, but she refused to go even as her hair-pulling intensified. Clomipramine was gradually increased to 75 mg nightly, briefly decreasing her pulling, then to 100 mg nightly when symptoms re-emerged. Clomipramine blood levels were monitored with each dosage change to guard against CNS and cardiac toxicity and other side effects (GI complaints, dizziness, cardiac arrhythmias, somnolence).

 

 

After watching for seasonal patterns, I found that Ms. D’s hair-pulling worsened during the winter, although no seasonal change in mood was detected. Phototherapy produced initial success, but Ms. D continued to pull her eyelashes.

Eight weeks later, however, Ms. D’s symptoms escalated. Clomipramine was increased to 150 mg nightly, resulting in blood levels of 99 ng/mL for clomipramine and 204 ng/mL for the metabolite N-desmethylclomipramine—both within the therapeutic range. As she continued pulling, I added buspirone, 10 mg/d.

At this point, would you:

  • continue clomipramine and increase the dosage?
  • discontinue clomipramine and start another psychotropic?
  • or maintain clomipramine at the same dosage and add another psychotropic?

Dr. Lundt’s observations

Drug treatment of trichotillomania has not been studied extensively or long-term, and no consensus exists. Psychoanalysis, cognitive-behavioral therapy (CBT), and hypnotherapy are usually administered with psychotropics.8 Patients often respond to treatment at first, then reach a plateau and resume pulling their hair.9

Numerous psychotropics are used off-label to treat trichotillomania. Several agents have been shown in clinical trials and case reports to reduce hair pulling/eating behaviors (Table), but these findings are limited by small sample size, lack of control groups, and lack of a standard symptom rating scale.

Figure Trichotillomania: Excessive grooming, habit, or vicious circle?



Most clinicians begin with SSRIs because they are generally well tolerated, even at high dosages. Monotherapy often is not adequate for trichotillomania, however. Medication augmentation is common, although little empiric data support this practice.

When clomipramine did not work initally, I explored serotonergic combination strategies.

Table

Medications shown to benefit patients with trichotillomania

DrugEvidenceDosage range (mg/d)Potential side effects
First-line
Tricyclic
 ClomipramineSwedo et al10 50 to 300Sedation, weight gain, cardiac arrythmias
SSRIs
 Fluoxetinevan Minnen et al11 20 to 60GI symptoms, insomnia
 FluvoxamineGabriel12 50 to 250Sexual dysfunction, weight gain
 ParoxetineRavindran et al13 20 to 60Sexual dysfunction, weight gain
 SertralineBradford and Gratzer14 50 to 200Sexual dysfunction, GI symptoms, insomnia
Second-line
Antipsychotics
 HaloperidolVan Ameringen et al15 0.25 to 2Sedation, EPS
 OlanzapineGupta and Gupta16 2.5 to 10Sedation, weight gain
 PimozideStein and Hollander17 25 to 200Restlessness, EPS
 QuetiapineKhouzam et al8 25 to 200Sedation
 RisperidoneGabriel12 Senturk and Tanriverdi18 0.5 to 4.0Sedation, hyperprolactinemia
Mood stabilizer
 LithiumChristenson et al19 900 to 1,500Increased thirst, weight gain, tremor
EPS: extrapyramidal symptoms

Further treatment: Relapse, resection

Ms. D was lost to follow-up for 1 year. She returned in 1996, just after undergoing a laparotomy for removal of a trichobezoar large enough to fill two 2-inch-by-6-inch bags. She also had been treated for pneumonia and a pulmonary embolus.

Riddled with shame and embarrassment, Ms. D had stopped pulling for 10 months, during which time she was off medication. Her pulling behaviors re-emerged, however, and clomipramine was restarted and titrated to 250 mg nightly.

One year later, a second trichobezoar was resected. Her clomipramine/N-desmethylclomipramine level reached 1,535 ng/mL, although an ECG reading was normal. Subsequent clomipramine/N-desmethylclomipramine blood levels were within the therapeutic range. Fluvoxamine, 25 mg/d titrated across 6 weeks to 150 mg/d, was added.

Again, Ms. D stopped taking her medications and was lost to follow-up. Her gastroenterologist began managing her care and started sertraline, dosage unknown, to address her depressed mood. A third trichobezoar was removed.

When Ms. D returned to my practice, I resumed CBT and increased sertraline over 1 month from 100 to 300 mg/d. Adding olanzapine, 2.5 mg/d, diminished her anxiety and markedly decreased her hair pulling.

Months later, her hair-pulling/eating behaviors again intensified, resulting in a small-bowel obstruction and a fourth trichobezoar removal. Olanzapine was increased to 5 mg nightly without significant benefit and with sedating effects.

Clomipramine, 125 mg/d, was reintroduced and her symptoms improved dramatically. On a regimen of sertraline, clomipramine and olanzapine, Ms. D remained stable for 2 years.

Last year, however, a fifth trichobezoar measuring 20 x 15 cm was removed. Subsequent trials of methylphenidate, titrated to 72 mg every morning, and tramadol, titrated to 100 mg/d, were unsuccessful.

After 10 years of medication and psychotherapy with three different providers, Ms. D’s hair-pulling/eating behaviors persist. She is taking ziprasidone, 120 mg bid, and naltrexone, 100 mg bid, to help her impulse control, as well as sertraline, 300 mg/d, and clomipramine, 125 mg/d. Another trichobezoar removal—her sixth in 8 years—is scheduled.

What strategies exist for minimizing Ms. D’s hair-pulling behavior and keeping her in therapy?

Dr. Lundt’s observations

Trichotillomania’s waxing and waning course—and its destructive effects on a patient’s self-esteem—pose a clinical challenge. The disorder’s severity can range from cosmetically annoying to life-threatening, as in Ms. D’s case. Patients embarrassed by their behavior often prematurely leave treatment, desperate to cut off all social contact—including medical appointments.

It is crucial to maintain a nonjudgmental, inviting demeanor to alleviate the patient’s fears and facilitate a return to treatment. Support groups, especially online, can help decrease patients’ isolation and provide a reliable information network (see Related resources).

 

 

I have had excellent results with other trichotillomania patients—especially children and adolescents. Simply naming their condition and demystifying the problem can be therapeutic. Many patients have responded to SSRIs combined with CBT.

Not long ago, trichotillomania patients were met with ignorance and disbelief within the medical community as the disorder was poorly understood. We need to break this cycle of shame and continue investigating treatment strategies.

Related resources

  • StopPulling.com, an interactive behavioral program for persons with trichotillomania. www.stoppulling.com.
  • Penzel F. The hair pulling problem: a complete guide to trichotillomania. New York: Oxford University Press, 2003.
  • Keuthen NJ, Stein DJ, Christenson GA. Help for hair pullers: understanding and coping with trichotillomania. Oakland, CA: New Harbinger Publications, 2001.

Drug brand names

  • Amitriptyline • Elavil
  • Buspirone • BuSpar
  • Clomipramine • Anafranil
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Haloperidol • Haldol
  • Lithium • Eskalith, others
  • Methylphenidate • Concerta, Ritalin
  • Naltrexone • ReVia
  • Olanzapine • Zyprexa
  • Paroxetine • Paxil
  • Pimozide • Orap
  • Quetiapine • Seroquel
  • Risperidone • Risperdal
  • Sertraline • Zoloft
  • Tramadol • Ultram
  • Ziprasidone • Geodon

Disclosure

Dr. Lundt receives research grants from and/or is a speaker for Eli Lilly and Co., Pfizer Inc., GlaxoSmithKline, and Bristol-Myers Squibb Co.

History: Bald at age 9

Ms. D, age 41, began compulsively pulling out and eating her hair at age 8. When she didn’t get her way at home or was nervous about school, she would sit for hours, pulling and eating a strand or two at a time, ultimately ingesting a clump of hair.

By age 9, Ms. D was bald. In grade school, she often wore hats and scarves to class to avoid teasing from other children about her baldness. In high school, she kept to herself and frequently wore wigs.

Ms. D stops pulling for brief periods and her hair grows out, but she invariably resumes pulling when psychosocial stressors mount. Many of life’s normal anxieties—job interviews, work-related stress, social rejection—trigger episodes.

When she is bald, Ms. D pulls and eats hair off her wig. Over the years, she has spent thousands of dollars on custom-made wigs that mask her baldness while feeding her habit.

Ms. D’s episodes are increasingly interfering with her life. She has been steadily employed as an office assistant, but does not socialize with coworkers. She has not dated in years, and during an exacerbation leaves home only to go to work. She also pulls her eyelashes and eyebrows and picks her nails and cuticles.

Ms. D first presented in 1994 after seeing a television segment I did on trichotillomania. At intake, she was wearing a wig and exhibited anxious mood. She also has Crohn’s disease; a gastroenterologist monitors her closely.

Ms. D reports compulsive counting and checking but denies other similar behaviors. No immediate family members have exhibited obsessive-compulsive or hair-pulling behaviors. Her father abused alcohol and a sister has a stuttering problem, although Ms. D denies that these have affected her psychologically.

Ms. D’s hair-pulling behavior suggests:

  • a pica disorder
  • an impulse control disorder
  • or an obsessive-compulsive disorder?

Box

DSM-IV-TR diagnostic criteria for trichotillomania

  1. Recurrent pulling out of one’s own hair, resulting in noticeable hair loss.
  2. Increasing tension immediately before pulling out the hair or when attempting to resist the behavior.
  3. Pleasure, gratification, or relief while pulling out the hair.
  4. The disturbance is not better explained as another mental disorder and is not caused by a dermatological or other general medical condition.
  5. The disturbance causes clinically significant distress or impairment in social, occupational, or other important areas of functioning.

Source: Reprinted with permission from the Diagnostic and statistical manual of mental disorders, 4th ed., text revision. Copyright 2000. American Psychiatric Association.

Dr. Lundt’s observations

Trichotillomania, defined as compulsive pulling of hair, usually begins in childhood or adolescence. Scalp hair is most commonly pulled, but some patients also pull their eyelashes, pubic hair, and other body hair. Some, especially children, have reportedly pulled their pets’ hair.

Mansueto et al estimate that trichotillomania afflicts approximately 1.5% of males and 3.5% of females.1 These estimates, however, do not include persons with the disorder who are too embarrassed to seek treatment.

DSM-IV-TR classifies trichotillomania as an impulse control disorder (Box). Although comorbid anxiety and depressive disorders are common, Ms. D did not meet criteria for any other psychiatric disorder.

Trichotillomania often is episodic. Months or years of abstinence is common after periods of exacerbation, usually caused by stress (Figure).

Many clinicians mistakenly consider trichotillomania a benign disorder with few consequences beyond alopecia.2 Some patients, however, progress into trichophagia—ingestion of pulled hair. Trichophagia is a form of pica disorder, typically defined as persistent eating of non-nutritive substances. Patients often harbor tremendous shame over their hair-eating behavior and resist psychiatric or medical treatment.2

The undigested hair can form sometimes massive clumps called trichobezoars, which are most common among children and the developmentally disabled.4 Persons with trichophagia face a 37.5% risk of forming a trichobezoar.5 The mass can cause abdominal pain, nausea, vomiting, and weight loss; complications include GI obstruction, ulceration, perforation, and peritonitis.6 An untreated trichobezoar can be fatal,7 although such deaths are rare among patients being treated for trichotillomania.

Patients with trichotillomania often respond to medications used to treat obsessive-compulsive disorder, such as clomipramine. Some clinicians believe this agent is more effective than selective serotonin reuptake inhibitors (SSRIs) but more difficult to tolerate. For Ms. D, I started with both.

Treatment: ‘I Don’t need medication’

Initial treatments—including fluoxetine, 20 mg/d for 6 months; hypnotherapy; and clomipramine, 25 mg/d—were unsuccessful. Ms. D was only marginally compliant, believing that she did not need medication.

I referred Ms. D to an out-of-state residential behavioral program specializing in trichotillomania, but she refused to go even as her hair-pulling intensified. Clomipramine was gradually increased to 75 mg nightly, briefly decreasing her pulling, then to 100 mg nightly when symptoms re-emerged. Clomipramine blood levels were monitored with each dosage change to guard against CNS and cardiac toxicity and other side effects (GI complaints, dizziness, cardiac arrhythmias, somnolence).

 

 

After watching for seasonal patterns, I found that Ms. D’s hair-pulling worsened during the winter, although no seasonal change in mood was detected. Phototherapy produced initial success, but Ms. D continued to pull her eyelashes.

Eight weeks later, however, Ms. D’s symptoms escalated. Clomipramine was increased to 150 mg nightly, resulting in blood levels of 99 ng/mL for clomipramine and 204 ng/mL for the metabolite N-desmethylclomipramine—both within the therapeutic range. As she continued pulling, I added buspirone, 10 mg/d.

At this point, would you:

  • continue clomipramine and increase the dosage?
  • discontinue clomipramine and start another psychotropic?
  • or maintain clomipramine at the same dosage and add another psychotropic?

Dr. Lundt’s observations

Drug treatment of trichotillomania has not been studied extensively or long-term, and no consensus exists. Psychoanalysis, cognitive-behavioral therapy (CBT), and hypnotherapy are usually administered with psychotropics.8 Patients often respond to treatment at first, then reach a plateau and resume pulling their hair.9

Numerous psychotropics are used off-label to treat trichotillomania. Several agents have been shown in clinical trials and case reports to reduce hair pulling/eating behaviors (Table), but these findings are limited by small sample size, lack of control groups, and lack of a standard symptom rating scale.

Figure Trichotillomania: Excessive grooming, habit, or vicious circle?



Most clinicians begin with SSRIs because they are generally well tolerated, even at high dosages. Monotherapy often is not adequate for trichotillomania, however. Medication augmentation is common, although little empiric data support this practice.

When clomipramine did not work initally, I explored serotonergic combination strategies.

Table

Medications shown to benefit patients with trichotillomania

DrugEvidenceDosage range (mg/d)Potential side effects
First-line
Tricyclic
 ClomipramineSwedo et al10 50 to 300Sedation, weight gain, cardiac arrythmias
SSRIs
 Fluoxetinevan Minnen et al11 20 to 60GI symptoms, insomnia
 FluvoxamineGabriel12 50 to 250Sexual dysfunction, weight gain
 ParoxetineRavindran et al13 20 to 60Sexual dysfunction, weight gain
 SertralineBradford and Gratzer14 50 to 200Sexual dysfunction, GI symptoms, insomnia
Second-line
Antipsychotics
 HaloperidolVan Ameringen et al15 0.25 to 2Sedation, EPS
 OlanzapineGupta and Gupta16 2.5 to 10Sedation, weight gain
 PimozideStein and Hollander17 25 to 200Restlessness, EPS
 QuetiapineKhouzam et al8 25 to 200Sedation
 RisperidoneGabriel12 Senturk and Tanriverdi18 0.5 to 4.0Sedation, hyperprolactinemia
Mood stabilizer
 LithiumChristenson et al19 900 to 1,500Increased thirst, weight gain, tremor
EPS: extrapyramidal symptoms

Further treatment: Relapse, resection

Ms. D was lost to follow-up for 1 year. She returned in 1996, just after undergoing a laparotomy for removal of a trichobezoar large enough to fill two 2-inch-by-6-inch bags. She also had been treated for pneumonia and a pulmonary embolus.

Riddled with shame and embarrassment, Ms. D had stopped pulling for 10 months, during which time she was off medication. Her pulling behaviors re-emerged, however, and clomipramine was restarted and titrated to 250 mg nightly.

One year later, a second trichobezoar was resected. Her clomipramine/N-desmethylclomipramine level reached 1,535 ng/mL, although an ECG reading was normal. Subsequent clomipramine/N-desmethylclomipramine blood levels were within the therapeutic range. Fluvoxamine, 25 mg/d titrated across 6 weeks to 150 mg/d, was added.

Again, Ms. D stopped taking her medications and was lost to follow-up. Her gastroenterologist began managing her care and started sertraline, dosage unknown, to address her depressed mood. A third trichobezoar was removed.

When Ms. D returned to my practice, I resumed CBT and increased sertraline over 1 month from 100 to 300 mg/d. Adding olanzapine, 2.5 mg/d, diminished her anxiety and markedly decreased her hair pulling.

Months later, her hair-pulling/eating behaviors again intensified, resulting in a small-bowel obstruction and a fourth trichobezoar removal. Olanzapine was increased to 5 mg nightly without significant benefit and with sedating effects.

Clomipramine, 125 mg/d, was reintroduced and her symptoms improved dramatically. On a regimen of sertraline, clomipramine and olanzapine, Ms. D remained stable for 2 years.

Last year, however, a fifth trichobezoar measuring 20 x 15 cm was removed. Subsequent trials of methylphenidate, titrated to 72 mg every morning, and tramadol, titrated to 100 mg/d, were unsuccessful.

After 10 years of medication and psychotherapy with three different providers, Ms. D’s hair-pulling/eating behaviors persist. She is taking ziprasidone, 120 mg bid, and naltrexone, 100 mg bid, to help her impulse control, as well as sertraline, 300 mg/d, and clomipramine, 125 mg/d. Another trichobezoar removal—her sixth in 8 years—is scheduled.

What strategies exist for minimizing Ms. D’s hair-pulling behavior and keeping her in therapy?

Dr. Lundt’s observations

Trichotillomania’s waxing and waning course—and its destructive effects on a patient’s self-esteem—pose a clinical challenge. The disorder’s severity can range from cosmetically annoying to life-threatening, as in Ms. D’s case. Patients embarrassed by their behavior often prematurely leave treatment, desperate to cut off all social contact—including medical appointments.

It is crucial to maintain a nonjudgmental, inviting demeanor to alleviate the patient’s fears and facilitate a return to treatment. Support groups, especially online, can help decrease patients’ isolation and provide a reliable information network (see Related resources).

 

 

I have had excellent results with other trichotillomania patients—especially children and adolescents. Simply naming their condition and demystifying the problem can be therapeutic. Many patients have responded to SSRIs combined with CBT.

Not long ago, trichotillomania patients were met with ignorance and disbelief within the medical community as the disorder was poorly understood. We need to break this cycle of shame and continue investigating treatment strategies.

Related resources

  • StopPulling.com, an interactive behavioral program for persons with trichotillomania. www.stoppulling.com.
  • Penzel F. The hair pulling problem: a complete guide to trichotillomania. New York: Oxford University Press, 2003.
  • Keuthen NJ, Stein DJ, Christenson GA. Help for hair pullers: understanding and coping with trichotillomania. Oakland, CA: New Harbinger Publications, 2001.

Drug brand names

  • Amitriptyline • Elavil
  • Buspirone • BuSpar
  • Clomipramine • Anafranil
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Haloperidol • Haldol
  • Lithium • Eskalith, others
  • Methylphenidate • Concerta, Ritalin
  • Naltrexone • ReVia
  • Olanzapine • Zyprexa
  • Paroxetine • Paxil
  • Pimozide • Orap
  • Quetiapine • Seroquel
  • Risperidone • Risperdal
  • Sertraline • Zoloft
  • Tramadol • Ultram
  • Ziprasidone • Geodon

Disclosure

Dr. Lundt receives research grants from and/or is a speaker for Eli Lilly and Co., Pfizer Inc., GlaxoSmithKline, and Bristol-Myers Squibb Co.

References

1. Mansueto C, Ninan PT, Rothbaum B. Trichotillomania and its treatment in adults: a guide for clinicians. Available at: (http://www.trich.org/articles/view_default.asp?filename="0305_Cases" aid=22&yd= researchers_intro). Accessed April 13, 2004

2. Bouwer C, Stein DJ. Trichobezoars in trichotillomania. Psychosom Med 1998;60:658-60.

3. Philippopoulos GS. A case of trichotillomania (hair pulling). Doc Geigy Acta Psychosom 1961;9:304-12.

4. Lee J. Bezoars and foreign bodies in the stomach. Gastrointest Endosc 1996;6:605-19.

5. Christenson GA, Crow SJ. The characterization and treatment of trichotillomania. Clin Psychiatry 1996;57(suppl 8):42-7.

6. Lal MM, Dhall JC. Trichobezoar: A collective analysis of 39 cases from India with a case report. Indian J Pediatr 1975;12:351-3.

7. DeBakey M, Ochsner W. Bezoars and concretions: A comprehensive review of the literature with an analysis of 303 collected cases and a presentation of 8 additional cases. Surgery 1939;4:934-63.

8. Khouzam HR, Battista MA, Byers PE. An overview of trichotillomania and its response to treatment with quetiapine. Psychiatry 2002;65:261-70.

9. Keuthen NJ, Fraim C, Deckersbach T, et al. Longitudinal follow-up of naturalistic treatment outcome in patients with trichotillomania. J Clin Psychiatry 2001;62:101-7.

10. Swedo SE, Leonard HL, Rapoport JL, et al. A double-blind comparison of clomipramine and desipramine in the treatment of trichotillomania (hair pulling). N Engl J Med 1989;8:497-501.

11. van Minnen A, Hoogduin KA, Keijsers GP, et al. Treatment of trichotillomania with behavioral therapy or fluoxetine: a randomized, waiting-list controlled study. Arch Gen Psychiatry 2003;60:517-22.

12. Gabriel A. A case of resistant trichotillomania treated with risperidone-augmented fluvoxamine. Can J Psychiatry 2001;46:285-6.

13. Ravindran AV, Lapierre YD, Anisman H. Obsessive-compulsive spectrum disorders: Effective treatment with paroxetine. Can J Psychiatry 1999;44:805-7.

14. Bradford JM, Gratzer TG. A treatment for impulse control disorders and paraphilia: a case report. Can J Psychiatry 1995;40:4-5.

15. Van Ameringen M, Mancini C, Oakman JM, Farvolden p. The potential role of haloperidol in the treatment of trichotillomania. J Affect Disord 1999;56:219-26.

16. Gupta MA, Gupta AK. Olanzapine is effective in the management of some self-induced dermatoses: Three case reports. Cutis 2000;66:143-6.

17. Stein DJ, Hollander E. Low-dose pimozide augmentation of serotonin reuptake blockers in the treatment of trichotillomania. J Clin Psych 1992;53:123-6.

18. Senturk V, Tanriverdi N. Resistant trichotillomania and risperidone. Psychosomatics 2002;43:429-30

19. Christenson GA, Popkin MK, Mackenzie TB, Realmuto GM. Lithium treatment of chronic hair pulling. J Clin Psychiatry 1991;52:116-20.

References

1. Mansueto C, Ninan PT, Rothbaum B. Trichotillomania and its treatment in adults: a guide for clinicians. Available at: (http://www.trich.org/articles/view_default.asp?filename="0305_Cases" aid=22&yd= researchers_intro). Accessed April 13, 2004

2. Bouwer C, Stein DJ. Trichobezoars in trichotillomania. Psychosom Med 1998;60:658-60.

3. Philippopoulos GS. A case of trichotillomania (hair pulling). Doc Geigy Acta Psychosom 1961;9:304-12.

4. Lee J. Bezoars and foreign bodies in the stomach. Gastrointest Endosc 1996;6:605-19.

5. Christenson GA, Crow SJ. The characterization and treatment of trichotillomania. Clin Psychiatry 1996;57(suppl 8):42-7.

6. Lal MM, Dhall JC. Trichobezoar: A collective analysis of 39 cases from India with a case report. Indian J Pediatr 1975;12:351-3.

7. DeBakey M, Ochsner W. Bezoars and concretions: A comprehensive review of the literature with an analysis of 303 collected cases and a presentation of 8 additional cases. Surgery 1939;4:934-63.

8. Khouzam HR, Battista MA, Byers PE. An overview of trichotillomania and its response to treatment with quetiapine. Psychiatry 2002;65:261-70.

9. Keuthen NJ, Fraim C, Deckersbach T, et al. Longitudinal follow-up of naturalistic treatment outcome in patients with trichotillomania. J Clin Psychiatry 2001;62:101-7.

10. Swedo SE, Leonard HL, Rapoport JL, et al. A double-blind comparison of clomipramine and desipramine in the treatment of trichotillomania (hair pulling). N Engl J Med 1989;8:497-501.

11. van Minnen A, Hoogduin KA, Keijsers GP, et al. Treatment of trichotillomania with behavioral therapy or fluoxetine: a randomized, waiting-list controlled study. Arch Gen Psychiatry 2003;60:517-22.

12. Gabriel A. A case of resistant trichotillomania treated with risperidone-augmented fluvoxamine. Can J Psychiatry 2001;46:285-6.

13. Ravindran AV, Lapierre YD, Anisman H. Obsessive-compulsive spectrum disorders: Effective treatment with paroxetine. Can J Psychiatry 1999;44:805-7.

14. Bradford JM, Gratzer TG. A treatment for impulse control disorders and paraphilia: a case report. Can J Psychiatry 1995;40:4-5.

15. Van Ameringen M, Mancini C, Oakman JM, Farvolden p. The potential role of haloperidol in the treatment of trichotillomania. J Affect Disord 1999;56:219-26.

16. Gupta MA, Gupta AK. Olanzapine is effective in the management of some self-induced dermatoses: Three case reports. Cutis 2000;66:143-6.

17. Stein DJ, Hollander E. Low-dose pimozide augmentation of serotonin reuptake blockers in the treatment of trichotillomania. J Clin Psych 1992;53:123-6.

18. Senturk V, Tanriverdi N. Resistant trichotillomania and risperidone. Psychosomatics 2002;43:429-30

19. Christenson GA, Popkin MK, Mackenzie TB, Realmuto GM. Lithium treatment of chronic hair pulling. J Clin Psychiatry 1991;52:116-20.

Issue
Current Psychiatry - 03(05)
Issue
Current Psychiatry - 03(05)
Page Number
89-105
Page Number
89-105
Publications
Publications
Topics
Article Type
Display Headline
Trichotillomania: A heads-up on severe cases
Display Headline
Trichotillomania: A heads-up on severe cases
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

‘Nielsen’ survey tells us you like Current Psychiatry

Article Type
Changed
Mon, 04/16/2018 - 14:13
Display Headline
‘Nielsen’ survey tells us you like Current Psychiatry

We all know about the Nielsen ratings, which track how many households are watching any given television program. Medical journals have a similar rating system called Focus (produced by Nielsen sister company PERQ/HCI), which tracks how many doctors are reading each journal and how thoroughly they read—cover-to-cover, skim, or not at all.

The reason for the Nielsen and Focus surveys, of course, is advertising revenue. Current Psychiatry—like ABC, CBS, and NBC—is supported almost entirely by advertising, which enables you to receive these TV networks and this medical journal for free.

Twice a year, PERQ/HCI sends Focus surveys to 500 to 600 general psychiatrists, then extrapolates the responses to the universe of 32,797 potential readers. Any of you whom the AMA lists as practicing general psychiatrists—including residents—are in the Focus pool.

Our staff recently received the December 2003 Focus survey tabulations, and I was thrilled. More psychiatrists are reading Current Psychiatry than many other psychiatric journals, including those I assumed were the most widely read. Our readership increased more than 20% in the 6 months since June 2003. And although news tabloids consistently show the highest readership, Current Psychiatry—in only its third year of publication—is being read cover to cover more than any other psychiatric journal.

I interpret these results as validating Current Psychiatry’s founding principle: to publish authoritative information you can use in your practice this week. I welcome your ideas and suggestions ([email protected]).

Article PDF
Author and Disclosure Information

 


James Randolph Hillard, MD
Editor-in-Chief

Issue
Current Psychiatry - 03(05)
Publications
Page Number
11-11
Sections
Author and Disclosure Information

 


James Randolph Hillard, MD
Editor-in-Chief

Author and Disclosure Information

 


James Randolph Hillard, MD
Editor-in-Chief

Article PDF
Article PDF

We all know about the Nielsen ratings, which track how many households are watching any given television program. Medical journals have a similar rating system called Focus (produced by Nielsen sister company PERQ/HCI), which tracks how many doctors are reading each journal and how thoroughly they read—cover-to-cover, skim, or not at all.

The reason for the Nielsen and Focus surveys, of course, is advertising revenue. Current Psychiatry—like ABC, CBS, and NBC—is supported almost entirely by advertising, which enables you to receive these TV networks and this medical journal for free.

Twice a year, PERQ/HCI sends Focus surveys to 500 to 600 general psychiatrists, then extrapolates the responses to the universe of 32,797 potential readers. Any of you whom the AMA lists as practicing general psychiatrists—including residents—are in the Focus pool.

Our staff recently received the December 2003 Focus survey tabulations, and I was thrilled. More psychiatrists are reading Current Psychiatry than many other psychiatric journals, including those I assumed were the most widely read. Our readership increased more than 20% in the 6 months since June 2003. And although news tabloids consistently show the highest readership, Current Psychiatry—in only its third year of publication—is being read cover to cover more than any other psychiatric journal.

I interpret these results as validating Current Psychiatry’s founding principle: to publish authoritative information you can use in your practice this week. I welcome your ideas and suggestions ([email protected]).

We all know about the Nielsen ratings, which track how many households are watching any given television program. Medical journals have a similar rating system called Focus (produced by Nielsen sister company PERQ/HCI), which tracks how many doctors are reading each journal and how thoroughly they read—cover-to-cover, skim, or not at all.

The reason for the Nielsen and Focus surveys, of course, is advertising revenue. Current Psychiatry—like ABC, CBS, and NBC—is supported almost entirely by advertising, which enables you to receive these TV networks and this medical journal for free.

Twice a year, PERQ/HCI sends Focus surveys to 500 to 600 general psychiatrists, then extrapolates the responses to the universe of 32,797 potential readers. Any of you whom the AMA lists as practicing general psychiatrists—including residents—are in the Focus pool.

Our staff recently received the December 2003 Focus survey tabulations, and I was thrilled. More psychiatrists are reading Current Psychiatry than many other psychiatric journals, including those I assumed were the most widely read. Our readership increased more than 20% in the 6 months since June 2003. And although news tabloids consistently show the highest readership, Current Psychiatry—in only its third year of publication—is being read cover to cover more than any other psychiatric journal.

I interpret these results as validating Current Psychiatry’s founding principle: to publish authoritative information you can use in your practice this week. I welcome your ideas and suggestions ([email protected]).

Issue
Current Psychiatry - 03(05)
Issue
Current Psychiatry - 03(05)
Page Number
11-11
Page Number
11-11
Publications
Publications
Article Type
Display Headline
‘Nielsen’ survey tells us you like Current Psychiatry
Display Headline
‘Nielsen’ survey tells us you like Current Psychiatry
Sections
Article PDF Media

Hypnosis: Brief interventions offer key to managing pain and anxiety

Article Type
Changed
Tue, 12/11/2018 - 15:10
Display Headline
Hypnosis: Brief interventions offer key to managing pain and anxiety

Mr. M, a world-class athlete, collapsed suddenly in an alley. He was rushed to a hospital emergency room, where he nearly died of internal bleeding from a grapefruit-sized abdominal lymphoma. He was hospitalized and placed on chemotherapy.

Increasing doses of opiates hardly reduced his pain, and he became extremely anxious. Staff described him as “climbing the walls.” He lay in bed writhing, and his parents feared he was becoming a “drug addict.”

Anxiety about his life-threatening illness was clearly compounding his pain, so his attending physician ordered a psychiatric evaluation. When I interviewed the patient, I felt that hypnosis could help.

Hypnosis—as a state of highly focused attention—can help us treat patients’ anxiety, phobias, pain, posttraumatic stress disorder (PTSD), and dissociative disorders. With training, an experienced psychiatrist can quickly start using hypnosis as an adjunct to other therapies.

This article describes how hypnosis helped Mr. M and a young woman traumatized by a criminal assault. Based on my experience and the literature, I discuss what hypnosis is, what training is required, how to measure hypnotizability, and the value of hypnosis in helping patients control their anxiety, posttraumatic, and dissociative states.

Case continued: ‘Surfing’ in Hawaii

When I met Mr. M in the hospital, I acknowledged his distress and the reasons for it, saying “You don’t really want to be here, do you?”

“How many years of medical training did it take you to figure that out?” he replied.

“Well then,” I said, “let’s go somewhere else. Where would you like to be right now?”

He responded, “I’ve never surfed.”

“Good,” I replied, “let’s go to Hawaii.” In hypnosis, I had him picture himself surfing. He continued to groan, but the pattern changed. “What happened?” I asked. “I fell off the surfboard,” he replied. “OK, get back on, and do it right,” I told him.

He learned to practice self-hypnosis, which markedly reduced his anxiety and pain. Two days later he was off pain medications and joking with the nurses in the hall. The attending physician noted in the patient’s record: “Patient off pain meds. Tumor must be regressing.”

What is hypnosis?

Mr. M’s response, though unusually strong, underscores the fact that hypnosis can rapidly produce analgesia and anxiolysis in the medical setting. Hypnosis—often called “believed-in imagination”—is characterized by an ability to sustain a state of attentive, receptive, intense focal concentration with diminished peripheral awareness. The hypnotized person is awake and alert, not asleep. Hypnosis’ three main components are absorption, dissociation, and suggestibility.

Biological basis. The hypnotic state has no brain “signature” per se, but brain imaging portrays hypnosis as a state of alertness with altered anterior cingulate gyrus activation, which helps to focus attention.1-3 Hypnotized persons can demonstrably alter blood flow in brain regions involved in perceptual processing in response to suggestions of altered perception, whether somatosensory, visual, or olfactory.4,5 Thus, patients report not only reduced pain but changes in how they experience pain with hypnotic analgesia.

The brain’s dopamine neurotransmitter system—especially in the frontal lobes—also may be involved in hypnosis, as highly hypnotizable persons have elevated levels of dopamine metabolites in their cerebrospinal fluid.6

Hypnotic trance. The trance experience is often best explained to patients as similar to being absorbed in a good novel. One loses awareness of one’s surroundings and enters the imagined world. When the novel is finished, the reader requires a moment of reorientation to the surrounding world.

A trance is a state of sustained, attentive-receptive concentration in response to a signal from within or from someone else. The signal activates this shift of awareness and permits more-intensive concentration in a designated direction.

All hypnosis is self-hypnosis. Much of its clinical value is that it can be self-induced throughout the day and whenever symptoms emerge. During the first weeks, patients can be encouraged to practice every 1 or 2 hours.

Applying hypnosis to practice. A well-trained clinician can learn to use hypnosis in classes offered by the two professional hypnosis societies or the American Psychiatric Association (Box 1) Because hypnosis is not something “done to” a patient but rather a capacity to be measured, tapped, and utilized, psychiatrists can integrate hypnosis into clinical practice after some initial training, with ongoing learning and supervision.

Who can be hypnotized?

Not everyone is equally hypnotizable, and hypnotizability is a stable and measurable trait. Approximately one-quarter of adults cannot respond to hypnotic instructions, whereas 10% are extremely hypnotizable.7

Brief, clinically useful tests of hypnotic responsiveness have been developed, such as the Hypnotic Induction Profile (HIP).8 The clinician usually can induce the trance experience and systematically measure the patient’s response within 5 minutes. A HIP score of 5 indicates usable hypnotizability.

 

 

The HIP test includes instructions to produce a sense of lightness in the left arm and hand, with tests of response to this instruction. Response is characterized by dissociation, hand elevation after it is lowered, involuntariness, response to the cutoff signal, and altered sensation.

Turning hypnotic induction into a test of hypnotic capacity transforms the initial encounter by:

  • removing pressure on the clinician to successfully hypnotize the subject
  • reducing patients’ experiences of complying with the clinician’s wishes, rather than exploring and discovering their own hypnotic capacity.

Placing the hypnotic experience in the context of a test also makes it consonant with other medical examinations and procedures.8

Once a patient’s hypnotizability is determined, structured measurement is no longer necessary. The test-retest correlation for hypnotizability scores is 0.7 over 25 years, which is more consistent than IQ testing.7 Subsequent inductions usually can be generated by the patient or signaled by the clinician, and only seconds are required for the shift into trance.

Box 1

Sources of training in hypnosis for psychotherapy

Effective, safe work with hypnosis requires clinical expertise in diagnostic assessment and choosing treatment options. Psychiatrists can learn techniques for inducing, measuring, and using hypnotic responsiveness in introductory and advanced workshops, supplemented by local supervision.

Courses in hypnosis are offered by many medical schools. Postgraduate training is available at annual meetings of the American Psychiatric Association, Society for Clinical and Experimental Hypnosis, and American Society of Clinical Hypnosis. The two hypnosis societies offer intensive workshops for psychiatrists, psychologists, and other health care professionals.

Useful text books also are available:

  • Spiegel H, Spiegel D. Trance and treatment: clinical uses of hypnosis. Washington, DC: American Psychiatric Publishing, 2004.
  • Zarren JI, Eimer BN. Brief cognitive hypnosis: facilitating the change of dysfunctional behavior. New York: Springer Publishing, 2002.
  • Lynn SJ, Kirsch I, Rhue JW. Casebook of clinical hypnosis. Washington, DC: American Psychological Association, 1996.
  • Fromm E, Kahn SP. Self-hypnosis: the Chicago paradigm. New York: Guilford Press, 1990.

Reducing anxiety

Anxiety can be understood as a vaguely defined but immobilizing sense of distress. Lack of clarity about the discomfort’s source enhances the patient’s sense of helplessness and avoidance. One therapeutic challenge is to convert anxiety into fear—to give it a focus so that something can be done about it.

Box 2

‘Imagine yourself floating:’ Hypnotic instruction for treating anxiety

Imagine yourself floating in a bath, a lake, a hot tub, or just floating in space. With each breath out, let a little more tension out of your body. Just enjoy this pleasant sense of floating, and notice how you can use your store of memories and fantasies to help yourself and your body feel better.

“While you imagine yourself floating, in your mind’s eye visualize an imaginary screen: a movie, TV, or computer screen, or, if you wish, a piece of clear blue sky. On that screen project your thoughts, fears, worries, ideas, feelings, or memories, while you maintain the pleasant sense of floating in your body. You establish this clear sense of your body floating here, while you relate to your thoughts and ideas out there.

“Once you have established this screen, divide it in half. Use the left side as your ‘worry screen.’ Picture one thing that causes you anxiety on this screen and learn to manage the feelings of discomfort that accompany it. Now use the right side as your ‘problem-solving’ screen. Brainstorm something you can do about the problem on the left, all the while maintaining a sense of floating in your body.

“You may have to ‘freeze’ what is on the ‘worry screen’ and re-establish the floating several times. This allows you to develop new means of coping with the things that are making you anxious, one at a time.”

Anxiety sets up a negative feedback cycle between psychological preoccupation and somatic discomfort, a “snowball effect” in which subjective anxiety and somatic tension reinforce each other. Hypnosis can help reduce anxiety and induce relaxation,9 and its dissociative component can help separate anxiety’s psychological and somatic components.

Hypnosis is as effective at reducing anxiety as 1 mg of alprazolam, at least in a study of college students.10 Student volunteers with high and low hypnotizability were given alprazolam, 1 mg, and a hypnotic suggestion based on their reactions to the drug. Four days later, when students received hypnosis only and hypnosis plus alprazolam:

  • combination therapy reduced anxiety more effectively than did hypnosis or alprazolam alone, as measured by the Profile of Mood States tension-anxiety scale
  • improvement was comparable with hypnosis or alprazolam alone
  • highly hypnotizable students showed significantly greater relaxation than did those with low hypnotizability in all three treatment groups
  • EEG data showed similar frontal and occipital changes in the alprazolam and hypnotic suggestion groups.
 

 

In randomized trials, simple self-hypnosis training has reduced pain and anxiety during medical procedures, reducing procedure time by an average 17 minutes and resulting in fewer complications.11

A typical hypnotic instruction for managing anxiety is provided in Box 2. This approach teaches patients how to deal with stressors that complicate their anxiety and to control their somatic response. Hypnosis expands patients’ repertoire of responses and enables them to feel less helpless.

Confronting phobias

Phobic symptoms of fear and avoidance or exposure with distress respond especially well to brief hypnosis interventions. Although behavior modification and antidepressants also can treat phobias successfully, one or two hypnosis sessions often can reduce or cure phobic symptoms.

For example, one can help patients with airplane phobia prepare for flight by going into a hypnotic state and learning three concepts:

  • Think of the airplane as an extension of the body, such as a bicycle.
  • Float with the plane.
  • Think about the difference between probability and possibility.

The hypnotic state—with its focused attention and physical relaxation—can amplify this cognitive restructuring technique. Phobic patients can feel more in control of their somatic reactions and, by extrapolation, the flying experience itself. In one study, 52% of patients taught this self-hypnosis exercise remained improved or cured at least 7 years later.12

Treating traumatic reactions

Evidence is growing that trauma elicits dissociation. Thus, hypnosis could help us understand and treat traumatic reactions, including patients with acute and posttraumatic stress disorder (PTSD) and dissociative disorders.

The hypnotic state’s controlled dissociation can be used to model the uncontrolled dissociation represented by posttraumatic phenomena such as flashbacks, numbing, and amnesia.13 This view is supported by evidence that PTSD is associated with high hypnotizability.14,15

Acute stress disorder—as introduced in DSM-IV16—is characterized by prominent dissociative symptoms, with intrusion, avoidance, and hyperarousal. These diagnostic criteria recognize that acute dissociation is a common and predictable reaction to trauma.

Hypnosis involving grief work, exploration of trauma-related transference issues, and emotional expression are effective psychotherapies for persons exposed to trauma. Becoming familiar with hypnotic states can teach patients to recognize, understand, and control their dissociative states.

Evidence suggests that hypnosis’ intense concentration may reverse the dissociative mind fragmentation caused by trauma.17 Traumatic memories may seem less overwhelming and intrusive once patients discover they can:

  • exert greater control over memory access and retrieval
  • work through and assimilate disturbing thoughts.

The controlled experience of hypnotic abreaction (reliving traumatic and other memories with strong emotion) provides boundaries for psychotherapeutic grief work.18,19 Instead of telling patients not to ruminate over a traumatic event, the clinician instructs the patient how to think about the experience.

The inferred message is that the patient can work on other things—such as relationships and daily living problems—after this therapeutic work is done.

Patients are slowly separated from the victim role. The goal is to help them restructure their memories, both cognitively and emotionally. They bear the memories’ impact, yet come to see the information differently.7 Traumatic input becomes more bearable when linked to a cognitively restructured recognition of an adaptive response.,20 For example, patients may acknowledge what they did during a traumatic event that was self-protective or helped others.

PTSD. Hypnosis shares common elements with other cognitive and behavioral treatments for PTSD, including exposure to traumatic memories for cognitive and emotional processing. Few studies have examined using hypnosis to treat PTSD, but evidence suggests it is at least as effective as other cognitive-behavioral treatments.20,21

Patients can be taught to view PTSD’s intrusive memories and bodily symptoms as re-experiencing painful memories. The memories often intrude less frequently after patients find a controlled method—such as self-hypnosis—to access and work them through.22

Box 3

Split-screen revelation: ‘He wants to kill me’

Ms. J hoped hypnosis could help her better visualize the face of an assailant who had attacked her as she returned at dusk from the grocery store. She had fought off his attempt to drag her into her apartment and rape her. The police showed little interest in pursuing him, however, because the sexual assault had not been completed. After the police left, she had a grand mal seizure. She had suffered a basalar skull fracture.

Ms. J was highly hypnotizable and learned the split-screen technique. While visualizing the assault on the left screen, she realized something that had not been clear to her before: “From the look on his face, I can see he wants to kill me. If he gets me into my apartment, he will kill me.”

She focused on this realization and the image of his hatred and threat to her. The therapist asked her to picture on the right screen something she had done to protect herself. She said: “He is surprised that I am fighting so hard. He doesn’t expect me to put up such a fight.”

She emerged from hypnosis understanding that she had been in more danger than she realized. Thus, despite the disappointment of having no clearer idea of what he looked like (it was quite dark when he attacked her), she had a restructured perspective about what had occurred.

Before this session, Ms. J had felt guilty that she had gotten herself so seriously injured. Afterward, she could better tolerate the memory of the attack because it was coupled with cognitive awareness that her actions may have saved her life.

 

 

Self-blame. Many trauma victims would rather feel guilty than helpless. They blame themselves inappropriately for events over which they had no control, rather than accept their helplessness. They misuse hindsight about the trauma to assume the events were predictable and therefore avoidable. They imagine they can replay the events and change the outcome.

Such an approach to trauma can be profoundly demoralizing, leaving victims burdened by needless guilt and shame. Helping them face and bear the feelings associated with traumatic events can free them from efforts to “undo” or take responsibility for the trauma and accept what happened.

Split-screen technique. Using hypnosis with a “split-screen” technique can help patients restructure the memory of trauma. The left screen symbolizes the trauma in condensed form. The right screen helps patients focus on how they tried to master the situation. This grief work allows patients to acknowledge, bear, and put into perspective the humiliation of the experience and their loss of invulnerability, health, or loved ones (Box 3).18

Dissociation. Dissociating during a threatening situation may enable a person to put aside some awareness of the danger and take self-protective action. Persistent dissociation, however, may make it too easy to avoid working through the traumatic experiences later on.22-24

Dissociation makes subsequent exposure to reminders of the trauma more similar to a reexperiencing rather than a controlled remembering of it. This can trigger physiologic stress reactions and lead to or worsen PTSD.25-27

Dissociative disorders can be understood as chronic and severe PTSDs.28 Many individuals with dissociative disorders have histories of sexual and physical abuse.29-31 Clearly, traumatic experiences sensitize survivors to subsequent trauma through conditioned activation of fear circuitry involving the amygdala, hippocampus, and frontal lobes.32

Hypnosis can be especially helpful—both for diagnosis and therapy.33 It can assist the controlled recovery of memories, while allowing some images to remain dissociated from cognition until the patient is ready to deal with them. The patient can turn memories on and off by entering and exiting the hypnotic state and thereby recover and reprocess memories at a tolerable pace.

Related resources

References

1. Spiegel D, Jasiukaitis P. Hypnosis: Brain basis. In: Smith BH (ed). Elsevier’s encyclopedia of neuroscience. The Netherlands: Elsevier Science, 1999.

2. Rainville P, Hofbauer RK, Bushnell MC, et al. Hypnosis modulates activity in brain structures involved in the regulation of consciousness. J Cogn Neurosci 2002;14:887-901.

3. Rainville P, Duncan GH, Price DD, et al. Pain affect encoded in human anterior cingulate but not somatosensory cortex. Science 1997;277:968-71.

4. Kosslyn SM, Thompson WL, Costantini-Ferrando MF, et al. Hypnotic visual illusion alters color processing in the brain. Am J Psychiatry 2000;157:1279-84.

5. Spiegel D. Negative and positive visual hypnotic hallucinations: attending inside and out. Int J Clin Exp Hypn 2003;51:130-46.

6. Spiegel D, King R. Hypnotizability and CSF HVA levels among psychiatric patients. Biol Psychiatry 1992;31:95-8.

7. Piccione C, Hilgard ER, Zimbardo PG. On the degree of stability of measured hypnotizability over a 25-year period. J Pers Soc Psychol 1989;56:289-95.

8. Spiegel H, Spiegel D. Trance and treatment: Clinical uses of hypnosis. Washington, DC: American Psychiatric Press, 2004.

9. Wertz JM, Sayette MA. Effects of smoking opportunity on attentional bias in smokers. Psychol Addict Behav 2001;15:268-71.

10. Nishith P, Barabasz A, Barabasz M, Warner D. Brief hypnosis substitutes for alprazolam use in college students: transient experiences and quantitative EEG responses. Am J Clin Hypn 1999;41:262-8.

11. Lang EV, Benotsch EG, Fick LJ, et al. Adjunctive nonpharmacological analgesia for invasive medical procedures: a randomised trial. Lancet 2000;355:1486-90.

12. Spiegel D, Frischholz EJ, Maruffi B, Spiegel H. Hypnotic responsitivity and the treatment of flying phobia. Am J Clin Hypn 1981;23:239-47.

13. Butler LD, Duran EFD, Jasiukaitis P, et al. Hypnotizability and traumatic experience: a diathesis-stress model of dissociative symptomatology. Am J Psychiatry 1996;153:42-63.

14. Spiegel D. Dissociation and hypnosis in post-traumatic stress disorder. J Trauma Stress 1988;1:17-33.

15. Stutman RK, Bliss EL. Posttraumatic stress disorder, hypnotizability, and imagery. Am J Psychiatry 1985;142:741-3.

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

17. Maldonado JR, Spiegel D. Trauma, dissociation and hypnotizability. In: Marmar R, Bremmer D (eds). Trauma, memory and dissociation. Washington, DC: American Psychiatric Press, 1998.

18. Lindemann E. Symptomatology and management of acute grief. Am J Psychiatry 1994;151:155-60.

19. Spiegel D. Vietnam grief work using hypnosis. Am J Clin Hypn 1981;24:33-40.

20. Foa EB, Davidson JRT, Frances A. Treatment of posttraumatic stress disorder. J Clin Psychiatry 1999;50:4-69.

21. Brom D, Kleber RJ, Defare PB. Brief psychotherapy for post-traumatic stress disorder. J Consult Clin Psychol 1989;57:607-12.

22. Spiegel D. Hypnosis and implicit memory: automatic processing of explicit content. Am J Clin Hypn 1998;40:231-40.

23. Spiegel D. Multiple personality as a post-traumatic stress disorder. Psychiatr Clin North Am 1984;7:101-10.

24. Kluft RP. Dissociation as a response to extreme trauma. In: Kluft RP (ed). Childhood antecedents of multiple personality. Washington, DC: American Psychiatric Press, 1985:66-97.

25. Marmar CR, Weiss DS, Metzler T. Peritraumatic dissociation and posttraumatic stress disorder. In: Bremner JD, Marmar C (eds). Trauma, memory, and dissociation. Washington, DC: American Psychiatric Press, 1998;229-52.

26. Birmes P. Peritraumatic dissociation, acute stress, and early posttraumatic stress disorder in victims of general crime. Can J Psychiatry 2001;46:649-51.

27. Spiegel D. Hypnosis, dissociation and trauma. In: Burrows GD, Stanley RO, Bloom PB (eds). Clinical hypnosis. New York: John Wiley & Sons, 2001;143-58.

28. Spiegel D, Cardena E. Disintegrated experience: the dissociative disorders revisited. J Abnorm Psychol 1991;100:366-78.

29. Chu JA, Dill DL. Dissociative symptoms in relation to childhood physical and sexual abuse. Am J Psychiatry 1990;147:887-92.

30. Kluft RP. Childhood antecedents of multiple personality. Washington, DC: American Psychiatric Press, 1985.

31. Spiegel D. Dissociating damage. Am J Clin Hypn 1986;29:123-31.

32. LeDoux J. Synaptic self: How our brains become who we are. New York: Viking Press, 2002.

33. Putnam FW. Using hypnosis for therapeutic abreactions. Psychiatr Med 1992;10:51-65.

Article PDF
Author and Disclosure Information

David Spiegel, MD
Willson Professor in the School of Medicine Department of Psychiatry and Behavioral Sciences Stanford University School of Medicine Stanford, CA

Issue
Current Psychiatry - 03(04)
Publications
Topics
Page Number
48-59
Sections
Author and Disclosure Information

David Spiegel, MD
Willson Professor in the School of Medicine Department of Psychiatry and Behavioral Sciences Stanford University School of Medicine Stanford, CA

Author and Disclosure Information

David Spiegel, MD
Willson Professor in the School of Medicine Department of Psychiatry and Behavioral Sciences Stanford University School of Medicine Stanford, CA

Article PDF
Article PDF

Mr. M, a world-class athlete, collapsed suddenly in an alley. He was rushed to a hospital emergency room, where he nearly died of internal bleeding from a grapefruit-sized abdominal lymphoma. He was hospitalized and placed on chemotherapy.

Increasing doses of opiates hardly reduced his pain, and he became extremely anxious. Staff described him as “climbing the walls.” He lay in bed writhing, and his parents feared he was becoming a “drug addict.”

Anxiety about his life-threatening illness was clearly compounding his pain, so his attending physician ordered a psychiatric evaluation. When I interviewed the patient, I felt that hypnosis could help.

Hypnosis—as a state of highly focused attention—can help us treat patients’ anxiety, phobias, pain, posttraumatic stress disorder (PTSD), and dissociative disorders. With training, an experienced psychiatrist can quickly start using hypnosis as an adjunct to other therapies.

This article describes how hypnosis helped Mr. M and a young woman traumatized by a criminal assault. Based on my experience and the literature, I discuss what hypnosis is, what training is required, how to measure hypnotizability, and the value of hypnosis in helping patients control their anxiety, posttraumatic, and dissociative states.

Case continued: ‘Surfing’ in Hawaii

When I met Mr. M in the hospital, I acknowledged his distress and the reasons for it, saying “You don’t really want to be here, do you?”

“How many years of medical training did it take you to figure that out?” he replied.

“Well then,” I said, “let’s go somewhere else. Where would you like to be right now?”

He responded, “I’ve never surfed.”

“Good,” I replied, “let’s go to Hawaii.” In hypnosis, I had him picture himself surfing. He continued to groan, but the pattern changed. “What happened?” I asked. “I fell off the surfboard,” he replied. “OK, get back on, and do it right,” I told him.

He learned to practice self-hypnosis, which markedly reduced his anxiety and pain. Two days later he was off pain medications and joking with the nurses in the hall. The attending physician noted in the patient’s record: “Patient off pain meds. Tumor must be regressing.”

What is hypnosis?

Mr. M’s response, though unusually strong, underscores the fact that hypnosis can rapidly produce analgesia and anxiolysis in the medical setting. Hypnosis—often called “believed-in imagination”—is characterized by an ability to sustain a state of attentive, receptive, intense focal concentration with diminished peripheral awareness. The hypnotized person is awake and alert, not asleep. Hypnosis’ three main components are absorption, dissociation, and suggestibility.

Biological basis. The hypnotic state has no brain “signature” per se, but brain imaging portrays hypnosis as a state of alertness with altered anterior cingulate gyrus activation, which helps to focus attention.1-3 Hypnotized persons can demonstrably alter blood flow in brain regions involved in perceptual processing in response to suggestions of altered perception, whether somatosensory, visual, or olfactory.4,5 Thus, patients report not only reduced pain but changes in how they experience pain with hypnotic analgesia.

The brain’s dopamine neurotransmitter system—especially in the frontal lobes—also may be involved in hypnosis, as highly hypnotizable persons have elevated levels of dopamine metabolites in their cerebrospinal fluid.6

Hypnotic trance. The trance experience is often best explained to patients as similar to being absorbed in a good novel. One loses awareness of one’s surroundings and enters the imagined world. When the novel is finished, the reader requires a moment of reorientation to the surrounding world.

A trance is a state of sustained, attentive-receptive concentration in response to a signal from within or from someone else. The signal activates this shift of awareness and permits more-intensive concentration in a designated direction.

All hypnosis is self-hypnosis. Much of its clinical value is that it can be self-induced throughout the day and whenever symptoms emerge. During the first weeks, patients can be encouraged to practice every 1 or 2 hours.

Applying hypnosis to practice. A well-trained clinician can learn to use hypnosis in classes offered by the two professional hypnosis societies or the American Psychiatric Association (Box 1) Because hypnosis is not something “done to” a patient but rather a capacity to be measured, tapped, and utilized, psychiatrists can integrate hypnosis into clinical practice after some initial training, with ongoing learning and supervision.

Who can be hypnotized?

Not everyone is equally hypnotizable, and hypnotizability is a stable and measurable trait. Approximately one-quarter of adults cannot respond to hypnotic instructions, whereas 10% are extremely hypnotizable.7

Brief, clinically useful tests of hypnotic responsiveness have been developed, such as the Hypnotic Induction Profile (HIP).8 The clinician usually can induce the trance experience and systematically measure the patient’s response within 5 minutes. A HIP score of 5 indicates usable hypnotizability.

 

 

The HIP test includes instructions to produce a sense of lightness in the left arm and hand, with tests of response to this instruction. Response is characterized by dissociation, hand elevation after it is lowered, involuntariness, response to the cutoff signal, and altered sensation.

Turning hypnotic induction into a test of hypnotic capacity transforms the initial encounter by:

  • removing pressure on the clinician to successfully hypnotize the subject
  • reducing patients’ experiences of complying with the clinician’s wishes, rather than exploring and discovering their own hypnotic capacity.

Placing the hypnotic experience in the context of a test also makes it consonant with other medical examinations and procedures.8

Once a patient’s hypnotizability is determined, structured measurement is no longer necessary. The test-retest correlation for hypnotizability scores is 0.7 over 25 years, which is more consistent than IQ testing.7 Subsequent inductions usually can be generated by the patient or signaled by the clinician, and only seconds are required for the shift into trance.

Box 1

Sources of training in hypnosis for psychotherapy

Effective, safe work with hypnosis requires clinical expertise in diagnostic assessment and choosing treatment options. Psychiatrists can learn techniques for inducing, measuring, and using hypnotic responsiveness in introductory and advanced workshops, supplemented by local supervision.

Courses in hypnosis are offered by many medical schools. Postgraduate training is available at annual meetings of the American Psychiatric Association, Society for Clinical and Experimental Hypnosis, and American Society of Clinical Hypnosis. The two hypnosis societies offer intensive workshops for psychiatrists, psychologists, and other health care professionals.

Useful text books also are available:

  • Spiegel H, Spiegel D. Trance and treatment: clinical uses of hypnosis. Washington, DC: American Psychiatric Publishing, 2004.
  • Zarren JI, Eimer BN. Brief cognitive hypnosis: facilitating the change of dysfunctional behavior. New York: Springer Publishing, 2002.
  • Lynn SJ, Kirsch I, Rhue JW. Casebook of clinical hypnosis. Washington, DC: American Psychological Association, 1996.
  • Fromm E, Kahn SP. Self-hypnosis: the Chicago paradigm. New York: Guilford Press, 1990.

Reducing anxiety

Anxiety can be understood as a vaguely defined but immobilizing sense of distress. Lack of clarity about the discomfort’s source enhances the patient’s sense of helplessness and avoidance. One therapeutic challenge is to convert anxiety into fear—to give it a focus so that something can be done about it.

Box 2

‘Imagine yourself floating:’ Hypnotic instruction for treating anxiety

Imagine yourself floating in a bath, a lake, a hot tub, or just floating in space. With each breath out, let a little more tension out of your body. Just enjoy this pleasant sense of floating, and notice how you can use your store of memories and fantasies to help yourself and your body feel better.

“While you imagine yourself floating, in your mind’s eye visualize an imaginary screen: a movie, TV, or computer screen, or, if you wish, a piece of clear blue sky. On that screen project your thoughts, fears, worries, ideas, feelings, or memories, while you maintain the pleasant sense of floating in your body. You establish this clear sense of your body floating here, while you relate to your thoughts and ideas out there.

“Once you have established this screen, divide it in half. Use the left side as your ‘worry screen.’ Picture one thing that causes you anxiety on this screen and learn to manage the feelings of discomfort that accompany it. Now use the right side as your ‘problem-solving’ screen. Brainstorm something you can do about the problem on the left, all the while maintaining a sense of floating in your body.

“You may have to ‘freeze’ what is on the ‘worry screen’ and re-establish the floating several times. This allows you to develop new means of coping with the things that are making you anxious, one at a time.”

Anxiety sets up a negative feedback cycle between psychological preoccupation and somatic discomfort, a “snowball effect” in which subjective anxiety and somatic tension reinforce each other. Hypnosis can help reduce anxiety and induce relaxation,9 and its dissociative component can help separate anxiety’s psychological and somatic components.

Hypnosis is as effective at reducing anxiety as 1 mg of alprazolam, at least in a study of college students.10 Student volunteers with high and low hypnotizability were given alprazolam, 1 mg, and a hypnotic suggestion based on their reactions to the drug. Four days later, when students received hypnosis only and hypnosis plus alprazolam:

  • combination therapy reduced anxiety more effectively than did hypnosis or alprazolam alone, as measured by the Profile of Mood States tension-anxiety scale
  • improvement was comparable with hypnosis or alprazolam alone
  • highly hypnotizable students showed significantly greater relaxation than did those with low hypnotizability in all three treatment groups
  • EEG data showed similar frontal and occipital changes in the alprazolam and hypnotic suggestion groups.
 

 

In randomized trials, simple self-hypnosis training has reduced pain and anxiety during medical procedures, reducing procedure time by an average 17 minutes and resulting in fewer complications.11

A typical hypnotic instruction for managing anxiety is provided in Box 2. This approach teaches patients how to deal with stressors that complicate their anxiety and to control their somatic response. Hypnosis expands patients’ repertoire of responses and enables them to feel less helpless.

Confronting phobias

Phobic symptoms of fear and avoidance or exposure with distress respond especially well to brief hypnosis interventions. Although behavior modification and antidepressants also can treat phobias successfully, one or two hypnosis sessions often can reduce or cure phobic symptoms.

For example, one can help patients with airplane phobia prepare for flight by going into a hypnotic state and learning three concepts:

  • Think of the airplane as an extension of the body, such as a bicycle.
  • Float with the plane.
  • Think about the difference between probability and possibility.

The hypnotic state—with its focused attention and physical relaxation—can amplify this cognitive restructuring technique. Phobic patients can feel more in control of their somatic reactions and, by extrapolation, the flying experience itself. In one study, 52% of patients taught this self-hypnosis exercise remained improved or cured at least 7 years later.12

Treating traumatic reactions

Evidence is growing that trauma elicits dissociation. Thus, hypnosis could help us understand and treat traumatic reactions, including patients with acute and posttraumatic stress disorder (PTSD) and dissociative disorders.

The hypnotic state’s controlled dissociation can be used to model the uncontrolled dissociation represented by posttraumatic phenomena such as flashbacks, numbing, and amnesia.13 This view is supported by evidence that PTSD is associated with high hypnotizability.14,15

Acute stress disorder—as introduced in DSM-IV16—is characterized by prominent dissociative symptoms, with intrusion, avoidance, and hyperarousal. These diagnostic criteria recognize that acute dissociation is a common and predictable reaction to trauma.

Hypnosis involving grief work, exploration of trauma-related transference issues, and emotional expression are effective psychotherapies for persons exposed to trauma. Becoming familiar with hypnotic states can teach patients to recognize, understand, and control their dissociative states.

Evidence suggests that hypnosis’ intense concentration may reverse the dissociative mind fragmentation caused by trauma.17 Traumatic memories may seem less overwhelming and intrusive once patients discover they can:

  • exert greater control over memory access and retrieval
  • work through and assimilate disturbing thoughts.

The controlled experience of hypnotic abreaction (reliving traumatic and other memories with strong emotion) provides boundaries for psychotherapeutic grief work.18,19 Instead of telling patients not to ruminate over a traumatic event, the clinician instructs the patient how to think about the experience.

The inferred message is that the patient can work on other things—such as relationships and daily living problems—after this therapeutic work is done.

Patients are slowly separated from the victim role. The goal is to help them restructure their memories, both cognitively and emotionally. They bear the memories’ impact, yet come to see the information differently.7 Traumatic input becomes more bearable when linked to a cognitively restructured recognition of an adaptive response.,20 For example, patients may acknowledge what they did during a traumatic event that was self-protective or helped others.

PTSD. Hypnosis shares common elements with other cognitive and behavioral treatments for PTSD, including exposure to traumatic memories for cognitive and emotional processing. Few studies have examined using hypnosis to treat PTSD, but evidence suggests it is at least as effective as other cognitive-behavioral treatments.20,21

Patients can be taught to view PTSD’s intrusive memories and bodily symptoms as re-experiencing painful memories. The memories often intrude less frequently after patients find a controlled method—such as self-hypnosis—to access and work them through.22

Box 3

Split-screen revelation: ‘He wants to kill me’

Ms. J hoped hypnosis could help her better visualize the face of an assailant who had attacked her as she returned at dusk from the grocery store. She had fought off his attempt to drag her into her apartment and rape her. The police showed little interest in pursuing him, however, because the sexual assault had not been completed. After the police left, she had a grand mal seizure. She had suffered a basalar skull fracture.

Ms. J was highly hypnotizable and learned the split-screen technique. While visualizing the assault on the left screen, she realized something that had not been clear to her before: “From the look on his face, I can see he wants to kill me. If he gets me into my apartment, he will kill me.”

She focused on this realization and the image of his hatred and threat to her. The therapist asked her to picture on the right screen something she had done to protect herself. She said: “He is surprised that I am fighting so hard. He doesn’t expect me to put up such a fight.”

She emerged from hypnosis understanding that she had been in more danger than she realized. Thus, despite the disappointment of having no clearer idea of what he looked like (it was quite dark when he attacked her), she had a restructured perspective about what had occurred.

Before this session, Ms. J had felt guilty that she had gotten herself so seriously injured. Afterward, she could better tolerate the memory of the attack because it was coupled with cognitive awareness that her actions may have saved her life.

 

 

Self-blame. Many trauma victims would rather feel guilty than helpless. They blame themselves inappropriately for events over which they had no control, rather than accept their helplessness. They misuse hindsight about the trauma to assume the events were predictable and therefore avoidable. They imagine they can replay the events and change the outcome.

Such an approach to trauma can be profoundly demoralizing, leaving victims burdened by needless guilt and shame. Helping them face and bear the feelings associated with traumatic events can free them from efforts to “undo” or take responsibility for the trauma and accept what happened.

Split-screen technique. Using hypnosis with a “split-screen” technique can help patients restructure the memory of trauma. The left screen symbolizes the trauma in condensed form. The right screen helps patients focus on how they tried to master the situation. This grief work allows patients to acknowledge, bear, and put into perspective the humiliation of the experience and their loss of invulnerability, health, or loved ones (Box 3).18

Dissociation. Dissociating during a threatening situation may enable a person to put aside some awareness of the danger and take self-protective action. Persistent dissociation, however, may make it too easy to avoid working through the traumatic experiences later on.22-24

Dissociation makes subsequent exposure to reminders of the trauma more similar to a reexperiencing rather than a controlled remembering of it. This can trigger physiologic stress reactions and lead to or worsen PTSD.25-27

Dissociative disorders can be understood as chronic and severe PTSDs.28 Many individuals with dissociative disorders have histories of sexual and physical abuse.29-31 Clearly, traumatic experiences sensitize survivors to subsequent trauma through conditioned activation of fear circuitry involving the amygdala, hippocampus, and frontal lobes.32

Hypnosis can be especially helpful—both for diagnosis and therapy.33 It can assist the controlled recovery of memories, while allowing some images to remain dissociated from cognition until the patient is ready to deal with them. The patient can turn memories on and off by entering and exiting the hypnotic state and thereby recover and reprocess memories at a tolerable pace.

Related resources

Mr. M, a world-class athlete, collapsed suddenly in an alley. He was rushed to a hospital emergency room, where he nearly died of internal bleeding from a grapefruit-sized abdominal lymphoma. He was hospitalized and placed on chemotherapy.

Increasing doses of opiates hardly reduced his pain, and he became extremely anxious. Staff described him as “climbing the walls.” He lay in bed writhing, and his parents feared he was becoming a “drug addict.”

Anxiety about his life-threatening illness was clearly compounding his pain, so his attending physician ordered a psychiatric evaluation. When I interviewed the patient, I felt that hypnosis could help.

Hypnosis—as a state of highly focused attention—can help us treat patients’ anxiety, phobias, pain, posttraumatic stress disorder (PTSD), and dissociative disorders. With training, an experienced psychiatrist can quickly start using hypnosis as an adjunct to other therapies.

This article describes how hypnosis helped Mr. M and a young woman traumatized by a criminal assault. Based on my experience and the literature, I discuss what hypnosis is, what training is required, how to measure hypnotizability, and the value of hypnosis in helping patients control their anxiety, posttraumatic, and dissociative states.

Case continued: ‘Surfing’ in Hawaii

When I met Mr. M in the hospital, I acknowledged his distress and the reasons for it, saying “You don’t really want to be here, do you?”

“How many years of medical training did it take you to figure that out?” he replied.

“Well then,” I said, “let’s go somewhere else. Where would you like to be right now?”

He responded, “I’ve never surfed.”

“Good,” I replied, “let’s go to Hawaii.” In hypnosis, I had him picture himself surfing. He continued to groan, but the pattern changed. “What happened?” I asked. “I fell off the surfboard,” he replied. “OK, get back on, and do it right,” I told him.

He learned to practice self-hypnosis, which markedly reduced his anxiety and pain. Two days later he was off pain medications and joking with the nurses in the hall. The attending physician noted in the patient’s record: “Patient off pain meds. Tumor must be regressing.”

What is hypnosis?

Mr. M’s response, though unusually strong, underscores the fact that hypnosis can rapidly produce analgesia and anxiolysis in the medical setting. Hypnosis—often called “believed-in imagination”—is characterized by an ability to sustain a state of attentive, receptive, intense focal concentration with diminished peripheral awareness. The hypnotized person is awake and alert, not asleep. Hypnosis’ three main components are absorption, dissociation, and suggestibility.

Biological basis. The hypnotic state has no brain “signature” per se, but brain imaging portrays hypnosis as a state of alertness with altered anterior cingulate gyrus activation, which helps to focus attention.1-3 Hypnotized persons can demonstrably alter blood flow in brain regions involved in perceptual processing in response to suggestions of altered perception, whether somatosensory, visual, or olfactory.4,5 Thus, patients report not only reduced pain but changes in how they experience pain with hypnotic analgesia.

The brain’s dopamine neurotransmitter system—especially in the frontal lobes—also may be involved in hypnosis, as highly hypnotizable persons have elevated levels of dopamine metabolites in their cerebrospinal fluid.6

Hypnotic trance. The trance experience is often best explained to patients as similar to being absorbed in a good novel. One loses awareness of one’s surroundings and enters the imagined world. When the novel is finished, the reader requires a moment of reorientation to the surrounding world.

A trance is a state of sustained, attentive-receptive concentration in response to a signal from within or from someone else. The signal activates this shift of awareness and permits more-intensive concentration in a designated direction.

All hypnosis is self-hypnosis. Much of its clinical value is that it can be self-induced throughout the day and whenever symptoms emerge. During the first weeks, patients can be encouraged to practice every 1 or 2 hours.

Applying hypnosis to practice. A well-trained clinician can learn to use hypnosis in classes offered by the two professional hypnosis societies or the American Psychiatric Association (Box 1) Because hypnosis is not something “done to” a patient but rather a capacity to be measured, tapped, and utilized, psychiatrists can integrate hypnosis into clinical practice after some initial training, with ongoing learning and supervision.

Who can be hypnotized?

Not everyone is equally hypnotizable, and hypnotizability is a stable and measurable trait. Approximately one-quarter of adults cannot respond to hypnotic instructions, whereas 10% are extremely hypnotizable.7

Brief, clinically useful tests of hypnotic responsiveness have been developed, such as the Hypnotic Induction Profile (HIP).8 The clinician usually can induce the trance experience and systematically measure the patient’s response within 5 minutes. A HIP score of 5 indicates usable hypnotizability.

 

 

The HIP test includes instructions to produce a sense of lightness in the left arm and hand, with tests of response to this instruction. Response is characterized by dissociation, hand elevation after it is lowered, involuntariness, response to the cutoff signal, and altered sensation.

Turning hypnotic induction into a test of hypnotic capacity transforms the initial encounter by:

  • removing pressure on the clinician to successfully hypnotize the subject
  • reducing patients’ experiences of complying with the clinician’s wishes, rather than exploring and discovering their own hypnotic capacity.

Placing the hypnotic experience in the context of a test also makes it consonant with other medical examinations and procedures.8

Once a patient’s hypnotizability is determined, structured measurement is no longer necessary. The test-retest correlation for hypnotizability scores is 0.7 over 25 years, which is more consistent than IQ testing.7 Subsequent inductions usually can be generated by the patient or signaled by the clinician, and only seconds are required for the shift into trance.

Box 1

Sources of training in hypnosis for psychotherapy

Effective, safe work with hypnosis requires clinical expertise in diagnostic assessment and choosing treatment options. Psychiatrists can learn techniques for inducing, measuring, and using hypnotic responsiveness in introductory and advanced workshops, supplemented by local supervision.

Courses in hypnosis are offered by many medical schools. Postgraduate training is available at annual meetings of the American Psychiatric Association, Society for Clinical and Experimental Hypnosis, and American Society of Clinical Hypnosis. The two hypnosis societies offer intensive workshops for psychiatrists, psychologists, and other health care professionals.

Useful text books also are available:

  • Spiegel H, Spiegel D. Trance and treatment: clinical uses of hypnosis. Washington, DC: American Psychiatric Publishing, 2004.
  • Zarren JI, Eimer BN. Brief cognitive hypnosis: facilitating the change of dysfunctional behavior. New York: Springer Publishing, 2002.
  • Lynn SJ, Kirsch I, Rhue JW. Casebook of clinical hypnosis. Washington, DC: American Psychological Association, 1996.
  • Fromm E, Kahn SP. Self-hypnosis: the Chicago paradigm. New York: Guilford Press, 1990.

Reducing anxiety

Anxiety can be understood as a vaguely defined but immobilizing sense of distress. Lack of clarity about the discomfort’s source enhances the patient’s sense of helplessness and avoidance. One therapeutic challenge is to convert anxiety into fear—to give it a focus so that something can be done about it.

Box 2

‘Imagine yourself floating:’ Hypnotic instruction for treating anxiety

Imagine yourself floating in a bath, a lake, a hot tub, or just floating in space. With each breath out, let a little more tension out of your body. Just enjoy this pleasant sense of floating, and notice how you can use your store of memories and fantasies to help yourself and your body feel better.

“While you imagine yourself floating, in your mind’s eye visualize an imaginary screen: a movie, TV, or computer screen, or, if you wish, a piece of clear blue sky. On that screen project your thoughts, fears, worries, ideas, feelings, or memories, while you maintain the pleasant sense of floating in your body. You establish this clear sense of your body floating here, while you relate to your thoughts and ideas out there.

“Once you have established this screen, divide it in half. Use the left side as your ‘worry screen.’ Picture one thing that causes you anxiety on this screen and learn to manage the feelings of discomfort that accompany it. Now use the right side as your ‘problem-solving’ screen. Brainstorm something you can do about the problem on the left, all the while maintaining a sense of floating in your body.

“You may have to ‘freeze’ what is on the ‘worry screen’ and re-establish the floating several times. This allows you to develop new means of coping with the things that are making you anxious, one at a time.”

Anxiety sets up a negative feedback cycle between psychological preoccupation and somatic discomfort, a “snowball effect” in which subjective anxiety and somatic tension reinforce each other. Hypnosis can help reduce anxiety and induce relaxation,9 and its dissociative component can help separate anxiety’s psychological and somatic components.

Hypnosis is as effective at reducing anxiety as 1 mg of alprazolam, at least in a study of college students.10 Student volunteers with high and low hypnotizability were given alprazolam, 1 mg, and a hypnotic suggestion based on their reactions to the drug. Four days later, when students received hypnosis only and hypnosis plus alprazolam:

  • combination therapy reduced anxiety more effectively than did hypnosis or alprazolam alone, as measured by the Profile of Mood States tension-anxiety scale
  • improvement was comparable with hypnosis or alprazolam alone
  • highly hypnotizable students showed significantly greater relaxation than did those with low hypnotizability in all three treatment groups
  • EEG data showed similar frontal and occipital changes in the alprazolam and hypnotic suggestion groups.
 

 

In randomized trials, simple self-hypnosis training has reduced pain and anxiety during medical procedures, reducing procedure time by an average 17 minutes and resulting in fewer complications.11

A typical hypnotic instruction for managing anxiety is provided in Box 2. This approach teaches patients how to deal with stressors that complicate their anxiety and to control their somatic response. Hypnosis expands patients’ repertoire of responses and enables them to feel less helpless.

Confronting phobias

Phobic symptoms of fear and avoidance or exposure with distress respond especially well to brief hypnosis interventions. Although behavior modification and antidepressants also can treat phobias successfully, one or two hypnosis sessions often can reduce or cure phobic symptoms.

For example, one can help patients with airplane phobia prepare for flight by going into a hypnotic state and learning three concepts:

  • Think of the airplane as an extension of the body, such as a bicycle.
  • Float with the plane.
  • Think about the difference between probability and possibility.

The hypnotic state—with its focused attention and physical relaxation—can amplify this cognitive restructuring technique. Phobic patients can feel more in control of their somatic reactions and, by extrapolation, the flying experience itself. In one study, 52% of patients taught this self-hypnosis exercise remained improved or cured at least 7 years later.12

Treating traumatic reactions

Evidence is growing that trauma elicits dissociation. Thus, hypnosis could help us understand and treat traumatic reactions, including patients with acute and posttraumatic stress disorder (PTSD) and dissociative disorders.

The hypnotic state’s controlled dissociation can be used to model the uncontrolled dissociation represented by posttraumatic phenomena such as flashbacks, numbing, and amnesia.13 This view is supported by evidence that PTSD is associated with high hypnotizability.14,15

Acute stress disorder—as introduced in DSM-IV16—is characterized by prominent dissociative symptoms, with intrusion, avoidance, and hyperarousal. These diagnostic criteria recognize that acute dissociation is a common and predictable reaction to trauma.

Hypnosis involving grief work, exploration of trauma-related transference issues, and emotional expression are effective psychotherapies for persons exposed to trauma. Becoming familiar with hypnotic states can teach patients to recognize, understand, and control their dissociative states.

Evidence suggests that hypnosis’ intense concentration may reverse the dissociative mind fragmentation caused by trauma.17 Traumatic memories may seem less overwhelming and intrusive once patients discover they can:

  • exert greater control over memory access and retrieval
  • work through and assimilate disturbing thoughts.

The controlled experience of hypnotic abreaction (reliving traumatic and other memories with strong emotion) provides boundaries for psychotherapeutic grief work.18,19 Instead of telling patients not to ruminate over a traumatic event, the clinician instructs the patient how to think about the experience.

The inferred message is that the patient can work on other things—such as relationships and daily living problems—after this therapeutic work is done.

Patients are slowly separated from the victim role. The goal is to help them restructure their memories, both cognitively and emotionally. They bear the memories’ impact, yet come to see the information differently.7 Traumatic input becomes more bearable when linked to a cognitively restructured recognition of an adaptive response.,20 For example, patients may acknowledge what they did during a traumatic event that was self-protective or helped others.

PTSD. Hypnosis shares common elements with other cognitive and behavioral treatments for PTSD, including exposure to traumatic memories for cognitive and emotional processing. Few studies have examined using hypnosis to treat PTSD, but evidence suggests it is at least as effective as other cognitive-behavioral treatments.20,21

Patients can be taught to view PTSD’s intrusive memories and bodily symptoms as re-experiencing painful memories. The memories often intrude less frequently after patients find a controlled method—such as self-hypnosis—to access and work them through.22

Box 3

Split-screen revelation: ‘He wants to kill me’

Ms. J hoped hypnosis could help her better visualize the face of an assailant who had attacked her as she returned at dusk from the grocery store. She had fought off his attempt to drag her into her apartment and rape her. The police showed little interest in pursuing him, however, because the sexual assault had not been completed. After the police left, she had a grand mal seizure. She had suffered a basalar skull fracture.

Ms. J was highly hypnotizable and learned the split-screen technique. While visualizing the assault on the left screen, she realized something that had not been clear to her before: “From the look on his face, I can see he wants to kill me. If he gets me into my apartment, he will kill me.”

She focused on this realization and the image of his hatred and threat to her. The therapist asked her to picture on the right screen something she had done to protect herself. She said: “He is surprised that I am fighting so hard. He doesn’t expect me to put up such a fight.”

She emerged from hypnosis understanding that she had been in more danger than she realized. Thus, despite the disappointment of having no clearer idea of what he looked like (it was quite dark when he attacked her), she had a restructured perspective about what had occurred.

Before this session, Ms. J had felt guilty that she had gotten herself so seriously injured. Afterward, she could better tolerate the memory of the attack because it was coupled with cognitive awareness that her actions may have saved her life.

 

 

Self-blame. Many trauma victims would rather feel guilty than helpless. They blame themselves inappropriately for events over which they had no control, rather than accept their helplessness. They misuse hindsight about the trauma to assume the events were predictable and therefore avoidable. They imagine they can replay the events and change the outcome.

Such an approach to trauma can be profoundly demoralizing, leaving victims burdened by needless guilt and shame. Helping them face and bear the feelings associated with traumatic events can free them from efforts to “undo” or take responsibility for the trauma and accept what happened.

Split-screen technique. Using hypnosis with a “split-screen” technique can help patients restructure the memory of trauma. The left screen symbolizes the trauma in condensed form. The right screen helps patients focus on how they tried to master the situation. This grief work allows patients to acknowledge, bear, and put into perspective the humiliation of the experience and their loss of invulnerability, health, or loved ones (Box 3).18

Dissociation. Dissociating during a threatening situation may enable a person to put aside some awareness of the danger and take self-protective action. Persistent dissociation, however, may make it too easy to avoid working through the traumatic experiences later on.22-24

Dissociation makes subsequent exposure to reminders of the trauma more similar to a reexperiencing rather than a controlled remembering of it. This can trigger physiologic stress reactions and lead to or worsen PTSD.25-27

Dissociative disorders can be understood as chronic and severe PTSDs.28 Many individuals with dissociative disorders have histories of sexual and physical abuse.29-31 Clearly, traumatic experiences sensitize survivors to subsequent trauma through conditioned activation of fear circuitry involving the amygdala, hippocampus, and frontal lobes.32

Hypnosis can be especially helpful—both for diagnosis and therapy.33 It can assist the controlled recovery of memories, while allowing some images to remain dissociated from cognition until the patient is ready to deal with them. The patient can turn memories on and off by entering and exiting the hypnotic state and thereby recover and reprocess memories at a tolerable pace.

Related resources

References

1. Spiegel D, Jasiukaitis P. Hypnosis: Brain basis. In: Smith BH (ed). Elsevier’s encyclopedia of neuroscience. The Netherlands: Elsevier Science, 1999.

2. Rainville P, Hofbauer RK, Bushnell MC, et al. Hypnosis modulates activity in brain structures involved in the regulation of consciousness. J Cogn Neurosci 2002;14:887-901.

3. Rainville P, Duncan GH, Price DD, et al. Pain affect encoded in human anterior cingulate but not somatosensory cortex. Science 1997;277:968-71.

4. Kosslyn SM, Thompson WL, Costantini-Ferrando MF, et al. Hypnotic visual illusion alters color processing in the brain. Am J Psychiatry 2000;157:1279-84.

5. Spiegel D. Negative and positive visual hypnotic hallucinations: attending inside and out. Int J Clin Exp Hypn 2003;51:130-46.

6. Spiegel D, King R. Hypnotizability and CSF HVA levels among psychiatric patients. Biol Psychiatry 1992;31:95-8.

7. Piccione C, Hilgard ER, Zimbardo PG. On the degree of stability of measured hypnotizability over a 25-year period. J Pers Soc Psychol 1989;56:289-95.

8. Spiegel H, Spiegel D. Trance and treatment: Clinical uses of hypnosis. Washington, DC: American Psychiatric Press, 2004.

9. Wertz JM, Sayette MA. Effects of smoking opportunity on attentional bias in smokers. Psychol Addict Behav 2001;15:268-71.

10. Nishith P, Barabasz A, Barabasz M, Warner D. Brief hypnosis substitutes for alprazolam use in college students: transient experiences and quantitative EEG responses. Am J Clin Hypn 1999;41:262-8.

11. Lang EV, Benotsch EG, Fick LJ, et al. Adjunctive nonpharmacological analgesia for invasive medical procedures: a randomised trial. Lancet 2000;355:1486-90.

12. Spiegel D, Frischholz EJ, Maruffi B, Spiegel H. Hypnotic responsitivity and the treatment of flying phobia. Am J Clin Hypn 1981;23:239-47.

13. Butler LD, Duran EFD, Jasiukaitis P, et al. Hypnotizability and traumatic experience: a diathesis-stress model of dissociative symptomatology. Am J Psychiatry 1996;153:42-63.

14. Spiegel D. Dissociation and hypnosis in post-traumatic stress disorder. J Trauma Stress 1988;1:17-33.

15. Stutman RK, Bliss EL. Posttraumatic stress disorder, hypnotizability, and imagery. Am J Psychiatry 1985;142:741-3.

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

17. Maldonado JR, Spiegel D. Trauma, dissociation and hypnotizability. In: Marmar R, Bremmer D (eds). Trauma, memory and dissociation. Washington, DC: American Psychiatric Press, 1998.

18. Lindemann E. Symptomatology and management of acute grief. Am J Psychiatry 1994;151:155-60.

19. Spiegel D. Vietnam grief work using hypnosis. Am J Clin Hypn 1981;24:33-40.

20. Foa EB, Davidson JRT, Frances A. Treatment of posttraumatic stress disorder. J Clin Psychiatry 1999;50:4-69.

21. Brom D, Kleber RJ, Defare PB. Brief psychotherapy for post-traumatic stress disorder. J Consult Clin Psychol 1989;57:607-12.

22. Spiegel D. Hypnosis and implicit memory: automatic processing of explicit content. Am J Clin Hypn 1998;40:231-40.

23. Spiegel D. Multiple personality as a post-traumatic stress disorder. Psychiatr Clin North Am 1984;7:101-10.

24. Kluft RP. Dissociation as a response to extreme trauma. In: Kluft RP (ed). Childhood antecedents of multiple personality. Washington, DC: American Psychiatric Press, 1985:66-97.

25. Marmar CR, Weiss DS, Metzler T. Peritraumatic dissociation and posttraumatic stress disorder. In: Bremner JD, Marmar C (eds). Trauma, memory, and dissociation. Washington, DC: American Psychiatric Press, 1998;229-52.

26. Birmes P. Peritraumatic dissociation, acute stress, and early posttraumatic stress disorder in victims of general crime. Can J Psychiatry 2001;46:649-51.

27. Spiegel D. Hypnosis, dissociation and trauma. In: Burrows GD, Stanley RO, Bloom PB (eds). Clinical hypnosis. New York: John Wiley & Sons, 2001;143-58.

28. Spiegel D, Cardena E. Disintegrated experience: the dissociative disorders revisited. J Abnorm Psychol 1991;100:366-78.

29. Chu JA, Dill DL. Dissociative symptoms in relation to childhood physical and sexual abuse. Am J Psychiatry 1990;147:887-92.

30. Kluft RP. Childhood antecedents of multiple personality. Washington, DC: American Psychiatric Press, 1985.

31. Spiegel D. Dissociating damage. Am J Clin Hypn 1986;29:123-31.

32. LeDoux J. Synaptic self: How our brains become who we are. New York: Viking Press, 2002.

33. Putnam FW. Using hypnosis for therapeutic abreactions. Psychiatr Med 1992;10:51-65.

References

1. Spiegel D, Jasiukaitis P. Hypnosis: Brain basis. In: Smith BH (ed). Elsevier’s encyclopedia of neuroscience. The Netherlands: Elsevier Science, 1999.

2. Rainville P, Hofbauer RK, Bushnell MC, et al. Hypnosis modulates activity in brain structures involved in the regulation of consciousness. J Cogn Neurosci 2002;14:887-901.

3. Rainville P, Duncan GH, Price DD, et al. Pain affect encoded in human anterior cingulate but not somatosensory cortex. Science 1997;277:968-71.

4. Kosslyn SM, Thompson WL, Costantini-Ferrando MF, et al. Hypnotic visual illusion alters color processing in the brain. Am J Psychiatry 2000;157:1279-84.

5. Spiegel D. Negative and positive visual hypnotic hallucinations: attending inside and out. Int J Clin Exp Hypn 2003;51:130-46.

6. Spiegel D, King R. Hypnotizability and CSF HVA levels among psychiatric patients. Biol Psychiatry 1992;31:95-8.

7. Piccione C, Hilgard ER, Zimbardo PG. On the degree of stability of measured hypnotizability over a 25-year period. J Pers Soc Psychol 1989;56:289-95.

8. Spiegel H, Spiegel D. Trance and treatment: Clinical uses of hypnosis. Washington, DC: American Psychiatric Press, 2004.

9. Wertz JM, Sayette MA. Effects of smoking opportunity on attentional bias in smokers. Psychol Addict Behav 2001;15:268-71.

10. Nishith P, Barabasz A, Barabasz M, Warner D. Brief hypnosis substitutes for alprazolam use in college students: transient experiences and quantitative EEG responses. Am J Clin Hypn 1999;41:262-8.

11. Lang EV, Benotsch EG, Fick LJ, et al. Adjunctive nonpharmacological analgesia for invasive medical procedures: a randomised trial. Lancet 2000;355:1486-90.

12. Spiegel D, Frischholz EJ, Maruffi B, Spiegel H. Hypnotic responsitivity and the treatment of flying phobia. Am J Clin Hypn 1981;23:239-47.

13. Butler LD, Duran EFD, Jasiukaitis P, et al. Hypnotizability and traumatic experience: a diathesis-stress model of dissociative symptomatology. Am J Psychiatry 1996;153:42-63.

14. Spiegel D. Dissociation and hypnosis in post-traumatic stress disorder. J Trauma Stress 1988;1:17-33.

15. Stutman RK, Bliss EL. Posttraumatic stress disorder, hypnotizability, and imagery. Am J Psychiatry 1985;142:741-3.

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

17. Maldonado JR, Spiegel D. Trauma, dissociation and hypnotizability. In: Marmar R, Bremmer D (eds). Trauma, memory and dissociation. Washington, DC: American Psychiatric Press, 1998.

18. Lindemann E. Symptomatology and management of acute grief. Am J Psychiatry 1994;151:155-60.

19. Spiegel D. Vietnam grief work using hypnosis. Am J Clin Hypn 1981;24:33-40.

20. Foa EB, Davidson JRT, Frances A. Treatment of posttraumatic stress disorder. J Clin Psychiatry 1999;50:4-69.

21. Brom D, Kleber RJ, Defare PB. Brief psychotherapy for post-traumatic stress disorder. J Consult Clin Psychol 1989;57:607-12.

22. Spiegel D. Hypnosis and implicit memory: automatic processing of explicit content. Am J Clin Hypn 1998;40:231-40.

23. Spiegel D. Multiple personality as a post-traumatic stress disorder. Psychiatr Clin North Am 1984;7:101-10.

24. Kluft RP. Dissociation as a response to extreme trauma. In: Kluft RP (ed). Childhood antecedents of multiple personality. Washington, DC: American Psychiatric Press, 1985:66-97.

25. Marmar CR, Weiss DS, Metzler T. Peritraumatic dissociation and posttraumatic stress disorder. In: Bremner JD, Marmar C (eds). Trauma, memory, and dissociation. Washington, DC: American Psychiatric Press, 1998;229-52.

26. Birmes P. Peritraumatic dissociation, acute stress, and early posttraumatic stress disorder in victims of general crime. Can J Psychiatry 2001;46:649-51.

27. Spiegel D. Hypnosis, dissociation and trauma. In: Burrows GD, Stanley RO, Bloom PB (eds). Clinical hypnosis. New York: John Wiley & Sons, 2001;143-58.

28. Spiegel D, Cardena E. Disintegrated experience: the dissociative disorders revisited. J Abnorm Psychol 1991;100:366-78.

29. Chu JA, Dill DL. Dissociative symptoms in relation to childhood physical and sexual abuse. Am J Psychiatry 1990;147:887-92.

30. Kluft RP. Childhood antecedents of multiple personality. Washington, DC: American Psychiatric Press, 1985.

31. Spiegel D. Dissociating damage. Am J Clin Hypn 1986;29:123-31.

32. LeDoux J. Synaptic self: How our brains become who we are. New York: Viking Press, 2002.

33. Putnam FW. Using hypnosis for therapeutic abreactions. Psychiatr Med 1992;10:51-65.

Issue
Current Psychiatry - 03(04)
Issue
Current Psychiatry - 03(04)
Page Number
48-59
Page Number
48-59
Publications
Publications
Topics
Article Type
Display Headline
Hypnosis: Brief interventions offer key to managing pain and anxiety
Display Headline
Hypnosis: Brief interventions offer key to managing pain and anxiety
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Olanzapine/fluoxetine combination: Evidence for using the first treatment indicated for bipolar depression

Article Type
Changed
Tue, 12/11/2018 - 15:37
Display Headline
Olanzapine/fluoxetine combination: Evidence for using the first treatment indicated for bipolar depression

Patients with bipolar disorder spend half their lives significantly symptomatic, mainly in the depressive phase.1 Treating bipolar depression poses a clinical challenge, although new research is starting to uncover some answers. Antidepressant drugs are commonly used, but recent data question the effectiveness of this practice.2

An olanzapine-fluoxetine combination (OFC), FDA-approved for treating bipolar type I depression, has demonstrated efficacy in clinical trials.

How it works

Most atypical antipsychotics—including olanzapine—are potent 5-HT2A (serotonin) receptor antagonists. This effect is similar to that of some antidepressants and may mediate some antidepressant and antianxiety effects of these drugs.3

Like most atypicals, olanzapine is also a potent 5-HT2C blocker. While binding to this receptor, serotonin inhibits dopamine release in the nucleus accumbens and frontal cortex.4 Thus, serotonin blockade would increase dopamine release in these areas. One study showed that olanzapine and fluoxetine together increased dopamine and norepinephrine in the frontal cortex of rats, compared with either drug given individually.5 Dopamine is critical to regulating motivation, defined as the ability to exert energy to obtain rewards.6 Olanzapine also interacts with dopaminergic (D1-5), muscarinic (M1-5), alpha1 adrenergic, histamine1, serotonin (5-HT2B,2C,3,6), and glutamate and other receptors.

Pharmacokinetics

Combining olanzapine and fluoxetine in one capsule raises potential kinetic problems. Olanzapine’s mean half-life is 30 hours,7 but fluoxetine’s is 24 to 72 hours and its principal active metabolite, norfluoxetine, has a half-life of 4 to 16 hours.7 Because fluoxetine and norfluoxetine inhibit the cytochrome P (CYP)-450 2D6 enzyme—which is involved in their metabolism—autoinhibition of degradation occurs with chronic dosing, thereby increasing the relative half-life of fluoxetine and norfluoxetine. Therefore, maximum steady-state plasma levels will be achieved with olanzapine and fluoxetine at very different rates, although this has not posed a problem in clinical trials. Still, consider this disparity when evaluating potential side effects or drug-drug interactions.

Table 1

Drugs that may interact with OFC

 

Drugs metabolized by CYP 2D6 isoenzymesDrugs metabolized by CYP 2C isoenzymes
CitalopramCitalopram
CodeineClomipramine
DextromethorphanDiazepam
HaloperidolImipramine
MetoprololNonsteroidal
Other SSRIsanti-inflammatory drugs
PerphenazineOmeprazole
PropafenonePhenytoin
PropranololProguanil
RisperidoneTolbutamide
ThioridazineTricyclic antidepressants
TrazodoneWarfarin
Tricyclic antidepressants (most) 
Venlafaxine 
Source: reference 8

Both compounds reach maximum concentration in 4 to 6 hours.7 Although food’s effect on OFC’s absorption has not been tested, a clinically important effect is unlikely. Food does not significantly alter absorption kinetics of olanzapine or fluoxetine.7

Avoid giving OFC concomitantly with drugs metabolized by CYP 2D6 and 2C (Table 1), because fluoxetine is a potent inhibitor of these isoenzymes. The resulting altered plasma concentrations could lead to drug-drug interactions.8

Efficacy

In an 8-week, double-blind, multinational trial,9 833 patients with bipolar I disorder in the depressive phase randomly received placebo, olanzapine alone (5 to 20 mg/d), or OFC in several fixed combinations (all shown as olanzapine/fluoxetine): 6/25 mg/d, 6/50 mg/d, or 12/50 mg/d. Dosage titration was allowed.

The researchers found that:

 

  • OFC was significantly more effective than placebo. A mean 18.5-point improvement in Montgomery-Asberg Depression Rating Scale (MADRS) scores was reported in the OFC group, compared with a mean 11.9-point improvement in the placebo group.
  • Olanzapine alone produced a mean 15-point MADRS score reduction. Remission criteria were achieved in 24.5%, 32.8%, and 48.8% of patients treated with placebo, olanzapine, and OFC, respectively.
  • Both OFC and olanzapine alone produced greater MADRS score reductions than did placebo at every follow-up week. Mania induction rates were low in the olanzapine and OFC treatment groups (5.7% and 6.4%, respectively) as measured with the Young Mania Rating Scale.

Shelton et al3 also compared OFC to olanzapine and fluoxetine alone in treatment-resistant unipolar depression. Thirty-two patients with major depression who responded inadequately to two types of antidepressants were treated with fluoxetine, up to 60 mg/d. After 7 weeks, 28 patients who did not respond to fluoxetine then received fluoxetine alone (mean modal dose: 52 mg/d), olanzapine alone (12.5 mg/d), or OFC (13.5 mg/52 mg/d) for another 8 weeks.

Olanzapine alone produced a transient effect at week 3 with relapse thereafter, possibly because of interactions between olanzapine and falling fluoxetine plasma concentrations over the first 3 weeks. Fluoxetine monotherapy produced minimal results across the 8-week random phase.

The OFC group, however, achieved significant improvement in MADRS scores compared with the placebo group after week one. The effect continued throughout the trial and during a subsequent 8-week open-label phase.3

Recent data suggest continued benefit in treatment- and nontreatment-resistant depressed patients for up to 1 year.10 Two follow-up trials—one using a lead-in with venlafaxine, the second with nortriptyline—produced negative results. In both studies, however, patients achieved a robust effect while continuing the same drug during the double-blind phase, suggesting that initial trials were inadequate.11,12 OFC showed early onset of effect in both studies. Other large-scale attempts at replication are anticipated.

 

 

Tolerability

Common side effects of OFC include increased appetite, weight gain, somnolence, fatigue, nausea, diarrhea, and dry mouth—the same effects associated with olanzapine or fluoxetine.

Combining the agents does not lessen the side effects, particularly olanzapine-induced weight gain. Simple, assertive dietary and exercise counseling can counteract olanzapine-induced weight gain.13 Sexual dysfunction was reported infrequently in clinical trials but is possible with exposure to fluoxetine.

Extrapyramidal side effects, including akathisia, appear to be relatively infrequent. Tardive dyskinesia (TD) is unlikely, although cases putatively associated with olanzapine have been reported.5 Many patients with TD have taken conventional antipsychotics, however, so the causal link with olanzapine is obscure. Still, alert patients and families to the possibility of TD and its emerging features.

Table 2

Olanzapine-fluoxetine: Fast facts

 

Drug brand name: Symbyax
Class: Combined atypical antipsychotic/selective serotonin reuptake inhibitor
FDA-approved indication: Bipolar type I depression
Approval date: Dec. 24, 2003
Manufacturer: Eli Lilly and Co.
Dosing forms: 6/25 mg/d, 12/50 mg/d, 12/25 mg/d, 12/50 mg/d
Dosing recommendations: Start at 6/25 mg at bedtime. Titrate according to tolerability and therapeutic benefit. Once the antidepressant effect is achieved, continue dosage indefinitely if no adverse effects occur. Dosages up to 18/75 mg/d have been used in clinical trials.

Although considered rare, isolated cases of neuroleptic malignant syndrome have been attributed to olanzapine.14 Cycle induction has not been reported in clinical trials, but be mindful of this possibility with long-term treatment.

Clinical implications

Taking olanzapine and fluoxetine as a single capsule could save the patient substantial cost. OFC comes in four dosing forms (Table 2), allowing for some flexibility.

It is unclear whether clinicians will prefer the single combination capsule or prescribe each drug separately to increase flexibility. Starting treatment with olanzapine and fluoxetine individually allows the psychiatrist to change the dosages independently and in smaller increments. Taken as separate agents, however, the two products are more expensive than the combined formula. OFC costs about the same as olanzapine alone. On the other hand, if the clinician begins the compounds individually, converting to the dosages in the combined product probably will not be exactly 1:1.

Tolerability is another major advantage of OFC; the combined agent exhibited a 10% dropout rate because of adverse effects compared with 4.6% for placebo.7 Moreover, some patients will prefer the convenience of using a single capsule instead of two medications.

Related resources

 

  • Tollefson GD, Sanger TM. Anxious-depressive symptoms in schizophrenia: a new treatment target for pharmacotherapy? Schizophr Res 1999;35(suppl):S13-S21.
  • Symbyax Web site. www.symbyax.com

Drug brand names

 

  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Diazepam • Valium
  • Fluoxetine • Prozac
  • Haloperidol • Haldol
  • Imipramine • Tofranil
  • Metoprolol succinate • Toprol
  • Nortriptyline • Aventyl
  • Olanzapine • Zyprexa
  • Omeprazole • Prilosec
  • Phenytoin • Dilantin
  • Proguanil • Malarone
  • Propafenone • Rythmol
  • Propranolol • Inderal
  • Risperidone • Risperdal
  • Tolbutamide • Orinase
  • Venlafaxine • Effexor
  • Warfarin • Coumadin

Disclosure

Dr. Shelton receives research grants from Abbott Laboratories, Eli Lilly and Co., GlaxoSmithKline, Janssen Pharmaceutica, Pfizer Inc., and Wyeth Pharmaceuticals; is a consultant to Janssen Pharmaceutica, Pfizer Inc., and Wyeth Pharmaceuticals; and is a speaker for Abbott Laboratories, Eli Lilly and Co., Forest Pharmaceuticals, GlaxoSmithKline, Janssen Pharmaceutica, Pfizer Inc., and Wyeth Pharmaceuticals

References

 

1. Judd LL, Akiskal HS, Schettler PJ, et al. The long-term natural history of the weekly symptomatic status of bipolar I disorder. Arch Gen Psychiatry 2002;59:530-7.

2. Nemeroff CB, Evans DL, Gyulai L, et al. Double-blind, placebo-controlled comparison of imipramine and paroxetine in the treatment of bipolar depression. Am J Psychiatry 2001;158:906-12.

3. Shelton RC, Tollefson GD, Tohen M, et al. A novel augmentation strategy for treating resistant major depression. Am J Psychiatry 2001;158:131-4.

4. Shelton RC. The combination of olanzapine and fluoxetine in mood disorders. Expert Opin Pharmacother 2003;4:1175-83.

5. Zhang W, Perry KW, Wong DT, et al. Synergistic effects of olanzapine and other antipsychotic agents in combination with fluoxetine on norepinephrine and dopamine release in rat prefrontal cortex. Neuropsychopharmacology 2000;23:250-62.

6. Salamone JD, Cousins MS, Snyder BJ. Behavioral functions of nucleus accumbens dopamine: empirical and conceptual problems with the anhedonia hypothesis. Neurosci Biobehav Rev 1997;21:341-59.

7. Symbyax package insert. Eli Lilly and Co., 2003.

8. Nemeroff CB, DeVane CL, Pollock BG. Newer antidepressants and the cytochrome P450 system. Am J Psychiatry 1996;153:311-20.

9. Tohen M, Vieta E, Calabrese J, et al. Efficacy of olanzapine and olanzapine-fluoxetine combination in the treatment of bipolar I depression. Arch Gen Psychiatry 2003;60:1079-88.

10. Corya SA, Andersen SW, Detke HC, et al. Long-term antidepressant efficacy and safety of olanzapine/fluoxetine combination: a 76-week open-label study. J Clin Psychiatry 2003;64:1349-56.

11. Dube S. Olanzapine-fluoxetine combination in treatment-resistant depression. Eur Psychiatry 2002;17(suppl 1):98.-

12. Dube S, Corya SA, Andersen SW, et al. Efficacy of olanzapine/fluoxetine combination in treatment resistant depression (presentation). San Juan, PR: American College of Neuropsychopharmacology annual meeting, 2002.

13. Ball MP, Coons VB, Buchanan RW. A program for treating olanzapine-related weight gain. Psychiatr Serv 2001;52:967-9.

14. Kogoj A, Velikonja I. Olanzapine-induced neuroleptic malignant syndrome—a case review. Hum Psychopharmacol 2003;18:301-9.

Author and Disclosure Information

 

Richard C. Shelton, MD
James K. Blakemore Professor, department of psychiatry Professor, department of pharmacology Vanderbilt University, Nashville, TN

Issue
Current Psychiatry - 03(04)
Publications
Topics
Page Number
85-88
Sections
Author and Disclosure Information

 

Richard C. Shelton, MD
James K. Blakemore Professor, department of psychiatry Professor, department of pharmacology Vanderbilt University, Nashville, TN

Author and Disclosure Information

 

Richard C. Shelton, MD
James K. Blakemore Professor, department of psychiatry Professor, department of pharmacology Vanderbilt University, Nashville, TN

Patients with bipolar disorder spend half their lives significantly symptomatic, mainly in the depressive phase.1 Treating bipolar depression poses a clinical challenge, although new research is starting to uncover some answers. Antidepressant drugs are commonly used, but recent data question the effectiveness of this practice.2

An olanzapine-fluoxetine combination (OFC), FDA-approved for treating bipolar type I depression, has demonstrated efficacy in clinical trials.

How it works

Most atypical antipsychotics—including olanzapine—are potent 5-HT2A (serotonin) receptor antagonists. This effect is similar to that of some antidepressants and may mediate some antidepressant and antianxiety effects of these drugs.3

Like most atypicals, olanzapine is also a potent 5-HT2C blocker. While binding to this receptor, serotonin inhibits dopamine release in the nucleus accumbens and frontal cortex.4 Thus, serotonin blockade would increase dopamine release in these areas. One study showed that olanzapine and fluoxetine together increased dopamine and norepinephrine in the frontal cortex of rats, compared with either drug given individually.5 Dopamine is critical to regulating motivation, defined as the ability to exert energy to obtain rewards.6 Olanzapine also interacts with dopaminergic (D1-5), muscarinic (M1-5), alpha1 adrenergic, histamine1, serotonin (5-HT2B,2C,3,6), and glutamate and other receptors.

Pharmacokinetics

Combining olanzapine and fluoxetine in one capsule raises potential kinetic problems. Olanzapine’s mean half-life is 30 hours,7 but fluoxetine’s is 24 to 72 hours and its principal active metabolite, norfluoxetine, has a half-life of 4 to 16 hours.7 Because fluoxetine and norfluoxetine inhibit the cytochrome P (CYP)-450 2D6 enzyme—which is involved in their metabolism—autoinhibition of degradation occurs with chronic dosing, thereby increasing the relative half-life of fluoxetine and norfluoxetine. Therefore, maximum steady-state plasma levels will be achieved with olanzapine and fluoxetine at very different rates, although this has not posed a problem in clinical trials. Still, consider this disparity when evaluating potential side effects or drug-drug interactions.

Table 1

Drugs that may interact with OFC

 

Drugs metabolized by CYP 2D6 isoenzymesDrugs metabolized by CYP 2C isoenzymes
CitalopramCitalopram
CodeineClomipramine
DextromethorphanDiazepam
HaloperidolImipramine
MetoprololNonsteroidal
Other SSRIsanti-inflammatory drugs
PerphenazineOmeprazole
PropafenonePhenytoin
PropranololProguanil
RisperidoneTolbutamide
ThioridazineTricyclic antidepressants
TrazodoneWarfarin
Tricyclic antidepressants (most) 
Venlafaxine 
Source: reference 8

Both compounds reach maximum concentration in 4 to 6 hours.7 Although food’s effect on OFC’s absorption has not been tested, a clinically important effect is unlikely. Food does not significantly alter absorption kinetics of olanzapine or fluoxetine.7

Avoid giving OFC concomitantly with drugs metabolized by CYP 2D6 and 2C (Table 1), because fluoxetine is a potent inhibitor of these isoenzymes. The resulting altered plasma concentrations could lead to drug-drug interactions.8

Efficacy

In an 8-week, double-blind, multinational trial,9 833 patients with bipolar I disorder in the depressive phase randomly received placebo, olanzapine alone (5 to 20 mg/d), or OFC in several fixed combinations (all shown as olanzapine/fluoxetine): 6/25 mg/d, 6/50 mg/d, or 12/50 mg/d. Dosage titration was allowed.

The researchers found that:

 

  • OFC was significantly more effective than placebo. A mean 18.5-point improvement in Montgomery-Asberg Depression Rating Scale (MADRS) scores was reported in the OFC group, compared with a mean 11.9-point improvement in the placebo group.
  • Olanzapine alone produced a mean 15-point MADRS score reduction. Remission criteria were achieved in 24.5%, 32.8%, and 48.8% of patients treated with placebo, olanzapine, and OFC, respectively.
  • Both OFC and olanzapine alone produced greater MADRS score reductions than did placebo at every follow-up week. Mania induction rates were low in the olanzapine and OFC treatment groups (5.7% and 6.4%, respectively) as measured with the Young Mania Rating Scale.

Shelton et al3 also compared OFC to olanzapine and fluoxetine alone in treatment-resistant unipolar depression. Thirty-two patients with major depression who responded inadequately to two types of antidepressants were treated with fluoxetine, up to 60 mg/d. After 7 weeks, 28 patients who did not respond to fluoxetine then received fluoxetine alone (mean modal dose: 52 mg/d), olanzapine alone (12.5 mg/d), or OFC (13.5 mg/52 mg/d) for another 8 weeks.

Olanzapine alone produced a transient effect at week 3 with relapse thereafter, possibly because of interactions between olanzapine and falling fluoxetine plasma concentrations over the first 3 weeks. Fluoxetine monotherapy produced minimal results across the 8-week random phase.

The OFC group, however, achieved significant improvement in MADRS scores compared with the placebo group after week one. The effect continued throughout the trial and during a subsequent 8-week open-label phase.3

Recent data suggest continued benefit in treatment- and nontreatment-resistant depressed patients for up to 1 year.10 Two follow-up trials—one using a lead-in with venlafaxine, the second with nortriptyline—produced negative results. In both studies, however, patients achieved a robust effect while continuing the same drug during the double-blind phase, suggesting that initial trials were inadequate.11,12 OFC showed early onset of effect in both studies. Other large-scale attempts at replication are anticipated.

 

 

Tolerability

Common side effects of OFC include increased appetite, weight gain, somnolence, fatigue, nausea, diarrhea, and dry mouth—the same effects associated with olanzapine or fluoxetine.

Combining the agents does not lessen the side effects, particularly olanzapine-induced weight gain. Simple, assertive dietary and exercise counseling can counteract olanzapine-induced weight gain.13 Sexual dysfunction was reported infrequently in clinical trials but is possible with exposure to fluoxetine.

Extrapyramidal side effects, including akathisia, appear to be relatively infrequent. Tardive dyskinesia (TD) is unlikely, although cases putatively associated with olanzapine have been reported.5 Many patients with TD have taken conventional antipsychotics, however, so the causal link with olanzapine is obscure. Still, alert patients and families to the possibility of TD and its emerging features.

Table 2

Olanzapine-fluoxetine: Fast facts

 

Drug brand name: Symbyax
Class: Combined atypical antipsychotic/selective serotonin reuptake inhibitor
FDA-approved indication: Bipolar type I depression
Approval date: Dec. 24, 2003
Manufacturer: Eli Lilly and Co.
Dosing forms: 6/25 mg/d, 12/50 mg/d, 12/25 mg/d, 12/50 mg/d
Dosing recommendations: Start at 6/25 mg at bedtime. Titrate according to tolerability and therapeutic benefit. Once the antidepressant effect is achieved, continue dosage indefinitely if no adverse effects occur. Dosages up to 18/75 mg/d have been used in clinical trials.

Although considered rare, isolated cases of neuroleptic malignant syndrome have been attributed to olanzapine.14 Cycle induction has not been reported in clinical trials, but be mindful of this possibility with long-term treatment.

Clinical implications

Taking olanzapine and fluoxetine as a single capsule could save the patient substantial cost. OFC comes in four dosing forms (Table 2), allowing for some flexibility.

It is unclear whether clinicians will prefer the single combination capsule or prescribe each drug separately to increase flexibility. Starting treatment with olanzapine and fluoxetine individually allows the psychiatrist to change the dosages independently and in smaller increments. Taken as separate agents, however, the two products are more expensive than the combined formula. OFC costs about the same as olanzapine alone. On the other hand, if the clinician begins the compounds individually, converting to the dosages in the combined product probably will not be exactly 1:1.

Tolerability is another major advantage of OFC; the combined agent exhibited a 10% dropout rate because of adverse effects compared with 4.6% for placebo.7 Moreover, some patients will prefer the convenience of using a single capsule instead of two medications.

Related resources

 

  • Tollefson GD, Sanger TM. Anxious-depressive symptoms in schizophrenia: a new treatment target for pharmacotherapy? Schizophr Res 1999;35(suppl):S13-S21.
  • Symbyax Web site. www.symbyax.com

Drug brand names

 

  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Diazepam • Valium
  • Fluoxetine • Prozac
  • Haloperidol • Haldol
  • Imipramine • Tofranil
  • Metoprolol succinate • Toprol
  • Nortriptyline • Aventyl
  • Olanzapine • Zyprexa
  • Omeprazole • Prilosec
  • Phenytoin • Dilantin
  • Proguanil • Malarone
  • Propafenone • Rythmol
  • Propranolol • Inderal
  • Risperidone • Risperdal
  • Tolbutamide • Orinase
  • Venlafaxine • Effexor
  • Warfarin • Coumadin

Disclosure

Dr. Shelton receives research grants from Abbott Laboratories, Eli Lilly and Co., GlaxoSmithKline, Janssen Pharmaceutica, Pfizer Inc., and Wyeth Pharmaceuticals; is a consultant to Janssen Pharmaceutica, Pfizer Inc., and Wyeth Pharmaceuticals; and is a speaker for Abbott Laboratories, Eli Lilly and Co., Forest Pharmaceuticals, GlaxoSmithKline, Janssen Pharmaceutica, Pfizer Inc., and Wyeth Pharmaceuticals

Patients with bipolar disorder spend half their lives significantly symptomatic, mainly in the depressive phase.1 Treating bipolar depression poses a clinical challenge, although new research is starting to uncover some answers. Antidepressant drugs are commonly used, but recent data question the effectiveness of this practice.2

An olanzapine-fluoxetine combination (OFC), FDA-approved for treating bipolar type I depression, has demonstrated efficacy in clinical trials.

How it works

Most atypical antipsychotics—including olanzapine—are potent 5-HT2A (serotonin) receptor antagonists. This effect is similar to that of some antidepressants and may mediate some antidepressant and antianxiety effects of these drugs.3

Like most atypicals, olanzapine is also a potent 5-HT2C blocker. While binding to this receptor, serotonin inhibits dopamine release in the nucleus accumbens and frontal cortex.4 Thus, serotonin blockade would increase dopamine release in these areas. One study showed that olanzapine and fluoxetine together increased dopamine and norepinephrine in the frontal cortex of rats, compared with either drug given individually.5 Dopamine is critical to regulating motivation, defined as the ability to exert energy to obtain rewards.6 Olanzapine also interacts with dopaminergic (D1-5), muscarinic (M1-5), alpha1 adrenergic, histamine1, serotonin (5-HT2B,2C,3,6), and glutamate and other receptors.

Pharmacokinetics

Combining olanzapine and fluoxetine in one capsule raises potential kinetic problems. Olanzapine’s mean half-life is 30 hours,7 but fluoxetine’s is 24 to 72 hours and its principal active metabolite, norfluoxetine, has a half-life of 4 to 16 hours.7 Because fluoxetine and norfluoxetine inhibit the cytochrome P (CYP)-450 2D6 enzyme—which is involved in their metabolism—autoinhibition of degradation occurs with chronic dosing, thereby increasing the relative half-life of fluoxetine and norfluoxetine. Therefore, maximum steady-state plasma levels will be achieved with olanzapine and fluoxetine at very different rates, although this has not posed a problem in clinical trials. Still, consider this disparity when evaluating potential side effects or drug-drug interactions.

Table 1

Drugs that may interact with OFC

 

Drugs metabolized by CYP 2D6 isoenzymesDrugs metabolized by CYP 2C isoenzymes
CitalopramCitalopram
CodeineClomipramine
DextromethorphanDiazepam
HaloperidolImipramine
MetoprololNonsteroidal
Other SSRIsanti-inflammatory drugs
PerphenazineOmeprazole
PropafenonePhenytoin
PropranololProguanil
RisperidoneTolbutamide
ThioridazineTricyclic antidepressants
TrazodoneWarfarin
Tricyclic antidepressants (most) 
Venlafaxine 
Source: reference 8

Both compounds reach maximum concentration in 4 to 6 hours.7 Although food’s effect on OFC’s absorption has not been tested, a clinically important effect is unlikely. Food does not significantly alter absorption kinetics of olanzapine or fluoxetine.7

Avoid giving OFC concomitantly with drugs metabolized by CYP 2D6 and 2C (Table 1), because fluoxetine is a potent inhibitor of these isoenzymes. The resulting altered plasma concentrations could lead to drug-drug interactions.8

Efficacy

In an 8-week, double-blind, multinational trial,9 833 patients with bipolar I disorder in the depressive phase randomly received placebo, olanzapine alone (5 to 20 mg/d), or OFC in several fixed combinations (all shown as olanzapine/fluoxetine): 6/25 mg/d, 6/50 mg/d, or 12/50 mg/d. Dosage titration was allowed.

The researchers found that:

 

  • OFC was significantly more effective than placebo. A mean 18.5-point improvement in Montgomery-Asberg Depression Rating Scale (MADRS) scores was reported in the OFC group, compared with a mean 11.9-point improvement in the placebo group.
  • Olanzapine alone produced a mean 15-point MADRS score reduction. Remission criteria were achieved in 24.5%, 32.8%, and 48.8% of patients treated with placebo, olanzapine, and OFC, respectively.
  • Both OFC and olanzapine alone produced greater MADRS score reductions than did placebo at every follow-up week. Mania induction rates were low in the olanzapine and OFC treatment groups (5.7% and 6.4%, respectively) as measured with the Young Mania Rating Scale.

Shelton et al3 also compared OFC to olanzapine and fluoxetine alone in treatment-resistant unipolar depression. Thirty-two patients with major depression who responded inadequately to two types of antidepressants were treated with fluoxetine, up to 60 mg/d. After 7 weeks, 28 patients who did not respond to fluoxetine then received fluoxetine alone (mean modal dose: 52 mg/d), olanzapine alone (12.5 mg/d), or OFC (13.5 mg/52 mg/d) for another 8 weeks.

Olanzapine alone produced a transient effect at week 3 with relapse thereafter, possibly because of interactions between olanzapine and falling fluoxetine plasma concentrations over the first 3 weeks. Fluoxetine monotherapy produced minimal results across the 8-week random phase.

The OFC group, however, achieved significant improvement in MADRS scores compared with the placebo group after week one. The effect continued throughout the trial and during a subsequent 8-week open-label phase.3

Recent data suggest continued benefit in treatment- and nontreatment-resistant depressed patients for up to 1 year.10 Two follow-up trials—one using a lead-in with venlafaxine, the second with nortriptyline—produced negative results. In both studies, however, patients achieved a robust effect while continuing the same drug during the double-blind phase, suggesting that initial trials were inadequate.11,12 OFC showed early onset of effect in both studies. Other large-scale attempts at replication are anticipated.

 

 

Tolerability

Common side effects of OFC include increased appetite, weight gain, somnolence, fatigue, nausea, diarrhea, and dry mouth—the same effects associated with olanzapine or fluoxetine.

Combining the agents does not lessen the side effects, particularly olanzapine-induced weight gain. Simple, assertive dietary and exercise counseling can counteract olanzapine-induced weight gain.13 Sexual dysfunction was reported infrequently in clinical trials but is possible with exposure to fluoxetine.

Extrapyramidal side effects, including akathisia, appear to be relatively infrequent. Tardive dyskinesia (TD) is unlikely, although cases putatively associated with olanzapine have been reported.5 Many patients with TD have taken conventional antipsychotics, however, so the causal link with olanzapine is obscure. Still, alert patients and families to the possibility of TD and its emerging features.

Table 2

Olanzapine-fluoxetine: Fast facts

 

Drug brand name: Symbyax
Class: Combined atypical antipsychotic/selective serotonin reuptake inhibitor
FDA-approved indication: Bipolar type I depression
Approval date: Dec. 24, 2003
Manufacturer: Eli Lilly and Co.
Dosing forms: 6/25 mg/d, 12/50 mg/d, 12/25 mg/d, 12/50 mg/d
Dosing recommendations: Start at 6/25 mg at bedtime. Titrate according to tolerability and therapeutic benefit. Once the antidepressant effect is achieved, continue dosage indefinitely if no adverse effects occur. Dosages up to 18/75 mg/d have been used in clinical trials.

Although considered rare, isolated cases of neuroleptic malignant syndrome have been attributed to olanzapine.14 Cycle induction has not been reported in clinical trials, but be mindful of this possibility with long-term treatment.

Clinical implications

Taking olanzapine and fluoxetine as a single capsule could save the patient substantial cost. OFC comes in four dosing forms (Table 2), allowing for some flexibility.

It is unclear whether clinicians will prefer the single combination capsule or prescribe each drug separately to increase flexibility. Starting treatment with olanzapine and fluoxetine individually allows the psychiatrist to change the dosages independently and in smaller increments. Taken as separate agents, however, the two products are more expensive than the combined formula. OFC costs about the same as olanzapine alone. On the other hand, if the clinician begins the compounds individually, converting to the dosages in the combined product probably will not be exactly 1:1.

Tolerability is another major advantage of OFC; the combined agent exhibited a 10% dropout rate because of adverse effects compared with 4.6% for placebo.7 Moreover, some patients will prefer the convenience of using a single capsule instead of two medications.

Related resources

 

  • Tollefson GD, Sanger TM. Anxious-depressive symptoms in schizophrenia: a new treatment target for pharmacotherapy? Schizophr Res 1999;35(suppl):S13-S21.
  • Symbyax Web site. www.symbyax.com

Drug brand names

 

  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Diazepam • Valium
  • Fluoxetine • Prozac
  • Haloperidol • Haldol
  • Imipramine • Tofranil
  • Metoprolol succinate • Toprol
  • Nortriptyline • Aventyl
  • Olanzapine • Zyprexa
  • Omeprazole • Prilosec
  • Phenytoin • Dilantin
  • Proguanil • Malarone
  • Propafenone • Rythmol
  • Propranolol • Inderal
  • Risperidone • Risperdal
  • Tolbutamide • Orinase
  • Venlafaxine • Effexor
  • Warfarin • Coumadin

Disclosure

Dr. Shelton receives research grants from Abbott Laboratories, Eli Lilly and Co., GlaxoSmithKline, Janssen Pharmaceutica, Pfizer Inc., and Wyeth Pharmaceuticals; is a consultant to Janssen Pharmaceutica, Pfizer Inc., and Wyeth Pharmaceuticals; and is a speaker for Abbott Laboratories, Eli Lilly and Co., Forest Pharmaceuticals, GlaxoSmithKline, Janssen Pharmaceutica, Pfizer Inc., and Wyeth Pharmaceuticals

References

 

1. Judd LL, Akiskal HS, Schettler PJ, et al. The long-term natural history of the weekly symptomatic status of bipolar I disorder. Arch Gen Psychiatry 2002;59:530-7.

2. Nemeroff CB, Evans DL, Gyulai L, et al. Double-blind, placebo-controlled comparison of imipramine and paroxetine in the treatment of bipolar depression. Am J Psychiatry 2001;158:906-12.

3. Shelton RC, Tollefson GD, Tohen M, et al. A novel augmentation strategy for treating resistant major depression. Am J Psychiatry 2001;158:131-4.

4. Shelton RC. The combination of olanzapine and fluoxetine in mood disorders. Expert Opin Pharmacother 2003;4:1175-83.

5. Zhang W, Perry KW, Wong DT, et al. Synergistic effects of olanzapine and other antipsychotic agents in combination with fluoxetine on norepinephrine and dopamine release in rat prefrontal cortex. Neuropsychopharmacology 2000;23:250-62.

6. Salamone JD, Cousins MS, Snyder BJ. Behavioral functions of nucleus accumbens dopamine: empirical and conceptual problems with the anhedonia hypothesis. Neurosci Biobehav Rev 1997;21:341-59.

7. Symbyax package insert. Eli Lilly and Co., 2003.

8. Nemeroff CB, DeVane CL, Pollock BG. Newer antidepressants and the cytochrome P450 system. Am J Psychiatry 1996;153:311-20.

9. Tohen M, Vieta E, Calabrese J, et al. Efficacy of olanzapine and olanzapine-fluoxetine combination in the treatment of bipolar I depression. Arch Gen Psychiatry 2003;60:1079-88.

10. Corya SA, Andersen SW, Detke HC, et al. Long-term antidepressant efficacy and safety of olanzapine/fluoxetine combination: a 76-week open-label study. J Clin Psychiatry 2003;64:1349-56.

11. Dube S. Olanzapine-fluoxetine combination in treatment-resistant depression. Eur Psychiatry 2002;17(suppl 1):98.-

12. Dube S, Corya SA, Andersen SW, et al. Efficacy of olanzapine/fluoxetine combination in treatment resistant depression (presentation). San Juan, PR: American College of Neuropsychopharmacology annual meeting, 2002.

13. Ball MP, Coons VB, Buchanan RW. A program for treating olanzapine-related weight gain. Psychiatr Serv 2001;52:967-9.

14. Kogoj A, Velikonja I. Olanzapine-induced neuroleptic malignant syndrome—a case review. Hum Psychopharmacol 2003;18:301-9.

References

 

1. Judd LL, Akiskal HS, Schettler PJ, et al. The long-term natural history of the weekly symptomatic status of bipolar I disorder. Arch Gen Psychiatry 2002;59:530-7.

2. Nemeroff CB, Evans DL, Gyulai L, et al. Double-blind, placebo-controlled comparison of imipramine and paroxetine in the treatment of bipolar depression. Am J Psychiatry 2001;158:906-12.

3. Shelton RC, Tollefson GD, Tohen M, et al. A novel augmentation strategy for treating resistant major depression. Am J Psychiatry 2001;158:131-4.

4. Shelton RC. The combination of olanzapine and fluoxetine in mood disorders. Expert Opin Pharmacother 2003;4:1175-83.

5. Zhang W, Perry KW, Wong DT, et al. Synergistic effects of olanzapine and other antipsychotic agents in combination with fluoxetine on norepinephrine and dopamine release in rat prefrontal cortex. Neuropsychopharmacology 2000;23:250-62.

6. Salamone JD, Cousins MS, Snyder BJ. Behavioral functions of nucleus accumbens dopamine: empirical and conceptual problems with the anhedonia hypothesis. Neurosci Biobehav Rev 1997;21:341-59.

7. Symbyax package insert. Eli Lilly and Co., 2003.

8. Nemeroff CB, DeVane CL, Pollock BG. Newer antidepressants and the cytochrome P450 system. Am J Psychiatry 1996;153:311-20.

9. Tohen M, Vieta E, Calabrese J, et al. Efficacy of olanzapine and olanzapine-fluoxetine combination in the treatment of bipolar I depression. Arch Gen Psychiatry 2003;60:1079-88.

10. Corya SA, Andersen SW, Detke HC, et al. Long-term antidepressant efficacy and safety of olanzapine/fluoxetine combination: a 76-week open-label study. J Clin Psychiatry 2003;64:1349-56.

11. Dube S. Olanzapine-fluoxetine combination in treatment-resistant depression. Eur Psychiatry 2002;17(suppl 1):98.-

12. Dube S, Corya SA, Andersen SW, et al. Efficacy of olanzapine/fluoxetine combination in treatment resistant depression (presentation). San Juan, PR: American College of Neuropsychopharmacology annual meeting, 2002.

13. Ball MP, Coons VB, Buchanan RW. A program for treating olanzapine-related weight gain. Psychiatr Serv 2001;52:967-9.

14. Kogoj A, Velikonja I. Olanzapine-induced neuroleptic malignant syndrome—a case review. Hum Psychopharmacol 2003;18:301-9.

Issue
Current Psychiatry - 03(04)
Issue
Current Psychiatry - 03(04)
Page Number
85-88
Page Number
85-88
Publications
Publications
Topics
Article Type
Display Headline
Olanzapine/fluoxetine combination: Evidence for using the first treatment indicated for bipolar depression
Display Headline
Olanzapine/fluoxetine combination: Evidence for using the first treatment indicated for bipolar depression
Sections
Disallow All Ads
Alternative CME

Bipolar maintenance: Are atypical antipsychotics really ‘mood stabilizers’?

Article Type
Changed
Tue, 12/11/2018 - 15:10
Display Headline
Bipolar maintenance: Are atypical antipsychotics really ‘mood stabilizers’?

Maintenance therapy with mood stabilizers is the most critical phase of bipolar disorder treatment but the stage with the least available evidence about medication risks and benefits. The FDA’s recent approval of olanzapine for bipolar maintenance raises the question of whether atypical antipsychotics are really mood stabilizers. This article attempts to answer that question by:

  • describing the “ideal” mood stabilizer
  • discussing atypicals’ advantages over conventional antipsychotics in bipolar patients
  • comparing efficacy data for the six available atypicals
  • recommending strategies to prevent and treat atypicals’ potentially serious side effects during long-term therapy.

What is a ‘mood stabilizer’?

Successful mood stabilizer maintenance therapy decreases the time patients are sick and disabled. Although somewhat dated after only 2 years, the most recent American Psychiatric Association (APA) practice guidelines1 support using mood stabilizers for patients with bipolar I and bipolar II disorders.

Table 1

Bipolar maintenance treatment goals

  • Prevent manic or depressive relapse
  • Reduce subthreshold symptoms
  • Reduce suicide risk
  • Reduce cycling frequency
  • Reduce mood instability
  • Improve functioning
Adapted from American Psychiatric Association practice guidelines for treating patients with bipolar disorder (reference 1)

The goals of maintenance therapy are listed in Table 1. The ideal mood stabilizer would work in maintenance and all bipolar phases and treatment stages—from treating acute depression, mania, hypomania, and mixed states to preventing abnormal mood elevations and depressions. It would not precipitate depression or mania, rapid cycling, or cycle acceleration.

In other words, the best “mood stabilizer” would work in all four treatment roles of bipolar disorder: treating highs and lows, and preventing highs and lows. No such mood stabilizer exists, although lithium may come closest to the ideal.2

Most U.S. psychiatrists use combination therapies for bipolar disorder, particularly when treating acute manic states. The most common combination is a “known” mood stabilizer—such as lithium or divalproex—plus an antipsychotic to quickly control mania.

After mania remits, clinicians often try to eliminate the antipsychotic in hopes of maintaining mood stability and euthymia with the mood stabilizer alone. This was especially true before atypical antipsychotics were approved, given the risk for tardive dyskinesia (TD) associated with long-term use of conventional antipsychotics.

Unfortunately, patients frequently relapse with this strategy, so psychiatrists may leave their bipolar patients on atypical antipsychotics during long-term maintenance. But how good are atypicals as mood stabilizers? Perhaps more importantly, how safe is long-term use of atypicals in bipolar patients?

Antipsychotics as mood stabilizers

The 2002 APA practice guidelines discuss efficacy data for using lithium, divalproex or valproate, lamotrigine, carbamazepine, and electroconvulsive therapy for bipolar maintenance treatment. Two sentences on antipsychotic drug use note:

  • one placebo-controlled study of a conventional antipsychotic showing no efficacy
  • some data supporting clozapine as a prophylactic bipolar treatment.1

A 1998 review of five open trials3 touched on conventional depot antipsychotics’ value in reducing manic or affective illness. However, the authors warned:

  • no controlled trials existed
  • maintenance antipsychotic treatment may be associated with increased risk for tardive movement disorders
  • conventional agents can exacerbate depressive symptoms in some patients.

Using conventional antipsychotics long-term in bipolar disorder is not advisable, with the possible exception of depot preparations in nonadhering patients with severe illness. Long-acting injectable atypicals—such as the recently approved IM risperidone—may displace any use of conventional antipsychotics in bipolar patients.

Atypical antipsychotics hold several advantages over conventional agents:

  • significantly reduced risk for TD and extrapyramidal symptoms (EPS)
  • lack of serum prolactin elevation (except with risperidone)
  • improved cognition
  • possible decreased suicidality, particularly with clozapine.4

Table 2

Tips for managing atypicals’ potentially serious side-effect risks

Weight gain/obesity
AssessmentPreventionTreatment
Evaluate comorbid conditions such as eating disorders or substance abuse
Take nutritional and exercise history
Check weight and waist circumference at baseline and every visit
Calculate body mass index at every visit
Prescribe healthy diet and exercise
Patient education, careful monitoring, and prevention are most-effective treatments
Drug therapy for persistent weight gain or early rapid gain (>7% in first 6 months). Agents of potential benefit include topiramate, sibutramine, metformin, zonisamide, and orlistat (see Table 3)
Glucose control/type 2 diabetes
AssessmentPreventionTreatment
Take history of glucose intolerance or diabetes
Ask about family history of diabetes, obesity, hypertension, heart disease
Check baseline weight and plasma glucose
Obtain fasting plasma glucose every 3 months for first year, then annually
Prescribe healthy diet and exercise
Primary prevention through careful monitoring is most effective
Discontinue atypical antipsychotic; use other mood stabilizer unless atypical is only effective drug for that patient
Oral hypoglycemics (metformin, others)
Hyperlipidemia
AssessmentPreventionTreatment
Take history of hyperlipidemia or cardiovascular disease
Ask about family history of hyperlipidemia
Check fasting lipid profile including triglycerides at baseline and every 3 months in first year
Prescribe healthy diet and exercise
Monitor diet, exercise, weight, lipids regularly
Change atypical antipsychotic or use other mood stabilizer (as described above)
Oral antilipemics (simvastatin, others)
 

 

Evidence for atypicals

Olanzapine is the only atypical FDA-approved for relapse prevention in bipolar disorder. This approval is supported by several studies, most notably two 1-year, double-blind trials:

  • Mean time to any mood relapse was 174 days in patients taking olanzapine, mean 12.5 mg/d (±5 mg), compared with 22 days in a placebo group (Eli Lilly and Co., data on file).
  • Manic relapse rate was 14.3% in patients treated with olanzapine, ~12 mg/d, compared with 28% in patients treated with lithium, ~1,100 mg/d (mean 0.76 mEq/L). The two treatments were similarly effective in preventing depressive relapse.5

As a mood stabilizer, olanzapine was as effective as divalproex in a 47-week randomized, double-blind study of 251 adults with bipolar I disorder.6 Patients treated with olanzapine improved more rapidly and had fewer manic symptoms than those treated with divalproex, but bipolar relapse rates were similar in both treatment groups.

Risperidone appears to have a role as a potential maintenance mood stabilizer in bipolar patients, although double-blind trials are lacking.

In a 6-month, open-label investigation, relapse rates were 16% for depression and 7% for mania in bipolar patients receiving risperidone (average 4 mg/d) combined with mood-stabilizing medications.7 These relapse rates are lower than those typically reported for mood-stabilizing monotherapy.

In another 6-month, open-label study, risperidone monotherapy (average 4 mg/d) was effective for treating mania and maintaining euthymia.8

IM risperidone is a useful option for bipolar patients chronically nonadherent with oral medications; it also substantially reduces the risk of neuroleptic side effects compared with older depot antipsychotics.

Quetiapine was recently approved as an antimanic agent and may possess mood-stabilizing properties. In a preliminary study of 10 patients with bipolar disorder, adding quetiapine (mean 200 mg/d) to existing mood stabilizer therapy for 12 weeks improved psychopathology, mania, and depression rating scale scores.9

More-recent unpublished data suggest dosing quetiapine to approximately 600 mg/d as monotherapy or an adjunct to treat acute mania, though controlled maintenance studies are lacking (AstraZeneca Pharmaceuticals, data on file).

Others. Some early evidence supports using ziprasidone and aripiprazole for bipolar mania:

  • Ziprasidone monotherapy, 40 to 80 mg bid, was significantly more effective than placebo in reducing acute mania symptoms in a 3-week, double-blind, randomized trial of 197 patients with bipolar I disorder.10
  • Aripiprazole monotherapy, 15 to 30 mg/d, had a significantly greater effect than placebo in a 3-week, double-blind, randomized trial of 262 patients in acute manic or mixed bipolar episodes. Response rates among patients with mania were 40% with aripiprazole and 19% with placebo.11

Both ziprasidone and aripiprazole were well-tolerated in these brief trials, although their efficacy as long-term mood-stabilizers in bipolar disorder is unclear.

Using clozapine raises concerns about potentially serious adverse events, although it remains the only agent with proven efficacy in treatment-refractory mania.12,13 Clozapine also appears to reduce hospitalization and affective relapse rates and improve symptoms and quality of life.14,15

Long-term safety

Compared with conventional antipsychotics, EPS are not a major concern with the atypical agents. Except for risperidone, atypicals’ effect on prolactin levels generally is not clinically meaningful. Atypicals appear to be “mood-friendly,” whereas conventional antipsychotics seem to contribute to dysphoria or cause depression in some patients.

Sedation or other annoying side effects such as dry mouth or dizziness can occur with any atypical. Other more-serious side effects may complicate antipsychotic treatment, as we are coming to understand from using atypicals for long-term schizophrenia management.

Table 3

Weight-loss medications for bipolar patients taking atypical antipsychotics

DrugDosageSide effectsRecommendations
Metformin500 to 1,000 mg bidHypoglycemia
Diarrhea
Nausea/vomiting
First-line in patients with comorbid type 2 diabetes
Orlistat120 mg tidGI distress
Change in bowel
habits
Second-line
For patients with BMI >27
Supplement fat-soluble vitamins
Sibutramine5 to 15 mg/dDry mouth
Anorexia
Insomnia
Constipation
Second-line
For patients with BMI 27 to 30
Risk of serotonin syndrome if given with serotonergic drugs
Topiramate50 to 250 mg/dSomnolence
Fatigue
Paresthesias
Consider first-line for its potential additive mood-stabilizing effect
May help comorbid binge-eating or seizure disorders
Zonisamide100 to 600 mg/dSomnolence
Dizziness
Anorexia
Consider first-line for its potential additive mood-stabilizing effect
May help comorbid binge-eating or seizure disorders

Movement disorders. Antipsychotics appear more likely to cause EPS in patients with mood disorders than with schizophrenia. In one study using conventional antipsychotics, bipolar patients were 4 to 5 times more likely than schizophrenia patients to experience acute dystonia.16

Although atypicals pose some small risk for acute EPS and TD, the risk is near placebo-level with clinically relevant and comparable dosages.17 Even so, it is important to educate patients to watch for emerging signs of TD during long-term treatment with any antipsychotic. EPS risk may be dose-dependent, particularly with risperidone.18

Weight gain and obesity. Patients with bipolar disorder are more likely to be overweight or obese (body mass index [BMI] > 30) than the general population,17,19 though the reasons are unknown. Studies suggest an obesity prevalence of 32% to 35% in bipolar patients, compared with 18% in the general population.20,21

 

 

All atypicals can cause weight gain, although olanzapine and clozapine are associated with the greatest mean weight gains. In three long-term trials (47 weeks to 18 months), bipolar patients who received olanzapine gained significantly more weight (mean 2 to 3 kg) than those receiving lithium or divalproex.19

Cases with much greater weight gain—even leading to clinical obesity—have been observed, particularly with olanzapine. Although evidence from registration trials and clinical experience show lesser weight gains with risperidone, quetiapine, ziprasidone, and aripiprazole, some of our patients do gain weight while taking these agents—either alone or in combination with lithium or divalproex.

Weight management. Because patients with bipolar disorder may be at increased risk for weight gain and obesity, weight management techniques may improve their health by:

  • decreasing morbidity and mortality tied to weight-related physical illnesses
  • enhancing psychological well-being.1

In addition to diet and exercise counseling, some bipolar patients taking long-term atypical antipsychotics may benefit from adjunctive weight-loss medications (Table 3). We generally use such medications for bipolar patients who:

  • persistently gain weight despite best dietary practices
  • gain substantial weight early in treatment with an atypical antipsychotic that is providing effective symptomatic relief.

Early weight gain—particularly gains of >7% within the first 6 weeks—might predict large weight gain over time.

Diabetes. In September 2003, the FDA requested a class-wide labeling change to warn about a possible link between atypical antipsychotics and diabetes. The FDA recommended blood sugar monitoring of patients taking atypicals, especially those with obesity risk factors or family history of diabetes.

Type 2 diabetes develops in some patients taking atypicals, whether or not they gain substantial weight.22 This suggests that weight gain associated with bipolar disorder and the use of atypical antipsychotics may be independent risk factors for diabetes—a clear concern when treating bipolar patients.

Evidence provides no clear answer as to which atypicals may increase diabetes risk. Cautious use and vigilant monitoring of blood glucose are therefore recommended for every patient taking an atypical for long-term therapy. Also watch for increases in triglycerides and cholesterol17 in patients taking atypicals as bipolar maintenance therapy.

Conclusion

Atypical antipsychotics are valuable therapies in preventing bipolar relapses, although olanzapine is the only atypical with this indication so far. Collective data and clinical experience suggest that atypicals are indeed mood stabilizers, although—like other mood stabilizers such as lithium or divalproex—they have limitations. None achieve ideal efficacy in all four bipolar treatment roles: treating the highs and lows, and preventing the highs and lows. Atypicals seem more effective in treating and preventing the highs than the lows, reminding us that effective depression treatment is the greatest unmet need in bipolar disorder.

More double-blind, randomized, controlled trials are needed to fully understand whether all atypicals are mood stabilizers and to determine their safety and side effects in long-term therapy for patients with bipolar disorder.

Related resources

  • Depression and Bipolar Support Alliance. www.dbsalliance.org
  • Muzina DJ, Calabrese JR. Guidelines for treatment of bipolar disorder.In: Stein DJ, Kupfer DJ, Schatzberg AF (eds). Textbook of mood disorders Washington, DC: American Psychiatric Publishing, 2004 (in press).

Drug brand names

  • Aripiprazole • Abilify
  • Carbamazepine • Tegretol
  • Clozapine • Clozaril
  • Divalproex/valproate • Depakote, Depakene
  • Lamotrigine • Lamictal
  • Lithium • Eskalith, Lithobid, et al
  • Metformin • Glucophage
  • Olanzapine • Zyprexa
  • Orlistat • Xenical
  • Quetiapine • Seroquel
  • Risperidone • Risperdal, Risperdal Consta
  • Sibutramine • Meridia
  • Simvastatin • Zocor
  • Topiramate • Topamax
  • Ziprasidone • Geodon
  • Zonisamide • Zonegran

Disclosure

Dr. Muzina receives research grants from AstraZeneca Pharmaceuticals, Eli Lilly and Co., and Abbott Laboratories, is a consultant to AstraZeneca Pharmaceuticals and Pfizer, Inc., and a speaker for AstraZeneca Pharmaceuticals, Pfizer Inc., Eli Lilly and Co., and GlaxoSmithKline.

References

1. Hirschfeld RM, Bowden CL, Gitlin MJ, et al. Practice guideline for the treatment of patients with bipolar disorder (rev). Am J Psychiatry 2002;159:1-50.

2. Bauer MS, Mitchner L. What is a “mood stabilizer?” An evidence-based response. Am J Psychiatry 2004;161(1):3-18.

3. Keck PE, Jr, McElroy SL, Strakowski SM. Anticonvulsants and antipsychotics in the treatment of bipolar disorder. J Clin Psychiatry 1998;59(suppl 6):74-81.

4. Sharma V. Atypical antipsychotics and suicide in mood and anxiety disorders. Bipolar Disord 2003;5(suppl 2):48-52.

5. Tohen M, Marneros A, Bowden C, et al. Olanzapine versus lithium in relapse prevention in bipolar disorder: a randomized double-blind controlled 12-month clinical trial (presentation). Freiberg, Germany: Stanley Foundation Bipolar Network, Sept. 11-14, 2002.

6. Tohen M, Ketter TA, Zarate CA, et al. Olanzapine versus divalproex sodium for the treatment of acute mania and maintenance of remission: a 47-week study. Am J Psychiatry 2003;160(7):1263-71.

7. Vieta E, Goikolea JM, Corbella B, et al. Risperidone safety and efficacy in the treatment of bipolar and schizoaffective disorders: results from a 6-month, multicenter, open study. J Clin Psychiatry 2001;62(10):818-25.

8. Vieta E, Brugue E, Goikolea JM, et al. Acute and continuation risperidone monotherapy in mania. Hum Psychopharmacol 2004;19(1):41-5.

9. Sajatovic M, Brescan DW, Perez DE, et al. Quetiapine alone and added to a mood stabilizer for serious mood disorders. J Clin Psychiatry 2001;62(9):728-32.

10. Keck PE, Jr, Versiani M, Potkin S, et al. Ziprasidone in the treatment of acute bipolar mania: a three-week, placebo-controlled, double-blind, randomized trial. Am J Psychiatry 2003;160(4):741-8.

11. Keck PE, Jr, Marcus R, Tourkodimitris S, et al. A placebo-controlled, double-blind study of the efficacy and safety of aripiprazole in patients with acute bipolar mania. Am J Psychiatry 2003;160(9):1651-8.

12. Calabrese JR, Kimmel SE, Woyshville MJ, et al. Clozapine for treatment-refractory mania. Am J Psychiatry 1996;153(6):759-64.

13. Green AI, Tohen M, Patel JK, et al. Clozapine in the treatment of refractory psychotic mania. Am J Psychiatry 2000;157(6):982-6.

14. Zarate CA, Jr, Tohen M, Banov MD, et al. Is clozapine a mood stabilizer? J Clin Psychiatry 1995;56(3):108-12.

15. Suppes T, Webb A, Paul B, et al. Clinical outcome in a randomized 1-year trial of clozapine versus treatment as usual for patients with treatment-resistant illness and a history of mania. Am J Psychiatry 1999;156(8):1164-9.

16. Nasrallah HA, Churchill CM, Hamdan-Allan GA. Higher frequency of neuroleptic-induced dystonia in mania than in schizophrenia. Am J Psychiatry 1988;145(11):1455-6.

17. Chue P, Kovacs CS. Safety and tolerability of atypical antipsychotics in patients with bipolar disorder: prevalence, monitoring and management. Bipolar Disord 2003;5(suppl 2):62-79.

18. Simpson GM, Lindenmayer JP. Extrapyramidal symptoms in patients treated with risperidone. J Clin Psychopharmacol 1997;17(3):194-201.

19. Keck PE, Jr, McElroy SL. Bipolar disorder, obesity, and pharmacotherapy-associated weight gain. J Clin Psychiatry 2003;64(12):1426-35.

20. Fagiolini A, Frank E, Houck PR, et al. Prevalence of obesity and weight change during treatment in patients with bipolar I disorder. J Clin Psychiatry 2002;63(6):528-33.

21. Fagiolini A, Kupfer DJ, Houck PR, et al. Obesity as a correlate of outcome in patients with bipolar I disorder. Am J Psychiatry 2003;160(1):112-17.

22. Haupt DW, Newcomer JW. Abnormalities in glucose regulation associated with mental illness and treatment. J Psychosom Res 2002;53(4):925-33.

Author and Disclosure Information

David J. Muzina, MD
Director, Bipolar Disorders Clinic Department of psychiatry and psychology The Cleveland Clinic Foundation Cleveland, Ohio

Issue
Current Psychiatry - 03(04)
Publications
Topics
Page Number
14-30
Sections
Author and Disclosure Information

David J. Muzina, MD
Director, Bipolar Disorders Clinic Department of psychiatry and psychology The Cleveland Clinic Foundation Cleveland, Ohio

Author and Disclosure Information

David J. Muzina, MD
Director, Bipolar Disorders Clinic Department of psychiatry and psychology The Cleveland Clinic Foundation Cleveland, Ohio

Maintenance therapy with mood stabilizers is the most critical phase of bipolar disorder treatment but the stage with the least available evidence about medication risks and benefits. The FDA’s recent approval of olanzapine for bipolar maintenance raises the question of whether atypical antipsychotics are really mood stabilizers. This article attempts to answer that question by:

  • describing the “ideal” mood stabilizer
  • discussing atypicals’ advantages over conventional antipsychotics in bipolar patients
  • comparing efficacy data for the six available atypicals
  • recommending strategies to prevent and treat atypicals’ potentially serious side effects during long-term therapy.

What is a ‘mood stabilizer’?

Successful mood stabilizer maintenance therapy decreases the time patients are sick and disabled. Although somewhat dated after only 2 years, the most recent American Psychiatric Association (APA) practice guidelines1 support using mood stabilizers for patients with bipolar I and bipolar II disorders.

Table 1

Bipolar maintenance treatment goals

  • Prevent manic or depressive relapse
  • Reduce subthreshold symptoms
  • Reduce suicide risk
  • Reduce cycling frequency
  • Reduce mood instability
  • Improve functioning
Adapted from American Psychiatric Association practice guidelines for treating patients with bipolar disorder (reference 1)

The goals of maintenance therapy are listed in Table 1. The ideal mood stabilizer would work in maintenance and all bipolar phases and treatment stages—from treating acute depression, mania, hypomania, and mixed states to preventing abnormal mood elevations and depressions. It would not precipitate depression or mania, rapid cycling, or cycle acceleration.

In other words, the best “mood stabilizer” would work in all four treatment roles of bipolar disorder: treating highs and lows, and preventing highs and lows. No such mood stabilizer exists, although lithium may come closest to the ideal.2

Most U.S. psychiatrists use combination therapies for bipolar disorder, particularly when treating acute manic states. The most common combination is a “known” mood stabilizer—such as lithium or divalproex—plus an antipsychotic to quickly control mania.

After mania remits, clinicians often try to eliminate the antipsychotic in hopes of maintaining mood stability and euthymia with the mood stabilizer alone. This was especially true before atypical antipsychotics were approved, given the risk for tardive dyskinesia (TD) associated with long-term use of conventional antipsychotics.

Unfortunately, patients frequently relapse with this strategy, so psychiatrists may leave their bipolar patients on atypical antipsychotics during long-term maintenance. But how good are atypicals as mood stabilizers? Perhaps more importantly, how safe is long-term use of atypicals in bipolar patients?

Antipsychotics as mood stabilizers

The 2002 APA practice guidelines discuss efficacy data for using lithium, divalproex or valproate, lamotrigine, carbamazepine, and electroconvulsive therapy for bipolar maintenance treatment. Two sentences on antipsychotic drug use note:

  • one placebo-controlled study of a conventional antipsychotic showing no efficacy
  • some data supporting clozapine as a prophylactic bipolar treatment.1

A 1998 review of five open trials3 touched on conventional depot antipsychotics’ value in reducing manic or affective illness. However, the authors warned:

  • no controlled trials existed
  • maintenance antipsychotic treatment may be associated with increased risk for tardive movement disorders
  • conventional agents can exacerbate depressive symptoms in some patients.

Using conventional antipsychotics long-term in bipolar disorder is not advisable, with the possible exception of depot preparations in nonadhering patients with severe illness. Long-acting injectable atypicals—such as the recently approved IM risperidone—may displace any use of conventional antipsychotics in bipolar patients.

Atypical antipsychotics hold several advantages over conventional agents:

  • significantly reduced risk for TD and extrapyramidal symptoms (EPS)
  • lack of serum prolactin elevation (except with risperidone)
  • improved cognition
  • possible decreased suicidality, particularly with clozapine.4

Table 2

Tips for managing atypicals’ potentially serious side-effect risks

Weight gain/obesity
AssessmentPreventionTreatment
Evaluate comorbid conditions such as eating disorders or substance abuse
Take nutritional and exercise history
Check weight and waist circumference at baseline and every visit
Calculate body mass index at every visit
Prescribe healthy diet and exercise
Patient education, careful monitoring, and prevention are most-effective treatments
Drug therapy for persistent weight gain or early rapid gain (>7% in first 6 months). Agents of potential benefit include topiramate, sibutramine, metformin, zonisamide, and orlistat (see Table 3)
Glucose control/type 2 diabetes
AssessmentPreventionTreatment
Take history of glucose intolerance or diabetes
Ask about family history of diabetes, obesity, hypertension, heart disease
Check baseline weight and plasma glucose
Obtain fasting plasma glucose every 3 months for first year, then annually
Prescribe healthy diet and exercise
Primary prevention through careful monitoring is most effective
Discontinue atypical antipsychotic; use other mood stabilizer unless atypical is only effective drug for that patient
Oral hypoglycemics (metformin, others)
Hyperlipidemia
AssessmentPreventionTreatment
Take history of hyperlipidemia or cardiovascular disease
Ask about family history of hyperlipidemia
Check fasting lipid profile including triglycerides at baseline and every 3 months in first year
Prescribe healthy diet and exercise
Monitor diet, exercise, weight, lipids regularly
Change atypical antipsychotic or use other mood stabilizer (as described above)
Oral antilipemics (simvastatin, others)
 

 

Evidence for atypicals

Olanzapine is the only atypical FDA-approved for relapse prevention in bipolar disorder. This approval is supported by several studies, most notably two 1-year, double-blind trials:

  • Mean time to any mood relapse was 174 days in patients taking olanzapine, mean 12.5 mg/d (±5 mg), compared with 22 days in a placebo group (Eli Lilly and Co., data on file).
  • Manic relapse rate was 14.3% in patients treated with olanzapine, ~12 mg/d, compared with 28% in patients treated with lithium, ~1,100 mg/d (mean 0.76 mEq/L). The two treatments were similarly effective in preventing depressive relapse.5

As a mood stabilizer, olanzapine was as effective as divalproex in a 47-week randomized, double-blind study of 251 adults with bipolar I disorder.6 Patients treated with olanzapine improved more rapidly and had fewer manic symptoms than those treated with divalproex, but bipolar relapse rates were similar in both treatment groups.

Risperidone appears to have a role as a potential maintenance mood stabilizer in bipolar patients, although double-blind trials are lacking.

In a 6-month, open-label investigation, relapse rates were 16% for depression and 7% for mania in bipolar patients receiving risperidone (average 4 mg/d) combined with mood-stabilizing medications.7 These relapse rates are lower than those typically reported for mood-stabilizing monotherapy.

In another 6-month, open-label study, risperidone monotherapy (average 4 mg/d) was effective for treating mania and maintaining euthymia.8

IM risperidone is a useful option for bipolar patients chronically nonadherent with oral medications; it also substantially reduces the risk of neuroleptic side effects compared with older depot antipsychotics.

Quetiapine was recently approved as an antimanic agent and may possess mood-stabilizing properties. In a preliminary study of 10 patients with bipolar disorder, adding quetiapine (mean 200 mg/d) to existing mood stabilizer therapy for 12 weeks improved psychopathology, mania, and depression rating scale scores.9

More-recent unpublished data suggest dosing quetiapine to approximately 600 mg/d as monotherapy or an adjunct to treat acute mania, though controlled maintenance studies are lacking (AstraZeneca Pharmaceuticals, data on file).

Others. Some early evidence supports using ziprasidone and aripiprazole for bipolar mania:

  • Ziprasidone monotherapy, 40 to 80 mg bid, was significantly more effective than placebo in reducing acute mania symptoms in a 3-week, double-blind, randomized trial of 197 patients with bipolar I disorder.10
  • Aripiprazole monotherapy, 15 to 30 mg/d, had a significantly greater effect than placebo in a 3-week, double-blind, randomized trial of 262 patients in acute manic or mixed bipolar episodes. Response rates among patients with mania were 40% with aripiprazole and 19% with placebo.11

Both ziprasidone and aripiprazole were well-tolerated in these brief trials, although their efficacy as long-term mood-stabilizers in bipolar disorder is unclear.

Using clozapine raises concerns about potentially serious adverse events, although it remains the only agent with proven efficacy in treatment-refractory mania.12,13 Clozapine also appears to reduce hospitalization and affective relapse rates and improve symptoms and quality of life.14,15

Long-term safety

Compared with conventional antipsychotics, EPS are not a major concern with the atypical agents. Except for risperidone, atypicals’ effect on prolactin levels generally is not clinically meaningful. Atypicals appear to be “mood-friendly,” whereas conventional antipsychotics seem to contribute to dysphoria or cause depression in some patients.

Sedation or other annoying side effects such as dry mouth or dizziness can occur with any atypical. Other more-serious side effects may complicate antipsychotic treatment, as we are coming to understand from using atypicals for long-term schizophrenia management.

Table 3

Weight-loss medications for bipolar patients taking atypical antipsychotics

DrugDosageSide effectsRecommendations
Metformin500 to 1,000 mg bidHypoglycemia
Diarrhea
Nausea/vomiting
First-line in patients with comorbid type 2 diabetes
Orlistat120 mg tidGI distress
Change in bowel
habits
Second-line
For patients with BMI >27
Supplement fat-soluble vitamins
Sibutramine5 to 15 mg/dDry mouth
Anorexia
Insomnia
Constipation
Second-line
For patients with BMI 27 to 30
Risk of serotonin syndrome if given with serotonergic drugs
Topiramate50 to 250 mg/dSomnolence
Fatigue
Paresthesias
Consider first-line for its potential additive mood-stabilizing effect
May help comorbid binge-eating or seizure disorders
Zonisamide100 to 600 mg/dSomnolence
Dizziness
Anorexia
Consider first-line for its potential additive mood-stabilizing effect
May help comorbid binge-eating or seizure disorders

Movement disorders. Antipsychotics appear more likely to cause EPS in patients with mood disorders than with schizophrenia. In one study using conventional antipsychotics, bipolar patients were 4 to 5 times more likely than schizophrenia patients to experience acute dystonia.16

Although atypicals pose some small risk for acute EPS and TD, the risk is near placebo-level with clinically relevant and comparable dosages.17 Even so, it is important to educate patients to watch for emerging signs of TD during long-term treatment with any antipsychotic. EPS risk may be dose-dependent, particularly with risperidone.18

Weight gain and obesity. Patients with bipolar disorder are more likely to be overweight or obese (body mass index [BMI] > 30) than the general population,17,19 though the reasons are unknown. Studies suggest an obesity prevalence of 32% to 35% in bipolar patients, compared with 18% in the general population.20,21

 

 

All atypicals can cause weight gain, although olanzapine and clozapine are associated with the greatest mean weight gains. In three long-term trials (47 weeks to 18 months), bipolar patients who received olanzapine gained significantly more weight (mean 2 to 3 kg) than those receiving lithium or divalproex.19

Cases with much greater weight gain—even leading to clinical obesity—have been observed, particularly with olanzapine. Although evidence from registration trials and clinical experience show lesser weight gains with risperidone, quetiapine, ziprasidone, and aripiprazole, some of our patients do gain weight while taking these agents—either alone or in combination with lithium or divalproex.

Weight management. Because patients with bipolar disorder may be at increased risk for weight gain and obesity, weight management techniques may improve their health by:

  • decreasing morbidity and mortality tied to weight-related physical illnesses
  • enhancing psychological well-being.1

In addition to diet and exercise counseling, some bipolar patients taking long-term atypical antipsychotics may benefit from adjunctive weight-loss medications (Table 3). We generally use such medications for bipolar patients who:

  • persistently gain weight despite best dietary practices
  • gain substantial weight early in treatment with an atypical antipsychotic that is providing effective symptomatic relief.

Early weight gain—particularly gains of >7% within the first 6 weeks—might predict large weight gain over time.

Diabetes. In September 2003, the FDA requested a class-wide labeling change to warn about a possible link between atypical antipsychotics and diabetes. The FDA recommended blood sugar monitoring of patients taking atypicals, especially those with obesity risk factors or family history of diabetes.

Type 2 diabetes develops in some patients taking atypicals, whether or not they gain substantial weight.22 This suggests that weight gain associated with bipolar disorder and the use of atypical antipsychotics may be independent risk factors for diabetes—a clear concern when treating bipolar patients.

Evidence provides no clear answer as to which atypicals may increase diabetes risk. Cautious use and vigilant monitoring of blood glucose are therefore recommended for every patient taking an atypical for long-term therapy. Also watch for increases in triglycerides and cholesterol17 in patients taking atypicals as bipolar maintenance therapy.

Conclusion

Atypical antipsychotics are valuable therapies in preventing bipolar relapses, although olanzapine is the only atypical with this indication so far. Collective data and clinical experience suggest that atypicals are indeed mood stabilizers, although—like other mood stabilizers such as lithium or divalproex—they have limitations. None achieve ideal efficacy in all four bipolar treatment roles: treating the highs and lows, and preventing the highs and lows. Atypicals seem more effective in treating and preventing the highs than the lows, reminding us that effective depression treatment is the greatest unmet need in bipolar disorder.

More double-blind, randomized, controlled trials are needed to fully understand whether all atypicals are mood stabilizers and to determine their safety and side effects in long-term therapy for patients with bipolar disorder.

Related resources

  • Depression and Bipolar Support Alliance. www.dbsalliance.org
  • Muzina DJ, Calabrese JR. Guidelines for treatment of bipolar disorder.In: Stein DJ, Kupfer DJ, Schatzberg AF (eds). Textbook of mood disorders Washington, DC: American Psychiatric Publishing, 2004 (in press).

Drug brand names

  • Aripiprazole • Abilify
  • Carbamazepine • Tegretol
  • Clozapine • Clozaril
  • Divalproex/valproate • Depakote, Depakene
  • Lamotrigine • Lamictal
  • Lithium • Eskalith, Lithobid, et al
  • Metformin • Glucophage
  • Olanzapine • Zyprexa
  • Orlistat • Xenical
  • Quetiapine • Seroquel
  • Risperidone • Risperdal, Risperdal Consta
  • Sibutramine • Meridia
  • Simvastatin • Zocor
  • Topiramate • Topamax
  • Ziprasidone • Geodon
  • Zonisamide • Zonegran

Disclosure

Dr. Muzina receives research grants from AstraZeneca Pharmaceuticals, Eli Lilly and Co., and Abbott Laboratories, is a consultant to AstraZeneca Pharmaceuticals and Pfizer, Inc., and a speaker for AstraZeneca Pharmaceuticals, Pfizer Inc., Eli Lilly and Co., and GlaxoSmithKline.

Maintenance therapy with mood stabilizers is the most critical phase of bipolar disorder treatment but the stage with the least available evidence about medication risks and benefits. The FDA’s recent approval of olanzapine for bipolar maintenance raises the question of whether atypical antipsychotics are really mood stabilizers. This article attempts to answer that question by:

  • describing the “ideal” mood stabilizer
  • discussing atypicals’ advantages over conventional antipsychotics in bipolar patients
  • comparing efficacy data for the six available atypicals
  • recommending strategies to prevent and treat atypicals’ potentially serious side effects during long-term therapy.

What is a ‘mood stabilizer’?

Successful mood stabilizer maintenance therapy decreases the time patients are sick and disabled. Although somewhat dated after only 2 years, the most recent American Psychiatric Association (APA) practice guidelines1 support using mood stabilizers for patients with bipolar I and bipolar II disorders.

Table 1

Bipolar maintenance treatment goals

  • Prevent manic or depressive relapse
  • Reduce subthreshold symptoms
  • Reduce suicide risk
  • Reduce cycling frequency
  • Reduce mood instability
  • Improve functioning
Adapted from American Psychiatric Association practice guidelines for treating patients with bipolar disorder (reference 1)

The goals of maintenance therapy are listed in Table 1. The ideal mood stabilizer would work in maintenance and all bipolar phases and treatment stages—from treating acute depression, mania, hypomania, and mixed states to preventing abnormal mood elevations and depressions. It would not precipitate depression or mania, rapid cycling, or cycle acceleration.

In other words, the best “mood stabilizer” would work in all four treatment roles of bipolar disorder: treating highs and lows, and preventing highs and lows. No such mood stabilizer exists, although lithium may come closest to the ideal.2

Most U.S. psychiatrists use combination therapies for bipolar disorder, particularly when treating acute manic states. The most common combination is a “known” mood stabilizer—such as lithium or divalproex—plus an antipsychotic to quickly control mania.

After mania remits, clinicians often try to eliminate the antipsychotic in hopes of maintaining mood stability and euthymia with the mood stabilizer alone. This was especially true before atypical antipsychotics were approved, given the risk for tardive dyskinesia (TD) associated with long-term use of conventional antipsychotics.

Unfortunately, patients frequently relapse with this strategy, so psychiatrists may leave their bipolar patients on atypical antipsychotics during long-term maintenance. But how good are atypicals as mood stabilizers? Perhaps more importantly, how safe is long-term use of atypicals in bipolar patients?

Antipsychotics as mood stabilizers

The 2002 APA practice guidelines discuss efficacy data for using lithium, divalproex or valproate, lamotrigine, carbamazepine, and electroconvulsive therapy for bipolar maintenance treatment. Two sentences on antipsychotic drug use note:

  • one placebo-controlled study of a conventional antipsychotic showing no efficacy
  • some data supporting clozapine as a prophylactic bipolar treatment.1

A 1998 review of five open trials3 touched on conventional depot antipsychotics’ value in reducing manic or affective illness. However, the authors warned:

  • no controlled trials existed
  • maintenance antipsychotic treatment may be associated with increased risk for tardive movement disorders
  • conventional agents can exacerbate depressive symptoms in some patients.

Using conventional antipsychotics long-term in bipolar disorder is not advisable, with the possible exception of depot preparations in nonadhering patients with severe illness. Long-acting injectable atypicals—such as the recently approved IM risperidone—may displace any use of conventional antipsychotics in bipolar patients.

Atypical antipsychotics hold several advantages over conventional agents:

  • significantly reduced risk for TD and extrapyramidal symptoms (EPS)
  • lack of serum prolactin elevation (except with risperidone)
  • improved cognition
  • possible decreased suicidality, particularly with clozapine.4

Table 2

Tips for managing atypicals’ potentially serious side-effect risks

Weight gain/obesity
AssessmentPreventionTreatment
Evaluate comorbid conditions such as eating disorders or substance abuse
Take nutritional and exercise history
Check weight and waist circumference at baseline and every visit
Calculate body mass index at every visit
Prescribe healthy diet and exercise
Patient education, careful monitoring, and prevention are most-effective treatments
Drug therapy for persistent weight gain or early rapid gain (>7% in first 6 months). Agents of potential benefit include topiramate, sibutramine, metformin, zonisamide, and orlistat (see Table 3)
Glucose control/type 2 diabetes
AssessmentPreventionTreatment
Take history of glucose intolerance or diabetes
Ask about family history of diabetes, obesity, hypertension, heart disease
Check baseline weight and plasma glucose
Obtain fasting plasma glucose every 3 months for first year, then annually
Prescribe healthy diet and exercise
Primary prevention through careful monitoring is most effective
Discontinue atypical antipsychotic; use other mood stabilizer unless atypical is only effective drug for that patient
Oral hypoglycemics (metformin, others)
Hyperlipidemia
AssessmentPreventionTreatment
Take history of hyperlipidemia or cardiovascular disease
Ask about family history of hyperlipidemia
Check fasting lipid profile including triglycerides at baseline and every 3 months in first year
Prescribe healthy diet and exercise
Monitor diet, exercise, weight, lipids regularly
Change atypical antipsychotic or use other mood stabilizer (as described above)
Oral antilipemics (simvastatin, others)
 

 

Evidence for atypicals

Olanzapine is the only atypical FDA-approved for relapse prevention in bipolar disorder. This approval is supported by several studies, most notably two 1-year, double-blind trials:

  • Mean time to any mood relapse was 174 days in patients taking olanzapine, mean 12.5 mg/d (±5 mg), compared with 22 days in a placebo group (Eli Lilly and Co., data on file).
  • Manic relapse rate was 14.3% in patients treated with olanzapine, ~12 mg/d, compared with 28% in patients treated with lithium, ~1,100 mg/d (mean 0.76 mEq/L). The two treatments were similarly effective in preventing depressive relapse.5

As a mood stabilizer, olanzapine was as effective as divalproex in a 47-week randomized, double-blind study of 251 adults with bipolar I disorder.6 Patients treated with olanzapine improved more rapidly and had fewer manic symptoms than those treated with divalproex, but bipolar relapse rates were similar in both treatment groups.

Risperidone appears to have a role as a potential maintenance mood stabilizer in bipolar patients, although double-blind trials are lacking.

In a 6-month, open-label investigation, relapse rates were 16% for depression and 7% for mania in bipolar patients receiving risperidone (average 4 mg/d) combined with mood-stabilizing medications.7 These relapse rates are lower than those typically reported for mood-stabilizing monotherapy.

In another 6-month, open-label study, risperidone monotherapy (average 4 mg/d) was effective for treating mania and maintaining euthymia.8

IM risperidone is a useful option for bipolar patients chronically nonadherent with oral medications; it also substantially reduces the risk of neuroleptic side effects compared with older depot antipsychotics.

Quetiapine was recently approved as an antimanic agent and may possess mood-stabilizing properties. In a preliminary study of 10 patients with bipolar disorder, adding quetiapine (mean 200 mg/d) to existing mood stabilizer therapy for 12 weeks improved psychopathology, mania, and depression rating scale scores.9

More-recent unpublished data suggest dosing quetiapine to approximately 600 mg/d as monotherapy or an adjunct to treat acute mania, though controlled maintenance studies are lacking (AstraZeneca Pharmaceuticals, data on file).

Others. Some early evidence supports using ziprasidone and aripiprazole for bipolar mania:

  • Ziprasidone monotherapy, 40 to 80 mg bid, was significantly more effective than placebo in reducing acute mania symptoms in a 3-week, double-blind, randomized trial of 197 patients with bipolar I disorder.10
  • Aripiprazole monotherapy, 15 to 30 mg/d, had a significantly greater effect than placebo in a 3-week, double-blind, randomized trial of 262 patients in acute manic or mixed bipolar episodes. Response rates among patients with mania were 40% with aripiprazole and 19% with placebo.11

Both ziprasidone and aripiprazole were well-tolerated in these brief trials, although their efficacy as long-term mood-stabilizers in bipolar disorder is unclear.

Using clozapine raises concerns about potentially serious adverse events, although it remains the only agent with proven efficacy in treatment-refractory mania.12,13 Clozapine also appears to reduce hospitalization and affective relapse rates and improve symptoms and quality of life.14,15

Long-term safety

Compared with conventional antipsychotics, EPS are not a major concern with the atypical agents. Except for risperidone, atypicals’ effect on prolactin levels generally is not clinically meaningful. Atypicals appear to be “mood-friendly,” whereas conventional antipsychotics seem to contribute to dysphoria or cause depression in some patients.

Sedation or other annoying side effects such as dry mouth or dizziness can occur with any atypical. Other more-serious side effects may complicate antipsychotic treatment, as we are coming to understand from using atypicals for long-term schizophrenia management.

Table 3

Weight-loss medications for bipolar patients taking atypical antipsychotics

DrugDosageSide effectsRecommendations
Metformin500 to 1,000 mg bidHypoglycemia
Diarrhea
Nausea/vomiting
First-line in patients with comorbid type 2 diabetes
Orlistat120 mg tidGI distress
Change in bowel
habits
Second-line
For patients with BMI >27
Supplement fat-soluble vitamins
Sibutramine5 to 15 mg/dDry mouth
Anorexia
Insomnia
Constipation
Second-line
For patients with BMI 27 to 30
Risk of serotonin syndrome if given with serotonergic drugs
Topiramate50 to 250 mg/dSomnolence
Fatigue
Paresthesias
Consider first-line for its potential additive mood-stabilizing effect
May help comorbid binge-eating or seizure disorders
Zonisamide100 to 600 mg/dSomnolence
Dizziness
Anorexia
Consider first-line for its potential additive mood-stabilizing effect
May help comorbid binge-eating or seizure disorders

Movement disorders. Antipsychotics appear more likely to cause EPS in patients with mood disorders than with schizophrenia. In one study using conventional antipsychotics, bipolar patients were 4 to 5 times more likely than schizophrenia patients to experience acute dystonia.16

Although atypicals pose some small risk for acute EPS and TD, the risk is near placebo-level with clinically relevant and comparable dosages.17 Even so, it is important to educate patients to watch for emerging signs of TD during long-term treatment with any antipsychotic. EPS risk may be dose-dependent, particularly with risperidone.18

Weight gain and obesity. Patients with bipolar disorder are more likely to be overweight or obese (body mass index [BMI] > 30) than the general population,17,19 though the reasons are unknown. Studies suggest an obesity prevalence of 32% to 35% in bipolar patients, compared with 18% in the general population.20,21

 

 

All atypicals can cause weight gain, although olanzapine and clozapine are associated with the greatest mean weight gains. In three long-term trials (47 weeks to 18 months), bipolar patients who received olanzapine gained significantly more weight (mean 2 to 3 kg) than those receiving lithium or divalproex.19

Cases with much greater weight gain—even leading to clinical obesity—have been observed, particularly with olanzapine. Although evidence from registration trials and clinical experience show lesser weight gains with risperidone, quetiapine, ziprasidone, and aripiprazole, some of our patients do gain weight while taking these agents—either alone or in combination with lithium or divalproex.

Weight management. Because patients with bipolar disorder may be at increased risk for weight gain and obesity, weight management techniques may improve their health by:

  • decreasing morbidity and mortality tied to weight-related physical illnesses
  • enhancing psychological well-being.1

In addition to diet and exercise counseling, some bipolar patients taking long-term atypical antipsychotics may benefit from adjunctive weight-loss medications (Table 3). We generally use such medications for bipolar patients who:

  • persistently gain weight despite best dietary practices
  • gain substantial weight early in treatment with an atypical antipsychotic that is providing effective symptomatic relief.

Early weight gain—particularly gains of >7% within the first 6 weeks—might predict large weight gain over time.

Diabetes. In September 2003, the FDA requested a class-wide labeling change to warn about a possible link between atypical antipsychotics and diabetes. The FDA recommended blood sugar monitoring of patients taking atypicals, especially those with obesity risk factors or family history of diabetes.

Type 2 diabetes develops in some patients taking atypicals, whether or not they gain substantial weight.22 This suggests that weight gain associated with bipolar disorder and the use of atypical antipsychotics may be independent risk factors for diabetes—a clear concern when treating bipolar patients.

Evidence provides no clear answer as to which atypicals may increase diabetes risk. Cautious use and vigilant monitoring of blood glucose are therefore recommended for every patient taking an atypical for long-term therapy. Also watch for increases in triglycerides and cholesterol17 in patients taking atypicals as bipolar maintenance therapy.

Conclusion

Atypical antipsychotics are valuable therapies in preventing bipolar relapses, although olanzapine is the only atypical with this indication so far. Collective data and clinical experience suggest that atypicals are indeed mood stabilizers, although—like other mood stabilizers such as lithium or divalproex—they have limitations. None achieve ideal efficacy in all four bipolar treatment roles: treating the highs and lows, and preventing the highs and lows. Atypicals seem more effective in treating and preventing the highs than the lows, reminding us that effective depression treatment is the greatest unmet need in bipolar disorder.

More double-blind, randomized, controlled trials are needed to fully understand whether all atypicals are mood stabilizers and to determine their safety and side effects in long-term therapy for patients with bipolar disorder.

Related resources

  • Depression and Bipolar Support Alliance. www.dbsalliance.org
  • Muzina DJ, Calabrese JR. Guidelines for treatment of bipolar disorder.In: Stein DJ, Kupfer DJ, Schatzberg AF (eds). Textbook of mood disorders Washington, DC: American Psychiatric Publishing, 2004 (in press).

Drug brand names

  • Aripiprazole • Abilify
  • Carbamazepine • Tegretol
  • Clozapine • Clozaril
  • Divalproex/valproate • Depakote, Depakene
  • Lamotrigine • Lamictal
  • Lithium • Eskalith, Lithobid, et al
  • Metformin • Glucophage
  • Olanzapine • Zyprexa
  • Orlistat • Xenical
  • Quetiapine • Seroquel
  • Risperidone • Risperdal, Risperdal Consta
  • Sibutramine • Meridia
  • Simvastatin • Zocor
  • Topiramate • Topamax
  • Ziprasidone • Geodon
  • Zonisamide • Zonegran

Disclosure

Dr. Muzina receives research grants from AstraZeneca Pharmaceuticals, Eli Lilly and Co., and Abbott Laboratories, is a consultant to AstraZeneca Pharmaceuticals and Pfizer, Inc., and a speaker for AstraZeneca Pharmaceuticals, Pfizer Inc., Eli Lilly and Co., and GlaxoSmithKline.

References

1. Hirschfeld RM, Bowden CL, Gitlin MJ, et al. Practice guideline for the treatment of patients with bipolar disorder (rev). Am J Psychiatry 2002;159:1-50.

2. Bauer MS, Mitchner L. What is a “mood stabilizer?” An evidence-based response. Am J Psychiatry 2004;161(1):3-18.

3. Keck PE, Jr, McElroy SL, Strakowski SM. Anticonvulsants and antipsychotics in the treatment of bipolar disorder. J Clin Psychiatry 1998;59(suppl 6):74-81.

4. Sharma V. Atypical antipsychotics and suicide in mood and anxiety disorders. Bipolar Disord 2003;5(suppl 2):48-52.

5. Tohen M, Marneros A, Bowden C, et al. Olanzapine versus lithium in relapse prevention in bipolar disorder: a randomized double-blind controlled 12-month clinical trial (presentation). Freiberg, Germany: Stanley Foundation Bipolar Network, Sept. 11-14, 2002.

6. Tohen M, Ketter TA, Zarate CA, et al. Olanzapine versus divalproex sodium for the treatment of acute mania and maintenance of remission: a 47-week study. Am J Psychiatry 2003;160(7):1263-71.

7. Vieta E, Goikolea JM, Corbella B, et al. Risperidone safety and efficacy in the treatment of bipolar and schizoaffective disorders: results from a 6-month, multicenter, open study. J Clin Psychiatry 2001;62(10):818-25.

8. Vieta E, Brugue E, Goikolea JM, et al. Acute and continuation risperidone monotherapy in mania. Hum Psychopharmacol 2004;19(1):41-5.

9. Sajatovic M, Brescan DW, Perez DE, et al. Quetiapine alone and added to a mood stabilizer for serious mood disorders. J Clin Psychiatry 2001;62(9):728-32.

10. Keck PE, Jr, Versiani M, Potkin S, et al. Ziprasidone in the treatment of acute bipolar mania: a three-week, placebo-controlled, double-blind, randomized trial. Am J Psychiatry 2003;160(4):741-8.

11. Keck PE, Jr, Marcus R, Tourkodimitris S, et al. A placebo-controlled, double-blind study of the efficacy and safety of aripiprazole in patients with acute bipolar mania. Am J Psychiatry 2003;160(9):1651-8.

12. Calabrese JR, Kimmel SE, Woyshville MJ, et al. Clozapine for treatment-refractory mania. Am J Psychiatry 1996;153(6):759-64.

13. Green AI, Tohen M, Patel JK, et al. Clozapine in the treatment of refractory psychotic mania. Am J Psychiatry 2000;157(6):982-6.

14. Zarate CA, Jr, Tohen M, Banov MD, et al. Is clozapine a mood stabilizer? J Clin Psychiatry 1995;56(3):108-12.

15. Suppes T, Webb A, Paul B, et al. Clinical outcome in a randomized 1-year trial of clozapine versus treatment as usual for patients with treatment-resistant illness and a history of mania. Am J Psychiatry 1999;156(8):1164-9.

16. Nasrallah HA, Churchill CM, Hamdan-Allan GA. Higher frequency of neuroleptic-induced dystonia in mania than in schizophrenia. Am J Psychiatry 1988;145(11):1455-6.

17. Chue P, Kovacs CS. Safety and tolerability of atypical antipsychotics in patients with bipolar disorder: prevalence, monitoring and management. Bipolar Disord 2003;5(suppl 2):62-79.

18. Simpson GM, Lindenmayer JP. Extrapyramidal symptoms in patients treated with risperidone. J Clin Psychopharmacol 1997;17(3):194-201.

19. Keck PE, Jr, McElroy SL. Bipolar disorder, obesity, and pharmacotherapy-associated weight gain. J Clin Psychiatry 2003;64(12):1426-35.

20. Fagiolini A, Frank E, Houck PR, et al. Prevalence of obesity and weight change during treatment in patients with bipolar I disorder. J Clin Psychiatry 2002;63(6):528-33.

21. Fagiolini A, Kupfer DJ, Houck PR, et al. Obesity as a correlate of outcome in patients with bipolar I disorder. Am J Psychiatry 2003;160(1):112-17.

22. Haupt DW, Newcomer JW. Abnormalities in glucose regulation associated with mental illness and treatment. J Psychosom Res 2002;53(4):925-33.

References

1. Hirschfeld RM, Bowden CL, Gitlin MJ, et al. Practice guideline for the treatment of patients with bipolar disorder (rev). Am J Psychiatry 2002;159:1-50.

2. Bauer MS, Mitchner L. What is a “mood stabilizer?” An evidence-based response. Am J Psychiatry 2004;161(1):3-18.

3. Keck PE, Jr, McElroy SL, Strakowski SM. Anticonvulsants and antipsychotics in the treatment of bipolar disorder. J Clin Psychiatry 1998;59(suppl 6):74-81.

4. Sharma V. Atypical antipsychotics and suicide in mood and anxiety disorders. Bipolar Disord 2003;5(suppl 2):48-52.

5. Tohen M, Marneros A, Bowden C, et al. Olanzapine versus lithium in relapse prevention in bipolar disorder: a randomized double-blind controlled 12-month clinical trial (presentation). Freiberg, Germany: Stanley Foundation Bipolar Network, Sept. 11-14, 2002.

6. Tohen M, Ketter TA, Zarate CA, et al. Olanzapine versus divalproex sodium for the treatment of acute mania and maintenance of remission: a 47-week study. Am J Psychiatry 2003;160(7):1263-71.

7. Vieta E, Goikolea JM, Corbella B, et al. Risperidone safety and efficacy in the treatment of bipolar and schizoaffective disorders: results from a 6-month, multicenter, open study. J Clin Psychiatry 2001;62(10):818-25.

8. Vieta E, Brugue E, Goikolea JM, et al. Acute and continuation risperidone monotherapy in mania. Hum Psychopharmacol 2004;19(1):41-5.

9. Sajatovic M, Brescan DW, Perez DE, et al. Quetiapine alone and added to a mood stabilizer for serious mood disorders. J Clin Psychiatry 2001;62(9):728-32.

10. Keck PE, Jr, Versiani M, Potkin S, et al. Ziprasidone in the treatment of acute bipolar mania: a three-week, placebo-controlled, double-blind, randomized trial. Am J Psychiatry 2003;160(4):741-8.

11. Keck PE, Jr, Marcus R, Tourkodimitris S, et al. A placebo-controlled, double-blind study of the efficacy and safety of aripiprazole in patients with acute bipolar mania. Am J Psychiatry 2003;160(9):1651-8.

12. Calabrese JR, Kimmel SE, Woyshville MJ, et al. Clozapine for treatment-refractory mania. Am J Psychiatry 1996;153(6):759-64.

13. Green AI, Tohen M, Patel JK, et al. Clozapine in the treatment of refractory psychotic mania. Am J Psychiatry 2000;157(6):982-6.

14. Zarate CA, Jr, Tohen M, Banov MD, et al. Is clozapine a mood stabilizer? J Clin Psychiatry 1995;56(3):108-12.

15. Suppes T, Webb A, Paul B, et al. Clinical outcome in a randomized 1-year trial of clozapine versus treatment as usual for patients with treatment-resistant illness and a history of mania. Am J Psychiatry 1999;156(8):1164-9.

16. Nasrallah HA, Churchill CM, Hamdan-Allan GA. Higher frequency of neuroleptic-induced dystonia in mania than in schizophrenia. Am J Psychiatry 1988;145(11):1455-6.

17. Chue P, Kovacs CS. Safety and tolerability of atypical antipsychotics in patients with bipolar disorder: prevalence, monitoring and management. Bipolar Disord 2003;5(suppl 2):62-79.

18. Simpson GM, Lindenmayer JP. Extrapyramidal symptoms in patients treated with risperidone. J Clin Psychopharmacol 1997;17(3):194-201.

19. Keck PE, Jr, McElroy SL. Bipolar disorder, obesity, and pharmacotherapy-associated weight gain. J Clin Psychiatry 2003;64(12):1426-35.

20. Fagiolini A, Frank E, Houck PR, et al. Prevalence of obesity and weight change during treatment in patients with bipolar I disorder. J Clin Psychiatry 2002;63(6):528-33.

21. Fagiolini A, Kupfer DJ, Houck PR, et al. Obesity as a correlate of outcome in patients with bipolar I disorder. Am J Psychiatry 2003;160(1):112-17.

22. Haupt DW, Newcomer JW. Abnormalities in glucose regulation associated with mental illness and treatment. J Psychosom Res 2002;53(4):925-33.

Issue
Current Psychiatry - 03(04)
Issue
Current Psychiatry - 03(04)
Page Number
14-30
Page Number
14-30
Publications
Publications
Topics
Article Type
Display Headline
Bipolar maintenance: Are atypical antipsychotics really ‘mood stabilizers’?
Display Headline
Bipolar maintenance: Are atypical antipsychotics really ‘mood stabilizers’?
Sections
Article Source

PURLs Copyright

Inside the Article

How to reduce aggression in youths with conduct disorder

Article Type
Changed
Tue, 12/11/2018 - 15:10
Display Headline
How to reduce aggression in youths with conduct disorder

Families and schools often pressure clinicians to “do something” when children or adolescents persistently bully, threaten, or injure others. This demand poses a treatment dilemma when psychosocial and educational interventions have failed to manage pediatric aggression.

Aggression is the main reason for drug therapy in youths with conduct disorder, but very little safety and efficacy data exist to help us choose medications. This places young patients at risk for polypharmacy, unmanaged symptoms, short-term side effects, and unknown long-term consequences of exposure to psychotropics.

Table 1

4 precautions when prescribing for pediatric aggression

  • Data to support polypharmacy are limited
  • Most drugs used to treat aggression are not FDA-approved for children
  • Drug treatment requires informed consent
  • Psychosocial treatment must be included in the treatment plan
Source: American Academy of Child and Adolescent Psychiatry1

This article reviews the limited data on using medications to reduce aggression in children and adolescents, focusing on double-blind, placebo-controlled trials in conduct disorder. Based on this evidence and our clinical experience, we offer a sample case and treatment recommendations.

Prescribing principles

Precautions. When prescribing drugs to treat aggressive youth, remember the American Academy of Child and Adolescent Psychiatry’s precautions (Table 1)1 Recently published recommendations prepared by expert consensus are also valuable treatment guides.2

Linking treatment to diagnosis. Should we attempt to manage aggression as a manifestation of an underlying psychiatric disorder? Or should we treat it the same across all disorders? The latter approach is akin to the “fever model.”

Fever—regardless of cause—may be treated with a nonsteroidal anti-inflammatory drug. However, evidence from drug studies suggests that underlying psychiatric disorders should help determine the choice of aggression treatment. For example, a recent study in adults found that divalproex was effective for aggressive patients only within a specific diagnostic subgroup (in this case, cluster B personality disorders).3

Clinical experience also links aggression treatment with underlying diagnoses. For example, aggression secondary to agitated depression is treated with an antidepressant, whereas aggression secondary to command hallucinations in schizophrenia is treated with antipsychotics.

In treating aggression in conduct disorder (Table 2), first treat comorbid disorders—such as attention deficit/hyperactivity disorder (ADHD) or bipolar disorder—and address the child’s psychosocial and educational needs. Then if medication is appropriate, consider drugs with evidence of safety and efficacy, such as antipsychotics, lithium, and stimulants.

Antipsychotics

Three conventional antipsychotics—chlorpromazine, haloperidol, and thioridazine—are FDA-approved for controlling disruptive behaviors in children.4 No atypical antipsychotics are so indicated, but atypicals are preferred in children and adolescents because of lower risks for tardive dyskinesia, neuroleptic malignant syndrome, and extrapyramidal symptoms.2

Risperidone is the most-studied atypical antipsychotic for treating pediatric aggression, particularly in patients with low intellectual functioning or mental retardation. In a 6-week, double-blind, placebo-controlled trial, 118 children ages 5 to 12 with severely disruptive behavior and IQs of 36 to 84 were given low-dose risperidone (mean 1.16 mg/d). Risperidone reduced conduct problems significantly more than placebo, although aggression was not measured directly.5 Adverse events included somnolence, headache, vomiting, weight gain, and elevated serum prolactin. Similar results have been reported in other studies.6

Table 2

Diagnostic criteria for conduct disorder

A. A repetitive and persistent pattern of behavior in which the basic rights of others or major age-appropriate societal norms or rules are violated, as manifested by the persistence of three (or more) of the following criteria in the past 12 months, with at least one criterion present in the past 6 months:
Aggression to people and animals
1. often bullies, threatens, or intimidates others5. has been physically cruel to animals
2. often initiates physical fights6. has stolen while confronting a victim (such as mugging, purse snatching, extortion, armed robbery)
3. has used a weapon that can cause serious physical harm to others (such as a bat, brick, broken bottle, knife, gun)7. has forced someone into sexual activity
4. has been physically cruel to people 
Destruction of property
8. has deliberately engaged in fire setting with the intention of causing serious damage9. has deliberately destroyed others’ property (other than by fire setting)
Deceitfulness or theft
10. has broken into someone else’s house, building, or car12. has stolen items of nontrivial value without confronting a victim (such as shoplifting without breaking and entering, or forgery)
11. often lies to obtain goods or favors or to avoid obligations(ie, “cons” others) 
Serious violation of rules
13. often stays out at night despite parental prohibitions, beginning before age 1315. has run away from home overnight at least twice while living in parental or parental surrogate home (or once without returning for a lengthy period)
14. is often truant from school, beginning before age 13 
B. The disturbance in behavior causes clinically significant impairment in social, academic, or occupational functioning
C. If the individual is age 18 or older, criteria are not met for antisocial personality disorder.
Specify severity:
Mild: few if any conduct problems in excess of those required to make the diagnosis and conduct problems cause only minor harm to others (such as lying, truancy, staying out after dark without permission)
Moderate: number of conduct problems and effect on others intermediate between “mild” and severe” (such as stealing without confronting a victim, vandalism)
Severe: many conduct problems in excess of those required to make the diagnosis or conduct problems cause considerable harm to others (such as forced sex, physical cruelty, use of a weapon, stealing while confronting a victim, breaking and entering)
Source: Reprinted with permission from the Diagnostic and statistical manual of mental disorders, 4th ed., text revision. Copyright 2000. American Psychiatric Association.
 

 

Box 1

Case report: Multiple diagnoses and drugs

JM, age 12, presented with his mother to address symptoms of hyperactivity and impulsive aggression. The boy also complained that his medications made him fall asleep during the day.

He is receiving five medications: a long-acting stimulant, atypical antipsychotic, anticonvulsant, alpha agonist, and selective serotonin reuptake inhibitor (SSRI). He had received numerous other medications, but prescription records are unavailable or incomplete.

Diagnostic history. Since age 5, JM has been diagnosed as having attention-deficit/hyperactivity disorder (ADHD), oppositional defiant disorder, conduct disorder, bipolar disorder, major depressive disorder, and learning disorders. On examination, the boy met DSM-IV criteria for ADHD, learning disorders, and conduct disorder (Table 2). He has a history of starting fights with peers, bullying, destroying property, lying, and stealing from stores and peers.

His mother stated that her son had always had irritable and labile periods, especially when he did not get his way. She was told during a previous psychiatric evaluation that the boy’s "mood swings" indicated bipolar disorder. On examination, however, he had no other bipolar symptoms, and his condition was chronic, not cyclic.

JM typically cries when he does not get his way, his mother reported, but he has no history of sleep or appetite changes that could suggest depression. He is happy when he can do as he pleases.

Reducing medications. After reviewing JM’s medications and performing the psychiatric assessment, the psychiatrist developed a plan to maximize his psychosocial and educational treatments and alter his medications and dosages. The first step was to increase the stimulant dosage to determine whether JM would be less hyperactive and impulsively aggressive.

The psychiatrist was concerned that the anticonvulsant, alpha agonist, and SSRI were not helping and could cause adverse events. He discussed slowly weaning these drugs one at a time with JM and his mother, and they agreed. The goal was to manage JM over time and to reduce his medications to one (ideally) or two (if necessary), possibly continuing the atypical antipsychotic.

Risperidone also reduced aggression in children with normal intelligence in one small study.7 As a cautionary note, however, long-term risperidone treatment has been associated with withdrawal dyskinesias.8

Olanzapine, quetiapine, ziprasidone, and aripiprazole are less well-studied for treating pediatric aggression but are preferable to conventional agents when antipsychotics are considered.

Recommendation. Expert consensus opinion2 recommends using atypicals when psychosocial treatments and first-line medications for primary conditions have failed. Start with low dosages, and titrate up slowly while monitoring symptoms and side effects. Because no studies have compared any atypical’s efficacy over others for aggressive behavior, base your choices on:

  • discussions with the patient and family (Box 1)
  • medical comorbidities
  • how the patient responded to antipsychotics in the past
  • side-effect profile
  • long-term treatment planning.2

If the patient cannot tolerate the medication or does not respond after 4 to 6 weeks, try switching atypicals. To improve partial response, consider adding a mood stabilizer such as lithium or divalproex. If aggressive symptoms remit for 6 months or longer, attempt to taper or discontinue the antipsychotic.2

Lithium

In placebo-controlled trials, lithium reduced aggression in:

  • male prisoners ages 16 to 24.9
  • children ages 7 and 12 with conduct disorder10
  • children and adolescents ages 10 to 17 with conduct disorder.11

Among these studies, only ours11 specifically measured aggression. We randomly assigned 40 children to receive 4 weeks of lithium, 900 to 2,100 mg/d (mean 1,425 ± 321 mg/d), or placebo. Serum lithium levels were 0.78 to 1.55 mEq/L (mean 1.07 ± 0.19 mEq/L). We used the Overt Aggression Scale (OAS)12,13 (see Related resources) to track frequency and severity of verbal aggression, aggression against objects, aggression against others, and self-aggression.

Lithium reduced aggression more than did placebo, as measured by the clinician-rated Clinical Global Impressions (CGI) scale and staff-rated Global Clinical Judgments (Consensus) Scale (GCJCS). The CGI showed a 70% response rate with lithium and 20% with placebo. Similarly, the GCJCS scale showed 80% response with lithium and 30% with placebo.

The aggression reduction with lithium was statistically significant and clinically evident. Most subjects (37 of 40) experienced at least one adverse event, however, whether receiving lithium or placebo. Nausea, vomiting, and urinary frequency were significantly more common in the lithium-treated group than with placebo. Fewer adverse events were reported in a similar outpatient study,14 probably because of less-frequent monitoring.

Lithium did not reduce aggression in adolescent girls treated for 2 weeks15 or in an outpatient study of children with ADHD.16

Recommendation. Lithium has shown efficacy for reducing severe aggression in hospitalized children with conduct disorder but not in similar outpatients. Consider this drug to reduce severe aggression in children with conduct disorder, especially if they have failed other treatments.

 

 

Anticonvulsants

Anticonvulsants have been used to decrease aggression for more than 50 years, and epidemiologic data show their use is increasing markedly.17 Few controlled studies support this prescribing trend, however.18

Initial reports suggested that anticonvulsants reduce disruptive behaviors, but more-critically designed studies have not supported this finding. For example, phenytoin sodium (diphenylhydantoin) demonstrated efficacy in open trials, but controlled trials found this anticonvulsant no more effective than placebo. In fact, placebo may have reduced aggression more than the active drug. Likewise, earlier controlled trials of carbamazepine indicated efficacy, but more-carefully designed trials using specific measures of aggression did not.

Divalproex is the anticonvulsant most commonly used for aggression in children and adolescents. Only one small, placebo-controlled study has found it effective in reducing aggression in children.19

Twenty children ages 10 to 18 with conduct disorder or oppositional defiant disorder were randomized to divalproex, 750 to 1,500 mg/d, or placebo. Eighteen completed the first phase, and 15 crossed over to the other treatment. Concomitant drug treatment, including stimulants, was allowed. The authors reported that 12 of 15 subjects showed some response to divalproex.

A 7-week study compared divalproex in high dosages (up to 1,500 mg/d) versus low dosages (up to 250 mg/d). This study was not placebo-controlled, but aggression was reduced more in the high-dosage than in the low-dosage group.20

Recommendation. If you use an anticonvulsant, first obtain informed consent from the patient and parent. Divalproex causes weight gain and has been associated with increased risk of polycystic ovary syndrome with masculinizing effects.21

Double-blind, placebo-controlled studies of divalproex and other anticonvulsants in treating aggression are needed, particularly as prescriptions for these agents are rising.

Stimulants

Some small controlled studies suggest that stimulants can reduce aggression in children with ADHD, but their effects on aggression in conduct disorder have not been well studied. Aggression was not measured directly in the National Institute of Mental Health Multimodal Treatment Study of Children with ADHD.21 Most other studies have been small and included children with ADHD but not necessarily conduct disorder.

Recommendation. Stimulants may help reduce aggression in children with ADHD, but studies gauging their effects in conduct disorder are needed.

Alpha agonists

Alpha agonists such as clonidine and guanfacine are increasingly being used to treat children with disruptive disorders, despite limited evidence. The small controlled studies that examined alpha agonists as monotherapy or add-ons in this population did not directly measure aggression.22,23

Recommendation. Little data support alpha agonists for reducing aggression. They should probably be considered second-line treatment.

SSRIs

No double-blind, placebo-controlled studies have tested any selective serotonin reuptake inhibitor (SSRIs) for reducing aggression in conduct disorder. In a 6-week open study, citalopram (mean 27 mg/d) reduced impulsive aggression in 12 children with mixed diagnoses, as measured by the modified OAS,13 Child Behavior Checklist, and CGI.24

Recommendation. Use caution when prescribing SSRIs to aggressive youth, as these drugs may contribute to aggression in some mood-disordered children. More evidence of SSRIs’ safety and efficacy in this population is needed.

Related resources

  • Overt Aggression Scale. In: Coccaro EF, Harvey PD, Kupsaw-Lawrence E, et al. Development of neuropharmacologically-based behavioral assessments of impulsiveaggressive behavior. J Neuropsychiatry 1991;3(2):S44-S51.

Drug brand names

  • Aripiprazole • Abilify
  • Carbamazepine • Tegretol
  • Citalopram • Celexa
  • Chlorpromazine • Thorazine
  • Clonidine • Catapres
  • Phenytoin sodium • Dilantin
  • Divalproex • Depakote
  • Guanfacine • Tenex
  • Haloperidol • Haldol
  • Olanzapine • Zyprexa
  • Quetiapine • Seroquel
  • Risperidone • Risperdal
  • Thioridazine • Mellaril
  • Ziprasidone • Geodon

Disclosure

Dr. Malone receives research support from Pfizer Inc. and Eli Lilly and Co. and is a consultant to Janssen Pharmaceutica.

Dr. Delaney is a consultant to Shire Pharmaceuticals.

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

References

1. Prescribing psychoactive medications for children and adolescents American Academy of Child and Adolescent Psychiatry policy statement, adopted Sept. 20, 2001. Available at:http://www.aacap.org/publications/policy/ps41.htm Accessed Jan. 15, 2004.

2. Pappadopulos E, MacIntyre JC, Crismon ML, et al. Treatment recommendations for the use of antipsychotics for aggressive youth (TRAAY). Part II. J Am Acad Child Adolesc Psychiatry 2003;42(2):145-61.

3. Hollander E, Tracy KA, Swann AC, et al. Divalproex in the treatment of impulsive aggression: efficacy in Cluster B personality disorders. Neuropsychopharmacology 2003;28:1186-97.

4. Physician’s Desk Reference (57th ed). Montvale, NJ: Thomson Healthcare, 2003.

5. Aman MG, DeSmedt G, Derivan A, et al. Double-blind, placebo-controlled study of risperidone for the treatment of disruptive behaviors in children with subaverage intelligence. Am J Psychiatry 2002;159:1337-46.

6. Snyder R, Turgay A, Aman M, et al. Effects of risperidone on conduct and disruptive behavior disorders in children with subaverage IQs. J Am Acad Child Adolesc Psychiatry 2002;41(9):1026-36.

7. Findling RL, McNamara NK, Branicky LA, et al. A double-blind pilot study of risperidone in the treatment of conduct disorder. J Am Acad Child Adolesc Psychiatry 2000;39:509-16.

8. Malone RP, Maislin G, Choudhury MS, et al. Risperidone treatment in children and adolescents with autism: short- and long-term safety and effectiveness. J Am Acad Child Adolesc Psychiatry 2002;41(2):140-7.

9. Sheard MH, Marini JL, Bridges CI, Wagner E. The effect of lithium on impulsive aggressive behavior in man. Am J Psychiatry 1976;133(12):1409-13.

10. Campbell M, Adams PB, Small AM, et al. Lithium in hospitalized aggressive children with conduct disorder: a double-blind and placebo-controlled study. J Am Acad Child Adolesc Psychiatry 1995;34:445-53.

11. Malone RP, Delaney MA, Luebbert JF, et al. A double-blind placebo-controlled study of lithium in hospitalized aggressive children and adolescents with conduct disorder. Arch Gen Psychiatry 2000a;57(7):649-54.

12. Yudofsky SC, Silver JM, Jackson W, et al. The Overt Aggression Scale for the objective rating of verbal and physical aggression. Am J Psychiatry 1986;143:35-9.

13. Coccaro EF, Harvey PD, Kupsaw-Lawrence E, et al. Development of neuropharmacologically-based behavioral assessments of impulsive aggressive behavior. J Neuropsychiatry 1991;3:S44-S5.

14. Malone RP, Delaney MA, Gifford C. Adverse events during lithium treatment in children varies by setting. Miami Beach, FL: American Academy of Child and Adolescent Psychiatry annual meeting, 2003.

15. Rifkin A, Karajgi B, Dicker R, et al. Lithium treatment of conduct disorders in adolescents. Am J Psychiatry 1997;154:554-5.

16. Klein RG. Preliminary results: lithium effects in conduct disorders. New Orleans: American Psychiatric Association annual meeting, 1991.

17. Zito JM, Safer DJ, DosReis S, et al. Psychotropic practice patterns for youth: a 10-year perspective. Arch Pediatr Adolesc Med 2003;157:17-25.

18. Malone RP, Delaney MA. Psychopharmacologic interventions in children with aggression: neuroleptics, lithium, and anticonvulsants. In: Coccaro EF (ed). Aggression: assessment and treatment. New York: Marcel Dekker, 2003b;331-49.

19. Donovan SJ, Stewart JW, Nunes EV, et al. Divalproex treatment for youth with explosive temper and mood lability: a double-blind, placebo-controlled crossover design. Am J Psychiatry 2000;157:818-20.

20. Steiner H. A randomized clinical trial of divalproex sodium in conduct disorders. J Clin Psychiatry (in press).

21. Isojarvi JT, Laatikainen TJ, Knip M, et al. Obesity and endocrine disorders in women taking valproate for epilepsy. Ann Neurol 1996;39:579-84.

22. MTA Cooperative Group. A 14-month randomized clinical trial of treatment strategies for attention deficit/hyperactivity disorder. Arch Gen Psychiatry 1999;56:1073-86.

23. Conner DF, Barkley RA, Davis HT. A pilot study of methylphenidate, clonidine, or the combination in ADHD comorbid with aggressive oppositional defiant or conduct disorder. Clin Pediatr 2000;39:15-25.

24. Hazell PL, Stuart JE. A randomized controlled trial of clonidine added to psychostimulant medication for hyperactive and aggressive children. J Am Acad Child Adolesc Psychiatry. 2003;886-94.

25. Armenteros JL, Lewis JE. Citalopram treatment for impulsive aggression in children and adolescents: an open pilot study. J Am Acad Child Adolesc Psychiatry 2002;41:522-9.

Author and Disclosure Information

Richard P. Malone, MD
Assistant professor

Mary Anne Delaney, MD
Assistant professor of psychiatry

Roomana Sheikh, MD
Assistant professor of psychiatry

Eastern Pennsylvania Psychiatric Institute Drexel University College of Medicine Philadelphia

Issue
Current Psychiatry - 03(04)
Publications
Topics
Page Number
65-79
Sections
Author and Disclosure Information

Richard P. Malone, MD
Assistant professor

Mary Anne Delaney, MD
Assistant professor of psychiatry

Roomana Sheikh, MD
Assistant professor of psychiatry

Eastern Pennsylvania Psychiatric Institute Drexel University College of Medicine Philadelphia

Author and Disclosure Information

Richard P. Malone, MD
Assistant professor

Mary Anne Delaney, MD
Assistant professor of psychiatry

Roomana Sheikh, MD
Assistant professor of psychiatry

Eastern Pennsylvania Psychiatric Institute Drexel University College of Medicine Philadelphia

Families and schools often pressure clinicians to “do something” when children or adolescents persistently bully, threaten, or injure others. This demand poses a treatment dilemma when psychosocial and educational interventions have failed to manage pediatric aggression.

Aggression is the main reason for drug therapy in youths with conduct disorder, but very little safety and efficacy data exist to help us choose medications. This places young patients at risk for polypharmacy, unmanaged symptoms, short-term side effects, and unknown long-term consequences of exposure to psychotropics.

Table 1

4 precautions when prescribing for pediatric aggression

  • Data to support polypharmacy are limited
  • Most drugs used to treat aggression are not FDA-approved for children
  • Drug treatment requires informed consent
  • Psychosocial treatment must be included in the treatment plan
Source: American Academy of Child and Adolescent Psychiatry1

This article reviews the limited data on using medications to reduce aggression in children and adolescents, focusing on double-blind, placebo-controlled trials in conduct disorder. Based on this evidence and our clinical experience, we offer a sample case and treatment recommendations.

Prescribing principles

Precautions. When prescribing drugs to treat aggressive youth, remember the American Academy of Child and Adolescent Psychiatry’s precautions (Table 1)1 Recently published recommendations prepared by expert consensus are also valuable treatment guides.2

Linking treatment to diagnosis. Should we attempt to manage aggression as a manifestation of an underlying psychiatric disorder? Or should we treat it the same across all disorders? The latter approach is akin to the “fever model.”

Fever—regardless of cause—may be treated with a nonsteroidal anti-inflammatory drug. However, evidence from drug studies suggests that underlying psychiatric disorders should help determine the choice of aggression treatment. For example, a recent study in adults found that divalproex was effective for aggressive patients only within a specific diagnostic subgroup (in this case, cluster B personality disorders).3

Clinical experience also links aggression treatment with underlying diagnoses. For example, aggression secondary to agitated depression is treated with an antidepressant, whereas aggression secondary to command hallucinations in schizophrenia is treated with antipsychotics.

In treating aggression in conduct disorder (Table 2), first treat comorbid disorders—such as attention deficit/hyperactivity disorder (ADHD) or bipolar disorder—and address the child’s psychosocial and educational needs. Then if medication is appropriate, consider drugs with evidence of safety and efficacy, such as antipsychotics, lithium, and stimulants.

Antipsychotics

Three conventional antipsychotics—chlorpromazine, haloperidol, and thioridazine—are FDA-approved for controlling disruptive behaviors in children.4 No atypical antipsychotics are so indicated, but atypicals are preferred in children and adolescents because of lower risks for tardive dyskinesia, neuroleptic malignant syndrome, and extrapyramidal symptoms.2

Risperidone is the most-studied atypical antipsychotic for treating pediatric aggression, particularly in patients with low intellectual functioning or mental retardation. In a 6-week, double-blind, placebo-controlled trial, 118 children ages 5 to 12 with severely disruptive behavior and IQs of 36 to 84 were given low-dose risperidone (mean 1.16 mg/d). Risperidone reduced conduct problems significantly more than placebo, although aggression was not measured directly.5 Adverse events included somnolence, headache, vomiting, weight gain, and elevated serum prolactin. Similar results have been reported in other studies.6

Table 2

Diagnostic criteria for conduct disorder

A. A repetitive and persistent pattern of behavior in which the basic rights of others or major age-appropriate societal norms or rules are violated, as manifested by the persistence of three (or more) of the following criteria in the past 12 months, with at least one criterion present in the past 6 months:
Aggression to people and animals
1. often bullies, threatens, or intimidates others5. has been physically cruel to animals
2. often initiates physical fights6. has stolen while confronting a victim (such as mugging, purse snatching, extortion, armed robbery)
3. has used a weapon that can cause serious physical harm to others (such as a bat, brick, broken bottle, knife, gun)7. has forced someone into sexual activity
4. has been physically cruel to people 
Destruction of property
8. has deliberately engaged in fire setting with the intention of causing serious damage9. has deliberately destroyed others’ property (other than by fire setting)
Deceitfulness or theft
10. has broken into someone else’s house, building, or car12. has stolen items of nontrivial value without confronting a victim (such as shoplifting without breaking and entering, or forgery)
11. often lies to obtain goods or favors or to avoid obligations(ie, “cons” others) 
Serious violation of rules
13. often stays out at night despite parental prohibitions, beginning before age 1315. has run away from home overnight at least twice while living in parental or parental surrogate home (or once without returning for a lengthy period)
14. is often truant from school, beginning before age 13 
B. The disturbance in behavior causes clinically significant impairment in social, academic, or occupational functioning
C. If the individual is age 18 or older, criteria are not met for antisocial personality disorder.
Specify severity:
Mild: few if any conduct problems in excess of those required to make the diagnosis and conduct problems cause only minor harm to others (such as lying, truancy, staying out after dark without permission)
Moderate: number of conduct problems and effect on others intermediate between “mild” and severe” (such as stealing without confronting a victim, vandalism)
Severe: many conduct problems in excess of those required to make the diagnosis or conduct problems cause considerable harm to others (such as forced sex, physical cruelty, use of a weapon, stealing while confronting a victim, breaking and entering)
Source: Reprinted with permission from the Diagnostic and statistical manual of mental disorders, 4th ed., text revision. Copyright 2000. American Psychiatric Association.
 

 

Box 1

Case report: Multiple diagnoses and drugs

JM, age 12, presented with his mother to address symptoms of hyperactivity and impulsive aggression. The boy also complained that his medications made him fall asleep during the day.

He is receiving five medications: a long-acting stimulant, atypical antipsychotic, anticonvulsant, alpha agonist, and selective serotonin reuptake inhibitor (SSRI). He had received numerous other medications, but prescription records are unavailable or incomplete.

Diagnostic history. Since age 5, JM has been diagnosed as having attention-deficit/hyperactivity disorder (ADHD), oppositional defiant disorder, conduct disorder, bipolar disorder, major depressive disorder, and learning disorders. On examination, the boy met DSM-IV criteria for ADHD, learning disorders, and conduct disorder (Table 2). He has a history of starting fights with peers, bullying, destroying property, lying, and stealing from stores and peers.

His mother stated that her son had always had irritable and labile periods, especially when he did not get his way. She was told during a previous psychiatric evaluation that the boy’s "mood swings" indicated bipolar disorder. On examination, however, he had no other bipolar symptoms, and his condition was chronic, not cyclic.

JM typically cries when he does not get his way, his mother reported, but he has no history of sleep or appetite changes that could suggest depression. He is happy when he can do as he pleases.

Reducing medications. After reviewing JM’s medications and performing the psychiatric assessment, the psychiatrist developed a plan to maximize his psychosocial and educational treatments and alter his medications and dosages. The first step was to increase the stimulant dosage to determine whether JM would be less hyperactive and impulsively aggressive.

The psychiatrist was concerned that the anticonvulsant, alpha agonist, and SSRI were not helping and could cause adverse events. He discussed slowly weaning these drugs one at a time with JM and his mother, and they agreed. The goal was to manage JM over time and to reduce his medications to one (ideally) or two (if necessary), possibly continuing the atypical antipsychotic.

Risperidone also reduced aggression in children with normal intelligence in one small study.7 As a cautionary note, however, long-term risperidone treatment has been associated with withdrawal dyskinesias.8

Olanzapine, quetiapine, ziprasidone, and aripiprazole are less well-studied for treating pediatric aggression but are preferable to conventional agents when antipsychotics are considered.

Recommendation. Expert consensus opinion2 recommends using atypicals when psychosocial treatments and first-line medications for primary conditions have failed. Start with low dosages, and titrate up slowly while monitoring symptoms and side effects. Because no studies have compared any atypical’s efficacy over others for aggressive behavior, base your choices on:

  • discussions with the patient and family (Box 1)
  • medical comorbidities
  • how the patient responded to antipsychotics in the past
  • side-effect profile
  • long-term treatment planning.2

If the patient cannot tolerate the medication or does not respond after 4 to 6 weeks, try switching atypicals. To improve partial response, consider adding a mood stabilizer such as lithium or divalproex. If aggressive symptoms remit for 6 months or longer, attempt to taper or discontinue the antipsychotic.2

Lithium

In placebo-controlled trials, lithium reduced aggression in:

  • male prisoners ages 16 to 24.9
  • children ages 7 and 12 with conduct disorder10
  • children and adolescents ages 10 to 17 with conduct disorder.11

Among these studies, only ours11 specifically measured aggression. We randomly assigned 40 children to receive 4 weeks of lithium, 900 to 2,100 mg/d (mean 1,425 ± 321 mg/d), or placebo. Serum lithium levels were 0.78 to 1.55 mEq/L (mean 1.07 ± 0.19 mEq/L). We used the Overt Aggression Scale (OAS)12,13 (see Related resources) to track frequency and severity of verbal aggression, aggression against objects, aggression against others, and self-aggression.

Lithium reduced aggression more than did placebo, as measured by the clinician-rated Clinical Global Impressions (CGI) scale and staff-rated Global Clinical Judgments (Consensus) Scale (GCJCS). The CGI showed a 70% response rate with lithium and 20% with placebo. Similarly, the GCJCS scale showed 80% response with lithium and 30% with placebo.

The aggression reduction with lithium was statistically significant and clinically evident. Most subjects (37 of 40) experienced at least one adverse event, however, whether receiving lithium or placebo. Nausea, vomiting, and urinary frequency were significantly more common in the lithium-treated group than with placebo. Fewer adverse events were reported in a similar outpatient study,14 probably because of less-frequent monitoring.

Lithium did not reduce aggression in adolescent girls treated for 2 weeks15 or in an outpatient study of children with ADHD.16

Recommendation. Lithium has shown efficacy for reducing severe aggression in hospitalized children with conduct disorder but not in similar outpatients. Consider this drug to reduce severe aggression in children with conduct disorder, especially if they have failed other treatments.

 

 

Anticonvulsants

Anticonvulsants have been used to decrease aggression for more than 50 years, and epidemiologic data show their use is increasing markedly.17 Few controlled studies support this prescribing trend, however.18

Initial reports suggested that anticonvulsants reduce disruptive behaviors, but more-critically designed studies have not supported this finding. For example, phenytoin sodium (diphenylhydantoin) demonstrated efficacy in open trials, but controlled trials found this anticonvulsant no more effective than placebo. In fact, placebo may have reduced aggression more than the active drug. Likewise, earlier controlled trials of carbamazepine indicated efficacy, but more-carefully designed trials using specific measures of aggression did not.

Divalproex is the anticonvulsant most commonly used for aggression in children and adolescents. Only one small, placebo-controlled study has found it effective in reducing aggression in children.19

Twenty children ages 10 to 18 with conduct disorder or oppositional defiant disorder were randomized to divalproex, 750 to 1,500 mg/d, or placebo. Eighteen completed the first phase, and 15 crossed over to the other treatment. Concomitant drug treatment, including stimulants, was allowed. The authors reported that 12 of 15 subjects showed some response to divalproex.

A 7-week study compared divalproex in high dosages (up to 1,500 mg/d) versus low dosages (up to 250 mg/d). This study was not placebo-controlled, but aggression was reduced more in the high-dosage than in the low-dosage group.20

Recommendation. If you use an anticonvulsant, first obtain informed consent from the patient and parent. Divalproex causes weight gain and has been associated with increased risk of polycystic ovary syndrome with masculinizing effects.21

Double-blind, placebo-controlled studies of divalproex and other anticonvulsants in treating aggression are needed, particularly as prescriptions for these agents are rising.

Stimulants

Some small controlled studies suggest that stimulants can reduce aggression in children with ADHD, but their effects on aggression in conduct disorder have not been well studied. Aggression was not measured directly in the National Institute of Mental Health Multimodal Treatment Study of Children with ADHD.21 Most other studies have been small and included children with ADHD but not necessarily conduct disorder.

Recommendation. Stimulants may help reduce aggression in children with ADHD, but studies gauging their effects in conduct disorder are needed.

Alpha agonists

Alpha agonists such as clonidine and guanfacine are increasingly being used to treat children with disruptive disorders, despite limited evidence. The small controlled studies that examined alpha agonists as monotherapy or add-ons in this population did not directly measure aggression.22,23

Recommendation. Little data support alpha agonists for reducing aggression. They should probably be considered second-line treatment.

SSRIs

No double-blind, placebo-controlled studies have tested any selective serotonin reuptake inhibitor (SSRIs) for reducing aggression in conduct disorder. In a 6-week open study, citalopram (mean 27 mg/d) reduced impulsive aggression in 12 children with mixed diagnoses, as measured by the modified OAS,13 Child Behavior Checklist, and CGI.24

Recommendation. Use caution when prescribing SSRIs to aggressive youth, as these drugs may contribute to aggression in some mood-disordered children. More evidence of SSRIs’ safety and efficacy in this population is needed.

Related resources

  • Overt Aggression Scale. In: Coccaro EF, Harvey PD, Kupsaw-Lawrence E, et al. Development of neuropharmacologically-based behavioral assessments of impulsiveaggressive behavior. J Neuropsychiatry 1991;3(2):S44-S51.

Drug brand names

  • Aripiprazole • Abilify
  • Carbamazepine • Tegretol
  • Citalopram • Celexa
  • Chlorpromazine • Thorazine
  • Clonidine • Catapres
  • Phenytoin sodium • Dilantin
  • Divalproex • Depakote
  • Guanfacine • Tenex
  • Haloperidol • Haldol
  • Olanzapine • Zyprexa
  • Quetiapine • Seroquel
  • Risperidone • Risperdal
  • Thioridazine • Mellaril
  • Ziprasidone • Geodon

Disclosure

Dr. Malone receives research support from Pfizer Inc. and Eli Lilly and Co. and is a consultant to Janssen Pharmaceutica.

Dr. Delaney is a consultant to Shire Pharmaceuticals.

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

Families and schools often pressure clinicians to “do something” when children or adolescents persistently bully, threaten, or injure others. This demand poses a treatment dilemma when psychosocial and educational interventions have failed to manage pediatric aggression.

Aggression is the main reason for drug therapy in youths with conduct disorder, but very little safety and efficacy data exist to help us choose medications. This places young patients at risk for polypharmacy, unmanaged symptoms, short-term side effects, and unknown long-term consequences of exposure to psychotropics.

Table 1

4 precautions when prescribing for pediatric aggression

  • Data to support polypharmacy are limited
  • Most drugs used to treat aggression are not FDA-approved for children
  • Drug treatment requires informed consent
  • Psychosocial treatment must be included in the treatment plan
Source: American Academy of Child and Adolescent Psychiatry1

This article reviews the limited data on using medications to reduce aggression in children and adolescents, focusing on double-blind, placebo-controlled trials in conduct disorder. Based on this evidence and our clinical experience, we offer a sample case and treatment recommendations.

Prescribing principles

Precautions. When prescribing drugs to treat aggressive youth, remember the American Academy of Child and Adolescent Psychiatry’s precautions (Table 1)1 Recently published recommendations prepared by expert consensus are also valuable treatment guides.2

Linking treatment to diagnosis. Should we attempt to manage aggression as a manifestation of an underlying psychiatric disorder? Or should we treat it the same across all disorders? The latter approach is akin to the “fever model.”

Fever—regardless of cause—may be treated with a nonsteroidal anti-inflammatory drug. However, evidence from drug studies suggests that underlying psychiatric disorders should help determine the choice of aggression treatment. For example, a recent study in adults found that divalproex was effective for aggressive patients only within a specific diagnostic subgroup (in this case, cluster B personality disorders).3

Clinical experience also links aggression treatment with underlying diagnoses. For example, aggression secondary to agitated depression is treated with an antidepressant, whereas aggression secondary to command hallucinations in schizophrenia is treated with antipsychotics.

In treating aggression in conduct disorder (Table 2), first treat comorbid disorders—such as attention deficit/hyperactivity disorder (ADHD) or bipolar disorder—and address the child’s psychosocial and educational needs. Then if medication is appropriate, consider drugs with evidence of safety and efficacy, such as antipsychotics, lithium, and stimulants.

Antipsychotics

Three conventional antipsychotics—chlorpromazine, haloperidol, and thioridazine—are FDA-approved for controlling disruptive behaviors in children.4 No atypical antipsychotics are so indicated, but atypicals are preferred in children and adolescents because of lower risks for tardive dyskinesia, neuroleptic malignant syndrome, and extrapyramidal symptoms.2

Risperidone is the most-studied atypical antipsychotic for treating pediatric aggression, particularly in patients with low intellectual functioning or mental retardation. In a 6-week, double-blind, placebo-controlled trial, 118 children ages 5 to 12 with severely disruptive behavior and IQs of 36 to 84 were given low-dose risperidone (mean 1.16 mg/d). Risperidone reduced conduct problems significantly more than placebo, although aggression was not measured directly.5 Adverse events included somnolence, headache, vomiting, weight gain, and elevated serum prolactin. Similar results have been reported in other studies.6

Table 2

Diagnostic criteria for conduct disorder

A. A repetitive and persistent pattern of behavior in which the basic rights of others or major age-appropriate societal norms or rules are violated, as manifested by the persistence of three (or more) of the following criteria in the past 12 months, with at least one criterion present in the past 6 months:
Aggression to people and animals
1. often bullies, threatens, or intimidates others5. has been physically cruel to animals
2. often initiates physical fights6. has stolen while confronting a victim (such as mugging, purse snatching, extortion, armed robbery)
3. has used a weapon that can cause serious physical harm to others (such as a bat, brick, broken bottle, knife, gun)7. has forced someone into sexual activity
4. has been physically cruel to people 
Destruction of property
8. has deliberately engaged in fire setting with the intention of causing serious damage9. has deliberately destroyed others’ property (other than by fire setting)
Deceitfulness or theft
10. has broken into someone else’s house, building, or car12. has stolen items of nontrivial value without confronting a victim (such as shoplifting without breaking and entering, or forgery)
11. often lies to obtain goods or favors or to avoid obligations(ie, “cons” others) 
Serious violation of rules
13. often stays out at night despite parental prohibitions, beginning before age 1315. has run away from home overnight at least twice while living in parental or parental surrogate home (or once without returning for a lengthy period)
14. is often truant from school, beginning before age 13 
B. The disturbance in behavior causes clinically significant impairment in social, academic, or occupational functioning
C. If the individual is age 18 or older, criteria are not met for antisocial personality disorder.
Specify severity:
Mild: few if any conduct problems in excess of those required to make the diagnosis and conduct problems cause only minor harm to others (such as lying, truancy, staying out after dark without permission)
Moderate: number of conduct problems and effect on others intermediate between “mild” and severe” (such as stealing without confronting a victim, vandalism)
Severe: many conduct problems in excess of those required to make the diagnosis or conduct problems cause considerable harm to others (such as forced sex, physical cruelty, use of a weapon, stealing while confronting a victim, breaking and entering)
Source: Reprinted with permission from the Diagnostic and statistical manual of mental disorders, 4th ed., text revision. Copyright 2000. American Psychiatric Association.
 

 

Box 1

Case report: Multiple diagnoses and drugs

JM, age 12, presented with his mother to address symptoms of hyperactivity and impulsive aggression. The boy also complained that his medications made him fall asleep during the day.

He is receiving five medications: a long-acting stimulant, atypical antipsychotic, anticonvulsant, alpha agonist, and selective serotonin reuptake inhibitor (SSRI). He had received numerous other medications, but prescription records are unavailable or incomplete.

Diagnostic history. Since age 5, JM has been diagnosed as having attention-deficit/hyperactivity disorder (ADHD), oppositional defiant disorder, conduct disorder, bipolar disorder, major depressive disorder, and learning disorders. On examination, the boy met DSM-IV criteria for ADHD, learning disorders, and conduct disorder (Table 2). He has a history of starting fights with peers, bullying, destroying property, lying, and stealing from stores and peers.

His mother stated that her son had always had irritable and labile periods, especially when he did not get his way. She was told during a previous psychiatric evaluation that the boy’s "mood swings" indicated bipolar disorder. On examination, however, he had no other bipolar symptoms, and his condition was chronic, not cyclic.

JM typically cries when he does not get his way, his mother reported, but he has no history of sleep or appetite changes that could suggest depression. He is happy when he can do as he pleases.

Reducing medications. After reviewing JM’s medications and performing the psychiatric assessment, the psychiatrist developed a plan to maximize his psychosocial and educational treatments and alter his medications and dosages. The first step was to increase the stimulant dosage to determine whether JM would be less hyperactive and impulsively aggressive.

The psychiatrist was concerned that the anticonvulsant, alpha agonist, and SSRI were not helping and could cause adverse events. He discussed slowly weaning these drugs one at a time with JM and his mother, and they agreed. The goal was to manage JM over time and to reduce his medications to one (ideally) or two (if necessary), possibly continuing the atypical antipsychotic.

Risperidone also reduced aggression in children with normal intelligence in one small study.7 As a cautionary note, however, long-term risperidone treatment has been associated with withdrawal dyskinesias.8

Olanzapine, quetiapine, ziprasidone, and aripiprazole are less well-studied for treating pediatric aggression but are preferable to conventional agents when antipsychotics are considered.

Recommendation. Expert consensus opinion2 recommends using atypicals when psychosocial treatments and first-line medications for primary conditions have failed. Start with low dosages, and titrate up slowly while monitoring symptoms and side effects. Because no studies have compared any atypical’s efficacy over others for aggressive behavior, base your choices on:

  • discussions with the patient and family (Box 1)
  • medical comorbidities
  • how the patient responded to antipsychotics in the past
  • side-effect profile
  • long-term treatment planning.2

If the patient cannot tolerate the medication or does not respond after 4 to 6 weeks, try switching atypicals. To improve partial response, consider adding a mood stabilizer such as lithium or divalproex. If aggressive symptoms remit for 6 months or longer, attempt to taper or discontinue the antipsychotic.2

Lithium

In placebo-controlled trials, lithium reduced aggression in:

  • male prisoners ages 16 to 24.9
  • children ages 7 and 12 with conduct disorder10
  • children and adolescents ages 10 to 17 with conduct disorder.11

Among these studies, only ours11 specifically measured aggression. We randomly assigned 40 children to receive 4 weeks of lithium, 900 to 2,100 mg/d (mean 1,425 ± 321 mg/d), or placebo. Serum lithium levels were 0.78 to 1.55 mEq/L (mean 1.07 ± 0.19 mEq/L). We used the Overt Aggression Scale (OAS)12,13 (see Related resources) to track frequency and severity of verbal aggression, aggression against objects, aggression against others, and self-aggression.

Lithium reduced aggression more than did placebo, as measured by the clinician-rated Clinical Global Impressions (CGI) scale and staff-rated Global Clinical Judgments (Consensus) Scale (GCJCS). The CGI showed a 70% response rate with lithium and 20% with placebo. Similarly, the GCJCS scale showed 80% response with lithium and 30% with placebo.

The aggression reduction with lithium was statistically significant and clinically evident. Most subjects (37 of 40) experienced at least one adverse event, however, whether receiving lithium or placebo. Nausea, vomiting, and urinary frequency were significantly more common in the lithium-treated group than with placebo. Fewer adverse events were reported in a similar outpatient study,14 probably because of less-frequent monitoring.

Lithium did not reduce aggression in adolescent girls treated for 2 weeks15 or in an outpatient study of children with ADHD.16

Recommendation. Lithium has shown efficacy for reducing severe aggression in hospitalized children with conduct disorder but not in similar outpatients. Consider this drug to reduce severe aggression in children with conduct disorder, especially if they have failed other treatments.

 

 

Anticonvulsants

Anticonvulsants have been used to decrease aggression for more than 50 years, and epidemiologic data show their use is increasing markedly.17 Few controlled studies support this prescribing trend, however.18

Initial reports suggested that anticonvulsants reduce disruptive behaviors, but more-critically designed studies have not supported this finding. For example, phenytoin sodium (diphenylhydantoin) demonstrated efficacy in open trials, but controlled trials found this anticonvulsant no more effective than placebo. In fact, placebo may have reduced aggression more than the active drug. Likewise, earlier controlled trials of carbamazepine indicated efficacy, but more-carefully designed trials using specific measures of aggression did not.

Divalproex is the anticonvulsant most commonly used for aggression in children and adolescents. Only one small, placebo-controlled study has found it effective in reducing aggression in children.19

Twenty children ages 10 to 18 with conduct disorder or oppositional defiant disorder were randomized to divalproex, 750 to 1,500 mg/d, or placebo. Eighteen completed the first phase, and 15 crossed over to the other treatment. Concomitant drug treatment, including stimulants, was allowed. The authors reported that 12 of 15 subjects showed some response to divalproex.

A 7-week study compared divalproex in high dosages (up to 1,500 mg/d) versus low dosages (up to 250 mg/d). This study was not placebo-controlled, but aggression was reduced more in the high-dosage than in the low-dosage group.20

Recommendation. If you use an anticonvulsant, first obtain informed consent from the patient and parent. Divalproex causes weight gain and has been associated with increased risk of polycystic ovary syndrome with masculinizing effects.21

Double-blind, placebo-controlled studies of divalproex and other anticonvulsants in treating aggression are needed, particularly as prescriptions for these agents are rising.

Stimulants

Some small controlled studies suggest that stimulants can reduce aggression in children with ADHD, but their effects on aggression in conduct disorder have not been well studied. Aggression was not measured directly in the National Institute of Mental Health Multimodal Treatment Study of Children with ADHD.21 Most other studies have been small and included children with ADHD but not necessarily conduct disorder.

Recommendation. Stimulants may help reduce aggression in children with ADHD, but studies gauging their effects in conduct disorder are needed.

Alpha agonists

Alpha agonists such as clonidine and guanfacine are increasingly being used to treat children with disruptive disorders, despite limited evidence. The small controlled studies that examined alpha agonists as monotherapy or add-ons in this population did not directly measure aggression.22,23

Recommendation. Little data support alpha agonists for reducing aggression. They should probably be considered second-line treatment.

SSRIs

No double-blind, placebo-controlled studies have tested any selective serotonin reuptake inhibitor (SSRIs) for reducing aggression in conduct disorder. In a 6-week open study, citalopram (mean 27 mg/d) reduced impulsive aggression in 12 children with mixed diagnoses, as measured by the modified OAS,13 Child Behavior Checklist, and CGI.24

Recommendation. Use caution when prescribing SSRIs to aggressive youth, as these drugs may contribute to aggression in some mood-disordered children. More evidence of SSRIs’ safety and efficacy in this population is needed.

Related resources

  • Overt Aggression Scale. In: Coccaro EF, Harvey PD, Kupsaw-Lawrence E, et al. Development of neuropharmacologically-based behavioral assessments of impulsiveaggressive behavior. J Neuropsychiatry 1991;3(2):S44-S51.

Drug brand names

  • Aripiprazole • Abilify
  • Carbamazepine • Tegretol
  • Citalopram • Celexa
  • Chlorpromazine • Thorazine
  • Clonidine • Catapres
  • Phenytoin sodium • Dilantin
  • Divalproex • Depakote
  • Guanfacine • Tenex
  • Haloperidol • Haldol
  • Olanzapine • Zyprexa
  • Quetiapine • Seroquel
  • Risperidone • Risperdal
  • Thioridazine • Mellaril
  • Ziprasidone • Geodon

Disclosure

Dr. Malone receives research support from Pfizer Inc. and Eli Lilly and Co. and is a consultant to Janssen Pharmaceutica.

Dr. Delaney is a consultant to Shire Pharmaceuticals.

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

References

1. Prescribing psychoactive medications for children and adolescents American Academy of Child and Adolescent Psychiatry policy statement, adopted Sept. 20, 2001. Available at:http://www.aacap.org/publications/policy/ps41.htm Accessed Jan. 15, 2004.

2. Pappadopulos E, MacIntyre JC, Crismon ML, et al. Treatment recommendations for the use of antipsychotics for aggressive youth (TRAAY). Part II. J Am Acad Child Adolesc Psychiatry 2003;42(2):145-61.

3. Hollander E, Tracy KA, Swann AC, et al. Divalproex in the treatment of impulsive aggression: efficacy in Cluster B personality disorders. Neuropsychopharmacology 2003;28:1186-97.

4. Physician’s Desk Reference (57th ed). Montvale, NJ: Thomson Healthcare, 2003.

5. Aman MG, DeSmedt G, Derivan A, et al. Double-blind, placebo-controlled study of risperidone for the treatment of disruptive behaviors in children with subaverage intelligence. Am J Psychiatry 2002;159:1337-46.

6. Snyder R, Turgay A, Aman M, et al. Effects of risperidone on conduct and disruptive behavior disorders in children with subaverage IQs. J Am Acad Child Adolesc Psychiatry 2002;41(9):1026-36.

7. Findling RL, McNamara NK, Branicky LA, et al. A double-blind pilot study of risperidone in the treatment of conduct disorder. J Am Acad Child Adolesc Psychiatry 2000;39:509-16.

8. Malone RP, Maislin G, Choudhury MS, et al. Risperidone treatment in children and adolescents with autism: short- and long-term safety and effectiveness. J Am Acad Child Adolesc Psychiatry 2002;41(2):140-7.

9. Sheard MH, Marini JL, Bridges CI, Wagner E. The effect of lithium on impulsive aggressive behavior in man. Am J Psychiatry 1976;133(12):1409-13.

10. Campbell M, Adams PB, Small AM, et al. Lithium in hospitalized aggressive children with conduct disorder: a double-blind and placebo-controlled study. J Am Acad Child Adolesc Psychiatry 1995;34:445-53.

11. Malone RP, Delaney MA, Luebbert JF, et al. A double-blind placebo-controlled study of lithium in hospitalized aggressive children and adolescents with conduct disorder. Arch Gen Psychiatry 2000a;57(7):649-54.

12. Yudofsky SC, Silver JM, Jackson W, et al. The Overt Aggression Scale for the objective rating of verbal and physical aggression. Am J Psychiatry 1986;143:35-9.

13. Coccaro EF, Harvey PD, Kupsaw-Lawrence E, et al. Development of neuropharmacologically-based behavioral assessments of impulsive aggressive behavior. J Neuropsychiatry 1991;3:S44-S5.

14. Malone RP, Delaney MA, Gifford C. Adverse events during lithium treatment in children varies by setting. Miami Beach, FL: American Academy of Child and Adolescent Psychiatry annual meeting, 2003.

15. Rifkin A, Karajgi B, Dicker R, et al. Lithium treatment of conduct disorders in adolescents. Am J Psychiatry 1997;154:554-5.

16. Klein RG. Preliminary results: lithium effects in conduct disorders. New Orleans: American Psychiatric Association annual meeting, 1991.

17. Zito JM, Safer DJ, DosReis S, et al. Psychotropic practice patterns for youth: a 10-year perspective. Arch Pediatr Adolesc Med 2003;157:17-25.

18. Malone RP, Delaney MA. Psychopharmacologic interventions in children with aggression: neuroleptics, lithium, and anticonvulsants. In: Coccaro EF (ed). Aggression: assessment and treatment. New York: Marcel Dekker, 2003b;331-49.

19. Donovan SJ, Stewart JW, Nunes EV, et al. Divalproex treatment for youth with explosive temper and mood lability: a double-blind, placebo-controlled crossover design. Am J Psychiatry 2000;157:818-20.

20. Steiner H. A randomized clinical trial of divalproex sodium in conduct disorders. J Clin Psychiatry (in press).

21. Isojarvi JT, Laatikainen TJ, Knip M, et al. Obesity and endocrine disorders in women taking valproate for epilepsy. Ann Neurol 1996;39:579-84.

22. MTA Cooperative Group. A 14-month randomized clinical trial of treatment strategies for attention deficit/hyperactivity disorder. Arch Gen Psychiatry 1999;56:1073-86.

23. Conner DF, Barkley RA, Davis HT. A pilot study of methylphenidate, clonidine, or the combination in ADHD comorbid with aggressive oppositional defiant or conduct disorder. Clin Pediatr 2000;39:15-25.

24. Hazell PL, Stuart JE. A randomized controlled trial of clonidine added to psychostimulant medication for hyperactive and aggressive children. J Am Acad Child Adolesc Psychiatry. 2003;886-94.

25. Armenteros JL, Lewis JE. Citalopram treatment for impulsive aggression in children and adolescents: an open pilot study. J Am Acad Child Adolesc Psychiatry 2002;41:522-9.

References

1. Prescribing psychoactive medications for children and adolescents American Academy of Child and Adolescent Psychiatry policy statement, adopted Sept. 20, 2001. Available at:http://www.aacap.org/publications/policy/ps41.htm Accessed Jan. 15, 2004.

2. Pappadopulos E, MacIntyre JC, Crismon ML, et al. Treatment recommendations for the use of antipsychotics for aggressive youth (TRAAY). Part II. J Am Acad Child Adolesc Psychiatry 2003;42(2):145-61.

3. Hollander E, Tracy KA, Swann AC, et al. Divalproex in the treatment of impulsive aggression: efficacy in Cluster B personality disorders. Neuropsychopharmacology 2003;28:1186-97.

4. Physician’s Desk Reference (57th ed). Montvale, NJ: Thomson Healthcare, 2003.

5. Aman MG, DeSmedt G, Derivan A, et al. Double-blind, placebo-controlled study of risperidone for the treatment of disruptive behaviors in children with subaverage intelligence. Am J Psychiatry 2002;159:1337-46.

6. Snyder R, Turgay A, Aman M, et al. Effects of risperidone on conduct and disruptive behavior disorders in children with subaverage IQs. J Am Acad Child Adolesc Psychiatry 2002;41(9):1026-36.

7. Findling RL, McNamara NK, Branicky LA, et al. A double-blind pilot study of risperidone in the treatment of conduct disorder. J Am Acad Child Adolesc Psychiatry 2000;39:509-16.

8. Malone RP, Maislin G, Choudhury MS, et al. Risperidone treatment in children and adolescents with autism: short- and long-term safety and effectiveness. J Am Acad Child Adolesc Psychiatry 2002;41(2):140-7.

9. Sheard MH, Marini JL, Bridges CI, Wagner E. The effect of lithium on impulsive aggressive behavior in man. Am J Psychiatry 1976;133(12):1409-13.

10. Campbell M, Adams PB, Small AM, et al. Lithium in hospitalized aggressive children with conduct disorder: a double-blind and placebo-controlled study. J Am Acad Child Adolesc Psychiatry 1995;34:445-53.

11. Malone RP, Delaney MA, Luebbert JF, et al. A double-blind placebo-controlled study of lithium in hospitalized aggressive children and adolescents with conduct disorder. Arch Gen Psychiatry 2000a;57(7):649-54.

12. Yudofsky SC, Silver JM, Jackson W, et al. The Overt Aggression Scale for the objective rating of verbal and physical aggression. Am J Psychiatry 1986;143:35-9.

13. Coccaro EF, Harvey PD, Kupsaw-Lawrence E, et al. Development of neuropharmacologically-based behavioral assessments of impulsive aggressive behavior. J Neuropsychiatry 1991;3:S44-S5.

14. Malone RP, Delaney MA, Gifford C. Adverse events during lithium treatment in children varies by setting. Miami Beach, FL: American Academy of Child and Adolescent Psychiatry annual meeting, 2003.

15. Rifkin A, Karajgi B, Dicker R, et al. Lithium treatment of conduct disorders in adolescents. Am J Psychiatry 1997;154:554-5.

16. Klein RG. Preliminary results: lithium effects in conduct disorders. New Orleans: American Psychiatric Association annual meeting, 1991.

17. Zito JM, Safer DJ, DosReis S, et al. Psychotropic practice patterns for youth: a 10-year perspective. Arch Pediatr Adolesc Med 2003;157:17-25.

18. Malone RP, Delaney MA. Psychopharmacologic interventions in children with aggression: neuroleptics, lithium, and anticonvulsants. In: Coccaro EF (ed). Aggression: assessment and treatment. New York: Marcel Dekker, 2003b;331-49.

19. Donovan SJ, Stewart JW, Nunes EV, et al. Divalproex treatment for youth with explosive temper and mood lability: a double-blind, placebo-controlled crossover design. Am J Psychiatry 2000;157:818-20.

20. Steiner H. A randomized clinical trial of divalproex sodium in conduct disorders. J Clin Psychiatry (in press).

21. Isojarvi JT, Laatikainen TJ, Knip M, et al. Obesity and endocrine disorders in women taking valproate for epilepsy. Ann Neurol 1996;39:579-84.

22. MTA Cooperative Group. A 14-month randomized clinical trial of treatment strategies for attention deficit/hyperactivity disorder. Arch Gen Psychiatry 1999;56:1073-86.

23. Conner DF, Barkley RA, Davis HT. A pilot study of methylphenidate, clonidine, or the combination in ADHD comorbid with aggressive oppositional defiant or conduct disorder. Clin Pediatr 2000;39:15-25.

24. Hazell PL, Stuart JE. A randomized controlled trial of clonidine added to psychostimulant medication for hyperactive and aggressive children. J Am Acad Child Adolesc Psychiatry. 2003;886-94.

25. Armenteros JL, Lewis JE. Citalopram treatment for impulsive aggression in children and adolescents: an open pilot study. J Am Acad Child Adolesc Psychiatry 2002;41:522-9.

Issue
Current Psychiatry - 03(04)
Issue
Current Psychiatry - 03(04)
Page Number
65-79
Page Number
65-79
Publications
Publications
Topics
Article Type
Display Headline
How to reduce aggression in youths with conduct disorder
Display Headline
How to reduce aggression in youths with conduct disorder
Sections
Article Source

PURLs Copyright

Inside the Article

The painful truth about depression

Article Type
Changed
Tue, 12/11/2018 - 15:31
Display Headline
The painful truth about depression

History: Initial symptoms

Ms. F, age 39, presented with depression, severe anxiety, and disturbed sleep. She denied psychiatric or medical history, but reported that her depressive symptoms hampered her performance at work and led to work-related stress.

Ms. F’s Beck Depression Inventory (BDI) score at baseline was 21, indicating moderate depression. The psychiatrist diagnosed her as having generalized anxiety disorder and adjustment disorder with depressed mood.

The patient was started on paroxetine, 10 mg/d. Five weeks later, her anxiety had decreased significantly and her BDI score had dropped to 10, indicating normal mood. Both the patient and clinician decided at this point that Ms. F had reached remission. The patient demanded that paroxetine be stopped, saying that she “does not like medication.” The psychiatrist reluctantly agreed.

Eight weeks later, during a follow-up examination, Ms. F complained of severely depressed mood with frequent crying spells. She complained of fatigue, nausea, headaches, decreased appetite, and dizziness. Her work performance, which had improved during the paroxetine trial, was again compromised. Her BDI score was 32, indicating severe depression.

Was the psychiatrist justified in stopping paroxetine therapy after 5 weeks?

Dr. Fishbain’s observations

Premature paroxetine discontinuation cannot be ruled out as a cause for Ms. F’s relapse. Sood et al1 found that duration of antidepressant therapy beyond treatment guidelines correlated with longer times to relapse.

By contrast, Ms. F’s initial therapy duration fell short of the 6 to 8 weeks recommended by the American Psychiatric Association.2

Patients commonly cite adverse events as a reason for wanting to stop antidepressant therapy.3 Ms. F reported no adverse effects, however; she said only that she did “not like medication.” Despite her insistence to the contrary, antidepressant therapy probably should not have been stopped.

Further history: A painful discovery

After questioning, Ms. F told the psychiatrist that she had been involved in a motor vehicle accident 2 weeks before the follow-up visit and had since been suffering lower back and neck pain.

After more questioning, Mr. F revealed that the pain was disrupting her sleep. She was getting about 4 hours of fragmented sleep per night, resulting in lack of energy during the day. The pain made it hard for her to sit, further impairing her work performance.

Ms. F was restarted on paroxetine, 10 mg/d titrated across 6 weeks to 60 mg/d for her depression, and zolpidem, 10 mg at bedtime, to help her sleep. After 6 weeks, her BDI score improved to 20, and she was less labile. Her depressive symptoms persisted, however, as did her pain, fatigue, headaches, nausea, dizziness, and sleep disturbances.

What role did Ms. F’s pain play in her relapse? How can clinicians detect somatic symptoms and gauge their effect on mood?

Dr. Fishbain’s observations

Pain most likely caused Ms. F’s depression relapse. McBeth et al4 have demonstrated that pain can contribute to depression’s development. In another study,5 43.4% of subjects who met criteria for major depression reported painful symptoms. The presence of a chronic painful condition was also found to contribute to major depression.5

Nakao et al6 screened 2,215 outpatients who were referred with mind/body complaints. Patients who were diagnosed with major depression had significantly higher rates of fatigue (86% vs. 65%), insomnia (79% vs. 58%), nausea/vomiting (51% vs. 40%), and low-back pain (36% vs. 24%) than those who were not. Within the major depression group, somatic symptoms were more abundant in patients with severe depressive episodes than in those with mild depressive episodes (5.8 vs. 3.7, P < 0.05).

Depression prevalence appears to increase when somatic symptoms are considered in the diagnosis. Posse and Hallstrom7 used a two-stage design to screen for depression. In the first stage, depression prevalence was 1.8%. In the second stage, 62 patients with high somatic complaint scores were re-evaluated. Of this group, 41 were diagnosed with a major depressive disorder or dysthymia.

Patients with continued pain after depression treatment are at high risk for depression recurrence.8 Diffuseness of pain and extent to which it interferes with daily activities strongly predict depression.9

Diagnostic challenges. Patients with depression often present to their primary care physicians with somatic rather than behavioral symptoms, making it hard for the family doctor to diagnose depression.10 By contrast, when presenting to a psychiatrist, depressed patients tend to discuss their emotional symptoms but not their physical complaints.11 This is because patients often:

  • attribute physical symptoms to an unrelated medical illness
  • consider aches and pains a normal part of aging
  • or are not aware that psychiatrists can treat physical symptoms.11

Psychiatrists in the past have emphasized emotional symptoms while barely addressing physical symptoms. This trend is changing, however, as the link between physical pain and depression has become clearer.

 

 

Be sure to include chronic pain or other somatic symptoms in the systems review. Screening tools such as the Visual Analogue Scale can measure pain intensity, while the Cornell Medical Index can uncover somatic symptoms. No all-inclusive tool exists to help detect depression-related somatic symptoms. however.

Should the psychiatrist continue to address Ms. F’s depressive symptoms, or should the focus shift to her somatic symptoms?

Dr. Fishbain’s observations

Patients who do not respond to depression treatment (ie, achieve >50% symptom reduction), or who respond without achieving remission, usually have residual physical symptoms—often fatigue, sleep disturbance, decreased appetite, anxiety, sexual dysfunction, and/or pain.12-14 Severe pain and other somatic symptoms are likely prolonging Ms. F’s depression, despite increased paroxetine dosages.

Paykel et al8 have associated residual depression symptoms with early relapse of depression. In their study, 94% of depressed patients with lingering depressive symptoms had mild to moderate physical symptoms. By contrast, degree of physical symptom improvement has been shown to correlate with likelihood of depression remission.15

Although emotional symptoms improve with antidepressants,16 some evidence17 indicates that physical symptoms associated with depression may be less responsive.

Also, because many psychiatrists have been taught to track emotional symptoms and only some physical symptoms, somatic symptoms of depression often are not targeted for treatment.17 Lack of rating scales to track somatic symptoms compounds this problem.17

Psychiatrists need to target both the physical and emotional symptoms of depression. When pain prolongs depression, it should be the primary target of antidepressant drug therapy (Algorithm).

To date, several meta-analyses18-20 have demonstrated that antidepressants have a separate analgesic effect on all forms of chronic pain. Evidence21,22 also indicates that the dualaction antidepressants—such as amitriptyline, bupropion, venlafaxine, and (awaiting FDA approval) duloxetine—have a more-consistent analgesic effect than do the serotonin reuptake inhibitors.

The analgesic effects of bupropion, duloxetine, and venlafaxine have not been compared with those of tricyclics or other older antidepressants. If one of the newer dualaction antidepressants does not reduce somatic conditions or produce an adequate response, consider switching to a tricyclic.

Treatment: No pain, some gain

Another psychiatrist who specializes in pain medicine targeted some of Ms. F’s somatic symptoms with antidepressants. Paroxetine and zolpidem were discontinued and the patient was started on:

  • venlafaxine, 37.5 mg bid, titrated to 225 mg/d across 2 weeks. Because of its activating properties, venlafaxine was chosen to address Ms. F’s pain and daytime fatigue.
  • amitriptyline, 50 mg at bedtime nightly, to promote sleep
  • prochlorperazine, 10 mg as needed, and meclizine, 25 mg as needed, to treat her nausea and dizziness, respectively.

Algorithm Suggested drug treatment of depression with somatic symptoms



Ms. F also was advised to take an abortive migraine compound (Midrin, 2 tablets at headache onset and 1 additional tablet every half-hour as needed, maximum 5 tablets per day). Mr. F’s primary care physician also referred her to a physical therapist to treat her neck and low-back pain; workup revealed no surgically treatable problem.

Four weeks later, Ms. F reported that her somatic symptoms significantly improved and that she was sleeping nearly 8 hours per night. Her BDI score was 12, indicating normal mood. She was functioning much more effectively at work and could once again routinely perform her daily activities.

Ms. F continued her medication regimen for 8 months, after which she was lost to follow-up. At her most recent visit, her depression remained in remission. Her pain persisted, though at a lower intensity.

Related resources

  • Fishbain DA, Cutler R, Rosomoff HL, Rosomoff RS. Chronic pain-associated depression: antecedent or consequence of chronic pain? A review. Clin J Pain 1997;13:116-37.

Drug brand names

  • Amitriptyline • Elavil
  • Duloxetine • Cymbalta
  • Meclizine • Antivert
  • Modafinil • Provigil
  • Paroxetine • Paxil
  • Prochlorperazine • Compazine
  • Venlafaxine • Effexor
  • Zolpidem • Ambien

Disclosure

Dr. Fishbain is a consultant to Eli Lilly and Co. and a speaker for Purdue Pharmaceuticals.

References

1. Sood N, Treglia M, Obenchain RL, et al. Determinants of antidepressant outcome. Am J Manag Care 2000;6:1327-36.

2. American Psychiatric Association. Practice guideline for the treatment of patients with major depressive disorder (revision). Am J Psychiatry 2000;157(suppl 4):1-45.

3. Lin EH, Von Korff M, Katon W, et al. The role of the primary care physician in patients’ adherence to antidepressant therapy. Med Care 1996;33:67-74.

4. McBeth J, MacFarlane G, Silman A. Does chronic pain predict future psychological distress? Pain 2002;96:239-45.

5. Ohayon M, Schatzberg A. Using chronic pain to predict depressive morbidity in the general population. Arch Gen Psychiatry 2003;60:39-47.

6. Nakao M, Yamanaka G, Kuboki T. Major depression and somatic symptoms in a mind/body medicine clinic. Psychopathology 2001;34:230-5.

7. Posse M, Hallstrom T. Depressive disorders among somatizing patients in primary health care. Acta Psychiatr Scand 1998;98:187-92.

8. Paykel ES, Ramana R, Cooper Z, et al. Residual symptoms after partial remission: an important outcome in depression. Psychol Med 1995;25:1171-80.

9. Von Korff M, Simon G. The relationship between pain and depression. Br J Psychiatry 1996;168:101-8.

10. Goldberg DP, Bridges K. Somatic presentations of psychiatric illness in primary care setting. J Psychosom Res 1988;32:137-44.

11. Sartorius N. Physical symptoms of depression as a public health concern. J Clinical Psych 2003;64(suppl 7):3-4.

12. Keller MB. Long-term treatment strategies in affective disorders. Psychopharmacol Bull 2002;36(suppl 2):36-48.

13. Keller MB, Berndt ER. Depression treatment: a lifelong commitment? Psychopharmacol Bull 2002;36(suppl 2):133-41.

14. Segal Z, Vincent P, Levitt A. Efficacy of combined, sequential and crossover psychotherapy and pharmacotherapy in improving outcomes in depression. J Psychiatry Neurosci 2002;27:281-90.

15. Denninger JW, Henderson PO, Fallis K. The relationship between somatic symptoms and depression (presentation). Philadelphia, PA: American Psychiatric Association annual meeting, 2002.

16. Worthington J, Fava M, Davidson K, et al. Patterns of improvement in depressive symptoms with fluoxetine treatment. Psychopharmacol Bull 1995;31:223-6.

17. Fava M. Somatic symptoms, depression, and antidepressant treatment. J Clin Psychiatry 2002;63:4-305-7.

18. Fishbain DA, Cutler RB, Rosomoff HL, et al. Do antidepressants have an analgesic effect in psychogenic pain and somatoform pain disorder? A meta-analysis. Psychosom Med 1998;60:503-9.

19. O’Malley PG, Belden E, Tomkins G, et al. Treatment of fibromyalgia with antidepressants: a meta-analysis. J Gen Intern Med 2000;15:659-66.

20. Salerno SM, Browning R, Jackson JL. The effect of antidepressant treatment on chronic back pain: a meta-analysis. Arch Intern Med 2002;162:19-24.

21. Fishbain DA. Evidence-based data on pain relief with antidepressants. Ann Med 2000;32:305-16.

22. Fishbain DA, Cutler R, Rosomoff HL, et al. Evidence-based data from animal and human experimental studies on pain relief with antidepressants: a structured review. Pain Med 2000;1:310-16.

Article PDF
Author and Disclosure Information

David A. Fishbain, MD
Professor of psychiatry Adjunct professor of neurological surgery and anesthesiology University of Miami School of Medicine, Miami, FL

Issue
Current Psychiatry - 03(04)
Publications
Topics
Page Number
89-93
Sections
Author and Disclosure Information

David A. Fishbain, MD
Professor of psychiatry Adjunct professor of neurological surgery and anesthesiology University of Miami School of Medicine, Miami, FL

Author and Disclosure Information

David A. Fishbain, MD
Professor of psychiatry Adjunct professor of neurological surgery and anesthesiology University of Miami School of Medicine, Miami, FL

Article PDF
Article PDF

History: Initial symptoms

Ms. F, age 39, presented with depression, severe anxiety, and disturbed sleep. She denied psychiatric or medical history, but reported that her depressive symptoms hampered her performance at work and led to work-related stress.

Ms. F’s Beck Depression Inventory (BDI) score at baseline was 21, indicating moderate depression. The psychiatrist diagnosed her as having generalized anxiety disorder and adjustment disorder with depressed mood.

The patient was started on paroxetine, 10 mg/d. Five weeks later, her anxiety had decreased significantly and her BDI score had dropped to 10, indicating normal mood. Both the patient and clinician decided at this point that Ms. F had reached remission. The patient demanded that paroxetine be stopped, saying that she “does not like medication.” The psychiatrist reluctantly agreed.

Eight weeks later, during a follow-up examination, Ms. F complained of severely depressed mood with frequent crying spells. She complained of fatigue, nausea, headaches, decreased appetite, and dizziness. Her work performance, which had improved during the paroxetine trial, was again compromised. Her BDI score was 32, indicating severe depression.

Was the psychiatrist justified in stopping paroxetine therapy after 5 weeks?

Dr. Fishbain’s observations

Premature paroxetine discontinuation cannot be ruled out as a cause for Ms. F’s relapse. Sood et al1 found that duration of antidepressant therapy beyond treatment guidelines correlated with longer times to relapse.

By contrast, Ms. F’s initial therapy duration fell short of the 6 to 8 weeks recommended by the American Psychiatric Association.2

Patients commonly cite adverse events as a reason for wanting to stop antidepressant therapy.3 Ms. F reported no adverse effects, however; she said only that she did “not like medication.” Despite her insistence to the contrary, antidepressant therapy probably should not have been stopped.

Further history: A painful discovery

After questioning, Ms. F told the psychiatrist that she had been involved in a motor vehicle accident 2 weeks before the follow-up visit and had since been suffering lower back and neck pain.

After more questioning, Mr. F revealed that the pain was disrupting her sleep. She was getting about 4 hours of fragmented sleep per night, resulting in lack of energy during the day. The pain made it hard for her to sit, further impairing her work performance.

Ms. F was restarted on paroxetine, 10 mg/d titrated across 6 weeks to 60 mg/d for her depression, and zolpidem, 10 mg at bedtime, to help her sleep. After 6 weeks, her BDI score improved to 20, and she was less labile. Her depressive symptoms persisted, however, as did her pain, fatigue, headaches, nausea, dizziness, and sleep disturbances.

What role did Ms. F’s pain play in her relapse? How can clinicians detect somatic symptoms and gauge their effect on mood?

Dr. Fishbain’s observations

Pain most likely caused Ms. F’s depression relapse. McBeth et al4 have demonstrated that pain can contribute to depression’s development. In another study,5 43.4% of subjects who met criteria for major depression reported painful symptoms. The presence of a chronic painful condition was also found to contribute to major depression.5

Nakao et al6 screened 2,215 outpatients who were referred with mind/body complaints. Patients who were diagnosed with major depression had significantly higher rates of fatigue (86% vs. 65%), insomnia (79% vs. 58%), nausea/vomiting (51% vs. 40%), and low-back pain (36% vs. 24%) than those who were not. Within the major depression group, somatic symptoms were more abundant in patients with severe depressive episodes than in those with mild depressive episodes (5.8 vs. 3.7, P < 0.05).

Depression prevalence appears to increase when somatic symptoms are considered in the diagnosis. Posse and Hallstrom7 used a two-stage design to screen for depression. In the first stage, depression prevalence was 1.8%. In the second stage, 62 patients with high somatic complaint scores were re-evaluated. Of this group, 41 were diagnosed with a major depressive disorder or dysthymia.

Patients with continued pain after depression treatment are at high risk for depression recurrence.8 Diffuseness of pain and extent to which it interferes with daily activities strongly predict depression.9

Diagnostic challenges. Patients with depression often present to their primary care physicians with somatic rather than behavioral symptoms, making it hard for the family doctor to diagnose depression.10 By contrast, when presenting to a psychiatrist, depressed patients tend to discuss their emotional symptoms but not their physical complaints.11 This is because patients often:

  • attribute physical symptoms to an unrelated medical illness
  • consider aches and pains a normal part of aging
  • or are not aware that psychiatrists can treat physical symptoms.11

Psychiatrists in the past have emphasized emotional symptoms while barely addressing physical symptoms. This trend is changing, however, as the link between physical pain and depression has become clearer.

 

 

Be sure to include chronic pain or other somatic symptoms in the systems review. Screening tools such as the Visual Analogue Scale can measure pain intensity, while the Cornell Medical Index can uncover somatic symptoms. No all-inclusive tool exists to help detect depression-related somatic symptoms. however.

Should the psychiatrist continue to address Ms. F’s depressive symptoms, or should the focus shift to her somatic symptoms?

Dr. Fishbain’s observations

Patients who do not respond to depression treatment (ie, achieve >50% symptom reduction), or who respond without achieving remission, usually have residual physical symptoms—often fatigue, sleep disturbance, decreased appetite, anxiety, sexual dysfunction, and/or pain.12-14 Severe pain and other somatic symptoms are likely prolonging Ms. F’s depression, despite increased paroxetine dosages.

Paykel et al8 have associated residual depression symptoms with early relapse of depression. In their study, 94% of depressed patients with lingering depressive symptoms had mild to moderate physical symptoms. By contrast, degree of physical symptom improvement has been shown to correlate with likelihood of depression remission.15

Although emotional symptoms improve with antidepressants,16 some evidence17 indicates that physical symptoms associated with depression may be less responsive.

Also, because many psychiatrists have been taught to track emotional symptoms and only some physical symptoms, somatic symptoms of depression often are not targeted for treatment.17 Lack of rating scales to track somatic symptoms compounds this problem.17

Psychiatrists need to target both the physical and emotional symptoms of depression. When pain prolongs depression, it should be the primary target of antidepressant drug therapy (Algorithm).

To date, several meta-analyses18-20 have demonstrated that antidepressants have a separate analgesic effect on all forms of chronic pain. Evidence21,22 also indicates that the dualaction antidepressants—such as amitriptyline, bupropion, venlafaxine, and (awaiting FDA approval) duloxetine—have a more-consistent analgesic effect than do the serotonin reuptake inhibitors.

The analgesic effects of bupropion, duloxetine, and venlafaxine have not been compared with those of tricyclics or other older antidepressants. If one of the newer dualaction antidepressants does not reduce somatic conditions or produce an adequate response, consider switching to a tricyclic.

Treatment: No pain, some gain

Another psychiatrist who specializes in pain medicine targeted some of Ms. F’s somatic symptoms with antidepressants. Paroxetine and zolpidem were discontinued and the patient was started on:

  • venlafaxine, 37.5 mg bid, titrated to 225 mg/d across 2 weeks. Because of its activating properties, venlafaxine was chosen to address Ms. F’s pain and daytime fatigue.
  • amitriptyline, 50 mg at bedtime nightly, to promote sleep
  • prochlorperazine, 10 mg as needed, and meclizine, 25 mg as needed, to treat her nausea and dizziness, respectively.

Algorithm Suggested drug treatment of depression with somatic symptoms



Ms. F also was advised to take an abortive migraine compound (Midrin, 2 tablets at headache onset and 1 additional tablet every half-hour as needed, maximum 5 tablets per day). Mr. F’s primary care physician also referred her to a physical therapist to treat her neck and low-back pain; workup revealed no surgically treatable problem.

Four weeks later, Ms. F reported that her somatic symptoms significantly improved and that she was sleeping nearly 8 hours per night. Her BDI score was 12, indicating normal mood. She was functioning much more effectively at work and could once again routinely perform her daily activities.

Ms. F continued her medication regimen for 8 months, after which she was lost to follow-up. At her most recent visit, her depression remained in remission. Her pain persisted, though at a lower intensity.

Related resources

  • Fishbain DA, Cutler R, Rosomoff HL, Rosomoff RS. Chronic pain-associated depression: antecedent or consequence of chronic pain? A review. Clin J Pain 1997;13:116-37.

Drug brand names

  • Amitriptyline • Elavil
  • Duloxetine • Cymbalta
  • Meclizine • Antivert
  • Modafinil • Provigil
  • Paroxetine • Paxil
  • Prochlorperazine • Compazine
  • Venlafaxine • Effexor
  • Zolpidem • Ambien

Disclosure

Dr. Fishbain is a consultant to Eli Lilly and Co. and a speaker for Purdue Pharmaceuticals.

History: Initial symptoms

Ms. F, age 39, presented with depression, severe anxiety, and disturbed sleep. She denied psychiatric or medical history, but reported that her depressive symptoms hampered her performance at work and led to work-related stress.

Ms. F’s Beck Depression Inventory (BDI) score at baseline was 21, indicating moderate depression. The psychiatrist diagnosed her as having generalized anxiety disorder and adjustment disorder with depressed mood.

The patient was started on paroxetine, 10 mg/d. Five weeks later, her anxiety had decreased significantly and her BDI score had dropped to 10, indicating normal mood. Both the patient and clinician decided at this point that Ms. F had reached remission. The patient demanded that paroxetine be stopped, saying that she “does not like medication.” The psychiatrist reluctantly agreed.

Eight weeks later, during a follow-up examination, Ms. F complained of severely depressed mood with frequent crying spells. She complained of fatigue, nausea, headaches, decreased appetite, and dizziness. Her work performance, which had improved during the paroxetine trial, was again compromised. Her BDI score was 32, indicating severe depression.

Was the psychiatrist justified in stopping paroxetine therapy after 5 weeks?

Dr. Fishbain’s observations

Premature paroxetine discontinuation cannot be ruled out as a cause for Ms. F’s relapse. Sood et al1 found that duration of antidepressant therapy beyond treatment guidelines correlated with longer times to relapse.

By contrast, Ms. F’s initial therapy duration fell short of the 6 to 8 weeks recommended by the American Psychiatric Association.2

Patients commonly cite adverse events as a reason for wanting to stop antidepressant therapy.3 Ms. F reported no adverse effects, however; she said only that she did “not like medication.” Despite her insistence to the contrary, antidepressant therapy probably should not have been stopped.

Further history: A painful discovery

After questioning, Ms. F told the psychiatrist that she had been involved in a motor vehicle accident 2 weeks before the follow-up visit and had since been suffering lower back and neck pain.

After more questioning, Mr. F revealed that the pain was disrupting her sleep. She was getting about 4 hours of fragmented sleep per night, resulting in lack of energy during the day. The pain made it hard for her to sit, further impairing her work performance.

Ms. F was restarted on paroxetine, 10 mg/d titrated across 6 weeks to 60 mg/d for her depression, and zolpidem, 10 mg at bedtime, to help her sleep. After 6 weeks, her BDI score improved to 20, and she was less labile. Her depressive symptoms persisted, however, as did her pain, fatigue, headaches, nausea, dizziness, and sleep disturbances.

What role did Ms. F’s pain play in her relapse? How can clinicians detect somatic symptoms and gauge their effect on mood?

Dr. Fishbain’s observations

Pain most likely caused Ms. F’s depression relapse. McBeth et al4 have demonstrated that pain can contribute to depression’s development. In another study,5 43.4% of subjects who met criteria for major depression reported painful symptoms. The presence of a chronic painful condition was also found to contribute to major depression.5

Nakao et al6 screened 2,215 outpatients who were referred with mind/body complaints. Patients who were diagnosed with major depression had significantly higher rates of fatigue (86% vs. 65%), insomnia (79% vs. 58%), nausea/vomiting (51% vs. 40%), and low-back pain (36% vs. 24%) than those who were not. Within the major depression group, somatic symptoms were more abundant in patients with severe depressive episodes than in those with mild depressive episodes (5.8 vs. 3.7, P < 0.05).

Depression prevalence appears to increase when somatic symptoms are considered in the diagnosis. Posse and Hallstrom7 used a two-stage design to screen for depression. In the first stage, depression prevalence was 1.8%. In the second stage, 62 patients with high somatic complaint scores were re-evaluated. Of this group, 41 were diagnosed with a major depressive disorder or dysthymia.

Patients with continued pain after depression treatment are at high risk for depression recurrence.8 Diffuseness of pain and extent to which it interferes with daily activities strongly predict depression.9

Diagnostic challenges. Patients with depression often present to their primary care physicians with somatic rather than behavioral symptoms, making it hard for the family doctor to diagnose depression.10 By contrast, when presenting to a psychiatrist, depressed patients tend to discuss their emotional symptoms but not their physical complaints.11 This is because patients often:

  • attribute physical symptoms to an unrelated medical illness
  • consider aches and pains a normal part of aging
  • or are not aware that psychiatrists can treat physical symptoms.11

Psychiatrists in the past have emphasized emotional symptoms while barely addressing physical symptoms. This trend is changing, however, as the link between physical pain and depression has become clearer.

 

 

Be sure to include chronic pain or other somatic symptoms in the systems review. Screening tools such as the Visual Analogue Scale can measure pain intensity, while the Cornell Medical Index can uncover somatic symptoms. No all-inclusive tool exists to help detect depression-related somatic symptoms. however.

Should the psychiatrist continue to address Ms. F’s depressive symptoms, or should the focus shift to her somatic symptoms?

Dr. Fishbain’s observations

Patients who do not respond to depression treatment (ie, achieve >50% symptom reduction), or who respond without achieving remission, usually have residual physical symptoms—often fatigue, sleep disturbance, decreased appetite, anxiety, sexual dysfunction, and/or pain.12-14 Severe pain and other somatic symptoms are likely prolonging Ms. F’s depression, despite increased paroxetine dosages.

Paykel et al8 have associated residual depression symptoms with early relapse of depression. In their study, 94% of depressed patients with lingering depressive symptoms had mild to moderate physical symptoms. By contrast, degree of physical symptom improvement has been shown to correlate with likelihood of depression remission.15

Although emotional symptoms improve with antidepressants,16 some evidence17 indicates that physical symptoms associated with depression may be less responsive.

Also, because many psychiatrists have been taught to track emotional symptoms and only some physical symptoms, somatic symptoms of depression often are not targeted for treatment.17 Lack of rating scales to track somatic symptoms compounds this problem.17

Psychiatrists need to target both the physical and emotional symptoms of depression. When pain prolongs depression, it should be the primary target of antidepressant drug therapy (Algorithm).

To date, several meta-analyses18-20 have demonstrated that antidepressants have a separate analgesic effect on all forms of chronic pain. Evidence21,22 also indicates that the dualaction antidepressants—such as amitriptyline, bupropion, venlafaxine, and (awaiting FDA approval) duloxetine—have a more-consistent analgesic effect than do the serotonin reuptake inhibitors.

The analgesic effects of bupropion, duloxetine, and venlafaxine have not been compared with those of tricyclics or other older antidepressants. If one of the newer dualaction antidepressants does not reduce somatic conditions or produce an adequate response, consider switching to a tricyclic.

Treatment: No pain, some gain

Another psychiatrist who specializes in pain medicine targeted some of Ms. F’s somatic symptoms with antidepressants. Paroxetine and zolpidem were discontinued and the patient was started on:

  • venlafaxine, 37.5 mg bid, titrated to 225 mg/d across 2 weeks. Because of its activating properties, venlafaxine was chosen to address Ms. F’s pain and daytime fatigue.
  • amitriptyline, 50 mg at bedtime nightly, to promote sleep
  • prochlorperazine, 10 mg as needed, and meclizine, 25 mg as needed, to treat her nausea and dizziness, respectively.

Algorithm Suggested drug treatment of depression with somatic symptoms



Ms. F also was advised to take an abortive migraine compound (Midrin, 2 tablets at headache onset and 1 additional tablet every half-hour as needed, maximum 5 tablets per day). Mr. F’s primary care physician also referred her to a physical therapist to treat her neck and low-back pain; workup revealed no surgically treatable problem.

Four weeks later, Ms. F reported that her somatic symptoms significantly improved and that she was sleeping nearly 8 hours per night. Her BDI score was 12, indicating normal mood. She was functioning much more effectively at work and could once again routinely perform her daily activities.

Ms. F continued her medication regimen for 8 months, after which she was lost to follow-up. At her most recent visit, her depression remained in remission. Her pain persisted, though at a lower intensity.

Related resources

  • Fishbain DA, Cutler R, Rosomoff HL, Rosomoff RS. Chronic pain-associated depression: antecedent or consequence of chronic pain? A review. Clin J Pain 1997;13:116-37.

Drug brand names

  • Amitriptyline • Elavil
  • Duloxetine • Cymbalta
  • Meclizine • Antivert
  • Modafinil • Provigil
  • Paroxetine • Paxil
  • Prochlorperazine • Compazine
  • Venlafaxine • Effexor
  • Zolpidem • Ambien

Disclosure

Dr. Fishbain is a consultant to Eli Lilly and Co. and a speaker for Purdue Pharmaceuticals.

References

1. Sood N, Treglia M, Obenchain RL, et al. Determinants of antidepressant outcome. Am J Manag Care 2000;6:1327-36.

2. American Psychiatric Association. Practice guideline for the treatment of patients with major depressive disorder (revision). Am J Psychiatry 2000;157(suppl 4):1-45.

3. Lin EH, Von Korff M, Katon W, et al. The role of the primary care physician in patients’ adherence to antidepressant therapy. Med Care 1996;33:67-74.

4. McBeth J, MacFarlane G, Silman A. Does chronic pain predict future psychological distress? Pain 2002;96:239-45.

5. Ohayon M, Schatzberg A. Using chronic pain to predict depressive morbidity in the general population. Arch Gen Psychiatry 2003;60:39-47.

6. Nakao M, Yamanaka G, Kuboki T. Major depression and somatic symptoms in a mind/body medicine clinic. Psychopathology 2001;34:230-5.

7. Posse M, Hallstrom T. Depressive disorders among somatizing patients in primary health care. Acta Psychiatr Scand 1998;98:187-92.

8. Paykel ES, Ramana R, Cooper Z, et al. Residual symptoms after partial remission: an important outcome in depression. Psychol Med 1995;25:1171-80.

9. Von Korff M, Simon G. The relationship between pain and depression. Br J Psychiatry 1996;168:101-8.

10. Goldberg DP, Bridges K. Somatic presentations of psychiatric illness in primary care setting. J Psychosom Res 1988;32:137-44.

11. Sartorius N. Physical symptoms of depression as a public health concern. J Clinical Psych 2003;64(suppl 7):3-4.

12. Keller MB. Long-term treatment strategies in affective disorders. Psychopharmacol Bull 2002;36(suppl 2):36-48.

13. Keller MB, Berndt ER. Depression treatment: a lifelong commitment? Psychopharmacol Bull 2002;36(suppl 2):133-41.

14. Segal Z, Vincent P, Levitt A. Efficacy of combined, sequential and crossover psychotherapy and pharmacotherapy in improving outcomes in depression. J Psychiatry Neurosci 2002;27:281-90.

15. Denninger JW, Henderson PO, Fallis K. The relationship between somatic symptoms and depression (presentation). Philadelphia, PA: American Psychiatric Association annual meeting, 2002.

16. Worthington J, Fava M, Davidson K, et al. Patterns of improvement in depressive symptoms with fluoxetine treatment. Psychopharmacol Bull 1995;31:223-6.

17. Fava M. Somatic symptoms, depression, and antidepressant treatment. J Clin Psychiatry 2002;63:4-305-7.

18. Fishbain DA, Cutler RB, Rosomoff HL, et al. Do antidepressants have an analgesic effect in psychogenic pain and somatoform pain disorder? A meta-analysis. Psychosom Med 1998;60:503-9.

19. O’Malley PG, Belden E, Tomkins G, et al. Treatment of fibromyalgia with antidepressants: a meta-analysis. J Gen Intern Med 2000;15:659-66.

20. Salerno SM, Browning R, Jackson JL. The effect of antidepressant treatment on chronic back pain: a meta-analysis. Arch Intern Med 2002;162:19-24.

21. Fishbain DA. Evidence-based data on pain relief with antidepressants. Ann Med 2000;32:305-16.

22. Fishbain DA, Cutler R, Rosomoff HL, et al. Evidence-based data from animal and human experimental studies on pain relief with antidepressants: a structured review. Pain Med 2000;1:310-16.

References

1. Sood N, Treglia M, Obenchain RL, et al. Determinants of antidepressant outcome. Am J Manag Care 2000;6:1327-36.

2. American Psychiatric Association. Practice guideline for the treatment of patients with major depressive disorder (revision). Am J Psychiatry 2000;157(suppl 4):1-45.

3. Lin EH, Von Korff M, Katon W, et al. The role of the primary care physician in patients’ adherence to antidepressant therapy. Med Care 1996;33:67-74.

4. McBeth J, MacFarlane G, Silman A. Does chronic pain predict future psychological distress? Pain 2002;96:239-45.

5. Ohayon M, Schatzberg A. Using chronic pain to predict depressive morbidity in the general population. Arch Gen Psychiatry 2003;60:39-47.

6. Nakao M, Yamanaka G, Kuboki T. Major depression and somatic symptoms in a mind/body medicine clinic. Psychopathology 2001;34:230-5.

7. Posse M, Hallstrom T. Depressive disorders among somatizing patients in primary health care. Acta Psychiatr Scand 1998;98:187-92.

8. Paykel ES, Ramana R, Cooper Z, et al. Residual symptoms after partial remission: an important outcome in depression. Psychol Med 1995;25:1171-80.

9. Von Korff M, Simon G. The relationship between pain and depression. Br J Psychiatry 1996;168:101-8.

10. Goldberg DP, Bridges K. Somatic presentations of psychiatric illness in primary care setting. J Psychosom Res 1988;32:137-44.

11. Sartorius N. Physical symptoms of depression as a public health concern. J Clinical Psych 2003;64(suppl 7):3-4.

12. Keller MB. Long-term treatment strategies in affective disorders. Psychopharmacol Bull 2002;36(suppl 2):36-48.

13. Keller MB, Berndt ER. Depression treatment: a lifelong commitment? Psychopharmacol Bull 2002;36(suppl 2):133-41.

14. Segal Z, Vincent P, Levitt A. Efficacy of combined, sequential and crossover psychotherapy and pharmacotherapy in improving outcomes in depression. J Psychiatry Neurosci 2002;27:281-90.

15. Denninger JW, Henderson PO, Fallis K. The relationship between somatic symptoms and depression (presentation). Philadelphia, PA: American Psychiatric Association annual meeting, 2002.

16. Worthington J, Fava M, Davidson K, et al. Patterns of improvement in depressive symptoms with fluoxetine treatment. Psychopharmacol Bull 1995;31:223-6.

17. Fava M. Somatic symptoms, depression, and antidepressant treatment. J Clin Psychiatry 2002;63:4-305-7.

18. Fishbain DA, Cutler RB, Rosomoff HL, et al. Do antidepressants have an analgesic effect in psychogenic pain and somatoform pain disorder? A meta-analysis. Psychosom Med 1998;60:503-9.

19. O’Malley PG, Belden E, Tomkins G, et al. Treatment of fibromyalgia with antidepressants: a meta-analysis. J Gen Intern Med 2000;15:659-66.

20. Salerno SM, Browning R, Jackson JL. The effect of antidepressant treatment on chronic back pain: a meta-analysis. Arch Intern Med 2002;162:19-24.

21. Fishbain DA. Evidence-based data on pain relief with antidepressants. Ann Med 2000;32:305-16.

22. Fishbain DA, Cutler R, Rosomoff HL, et al. Evidence-based data from animal and human experimental studies on pain relief with antidepressants: a structured review. Pain Med 2000;1:310-16.

Issue
Current Psychiatry - 03(04)
Issue
Current Psychiatry - 03(04)
Page Number
89-93
Page Number
89-93
Publications
Publications
Topics
Article Type
Display Headline
The painful truth about depression
Display Headline
The painful truth about depression
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Update on eating disorders: Binge-eating disorder

Article Type
Changed
Tue, 12/11/2018 - 15:10
Display Headline
Update on eating disorders: Binge-eating disorder

Clinical snapshot of BED

Managing patients with binge-eating disorder (BED) often requires behavioral, medical, and psychiatric interventions.

These patients suffer from recurrent episodes of distressing, uncontrollable overeating, but they do not purge or show other compensatory weight-loss behaviors common to bulimia nervosa1 and anorexia nervosa.2-10 As a result, they are often overweight or obese and may have obesity-related illnesses, such as hypertension or type 2 diabetes. Mild to severe depression—unipolar or bipolar—is a common psychopathology.

Because no one treatment fits all patients with binge eating disorder, their management usually requires an individualized program of:

  • behavioral weight control
  • psychotherapy
  • and sometimes medications.

In our weight management clinic, we consider medication options based on patient preference and whether BED is uncomplicated (Figure 1) or coexists with a mood disorder (Figure 2).

This article presents the evidence on which we base our comprehensive approach. General psychiatrists with knowledge of BED can treat patients with this eating disorder, although complicated cases may require referral for specialized treatment.

Figure 1 Medication options for uncomplicated BED


Clinical characteristics

Psychiatric comorbidity. BED often occurs in patients with mood, anxiety, substance-abuse, impulsecontrol, and personality disorders.4,6,10-12 Mood disorder—particularly depression—appears to be the most common comorbidity. BED can occur with bipolar disorder12—a comorbidity that in our experience is underrecognized both clinically and in the literature.

Patients with BED and bipolar disorder show increased impulsivity and mood lability. As bipolar II disorder and other “soft-spectrum” forms are more common than bipolar I disorder, BED is also more likely to occur with hypomania than mania.

Overweight. Not surprisingly, BED is associated with overweight and obesity.5,8,9,11 Not all patients with BED are overweight or obese, but most who participate in clinical trials of BED treatments are at least overweight. BED has been reported in up to:

  • 30% of participants in weight-loss programs7
  • 70% of participants in groups such as Overeaters Anonymous
  • 50% of patients who seek bariatric surgery.5

In our experience, patients are often more distressed by their weight than by their binge eating, depression, or anxiety. Indeed, overweight and obesity are the usual reasons patients with BED present for treatment at our center.

Diagnosis. BED’s validity as a clinical diagnosis has been controversial since the disorder was first included in DSM-IV (Table 1).3 Debate continues about some definitions in the DSM criteria, including what amount of food is “definitely larger” than most people would eat and what is “loss of control over eating.”

Nevertheless, screening for BED is relatively easy. Clinicians may use the eating disorder section of the Structured Clinical Interview for DSM-IV or the Eating Disorders Examination. Alternatively, simply ask patients if they have episodes of uncontrollable overeating, during which they eat unusually large amounts of food and their eating feels out of control.

Course. BED begins in adolescence or adulthood. Disease course is variable, with periods of remission, recurrence, and chronicity.6,7,10 Interestingly, one prospective study showed that even if the binge eating resolves, persons may still develop obesity.13

Prevalence. BED affects 1.5% to 3% of the U.S. population. It is more common in women than men, equally prevalent in whites and blacks, and more prevalent than anorexia nervosa and bulimia nervosa combined.11,14 Subthreshold BED—such as obesity with infrequent or nondistressing binge eating—appears to be much more common,10 although no data are available.

Theories of binge eating

BED’s cause is unknown, but biological, familial, and psychosocial factors have been implicated.

Biological factors. The neurotransmitters serotonin (5-HT) and dopamine—as well as various peptides—have been shown to help regulate feeding behavior.10

Table 1

Diagnostic criteria for binge-eating disorder*

  1. Recurrent episodes of binge eating are characterized by both of the following:
    • eating in a discrete period of time (as within any 2 hours) an amount of food that is definitely larger than what most people would eat in a similar period under similar circumstances
    • a sense of lack of control over eating during the episode (a feeling that one cannot stop eating or control what or how much one is eating)
  2. The binge-eating episodes are associated with three or more of the following:
    • eating much more rapidly than normal
    • eating until feeling uncomfortably full
    • eating large amounts of food when not feeling physically hungry
    • eating alone because of being embarrassed by how much one is eating
    • feeling disgusted with oneself, depressed, or very guilty after overeating
  3. Marked distress regarding binge eating is present.
  4. The binge eating occurs, on average, at least 2 days a week for 6 months.
  5. The binge eating is not associated with the regular use of inappropriate compensatory behaviors (purging, fasting, excessive exercise) and does not occur exclusively during the course of anorexia nervosa or bulimia nervosa.
* Research criteria, DSM-IV-TR appendix B.
Source: Reprinted with permission from the Diagnostic and statistical manual of mental disorders, 4th edition, text revision. Copyright 2000. American Psychiatric Association.
 

 

Serotonin. Reduced 5-HT transporter binding has been shown in obese women with BED.15 Their 5-HT binding improved and binge eating subsided with group psychotherapy and fluoxetine, although the women continued to gain weight.

Figure 2 Medication options for BED with obesity and a mood disorder*



Dopamine. Obese patients who compulsively overeat may have lower levels of dopamine D2 receptors than do normal-weight controls.16

Genetic factors. In severely obese patients (body mass index 44±2), those with a DSM-IV diagnosis of BED exhibited mutations of the melanocortin 4 receptor gene, which affects the anorectic properties of alpha melanocyte-stimulating hormone.17

Familial factors associated with BED include parental depression and obesity.18

Psychosocial correlates include physical and sexual abuse, bullying by peers, and discrimination because of being overweight.19

Treatment recommendations

Few systematic studies have examined BED treatment. Emerging research suggests that behavioral weight-loss treatment, specialized psychotherapies, and medications may be effective in some patients with BED.4,6,8

Behavioral weight-loss treatment’s main goal is to manage the patient’s weight with a lower-calorie, healthy diet and to increase exercise.20,21

Over the short term (<1 year), behavioral weight-loss treatment produces similar weight loss in obese patients with or without BED; long-term results in both groups, however, have not been satisfactory.20,21 No studies have examined the efficacy of specialized diets (such as low-carbohydrate regimens) in patients with BED.

Specialized psychotherapy’s goal is to modify bingeeating behavior with behavioral self-management strategies, reducing interpersonal dysfunction and stress, and/or managing affective dysregulation.

Cognitive-behavioral therapy (CBT) and interpersonal therapy (IPT) have been effective in reducing binge eating, both acutely and for up to 12 months4,20-24 but less effective in achieving and maintaining weight loss. Patients who achieve remission in binge eating after undergoing CBT or IPI often experience modest but stable weight loss.20-22 For example, in a comparison study of CBT and IPT:

  • After 20 weekly sessions, patients whose binge eating was in remission lost weight (mean body mass index [BMI] −0.5 ± 1.5 kg/m2), whereas those who continued to binge gained weight (mean BMI +0.4 ±2.0 kg/m2).
  • At 12 months’ follow-up, patients still in remission continued to lose weight (mean BMI −1.0 ± 3.0 kg/m2), whereas those no longer in remission gained weight (mean BMI +0.7 ±2.9 kg/m2[P = 0.01]).22

Self-help and dialectical behavioral therapy (DBT) may also help reduce binge eating in BED. As with CBT and IPT, they are less effective in weight loss. In the only controlled study of DBT,24 patients achieved an average 2.5-lb weight loss after 20 weeks of DBT, compared with an average 0.6-lb weight gain in the control group. This difference was not significant, and the report did not include data on weight loss maintenance.

In summary, CBT may be more effective than behavioral weight loss treatment for reducing binge eating, but behavioral weight loss is more effective for weight loss.

Medications for BED

Medications that have been tried for BED include antidepressants, appetite suppressants, and anticonvulsants.25,26 Antidepressants are used to treat BED because:

  • BED is often associated with depressive symptoms and disorders.
  • BED is related to bulimia nervosa, and placebo-controlled trials have shown that the binge eating of bulimia nervosa responds to several classes of antidepressants. The selective serotonin reuptake inhibitor (SSRI) fluoxetine is the only medication indicated for treating any eating disorder (bulimia nervosa).
  • Bupropion and venlafaxine—a serotonin-norepinephrine reuptake inhibitor (SNRI)—have weight-loss properties.

SSRIs are the most extensively studied antidepressants for treating BED. SSRIs have weightloss properties, but only short term.25-26 Citalopram, fluoxetine, fluvoxamine, and sertraline have reduced binge eating and body weight more effectively than placebo during 6 to 9 weeks of treatment (Table 2).25-26 However, one controlled study23 showed that fluoxetine was not significantly more effective than placebo in reducing binge frequency or body weight after 16 weeks.

TCAs. Studies of tricyclic antidepressants (TCAs) for BED are sparse, and results have been mixed. In one trial, imipramine was similar to placebo in reducing binge frequency and body weight. In a placebo-controlled study of patients with nonpurging bulimia nervosa, desipramine reduced binge eating but had no effect on body weight.25,26

Table 2

Drug therapies shown to be effective for BED*

MedicationBinge eatingWeightDepressionStudy sizeDuration (weeks)Dosage (mg/d)
Antidepressants
Citalopram++38620 to 60
Fluoxetine †+++60620 to 80
Fluvoxamine++85950 to 300
Sertraline++34650 to 200
Appetite suppressant
Sibutramine+++601215
Anticonvulsant
Topiramate++611450 to 600
+ Improvement
− No improvement
* Randomized, controlled trials. Antidepressants were studied in patients with BED; sibutramine and topiramate were studied in patients with BED and associated obesity.
† One 16-week trial of fluoxetine for BED (reference 23) did not show statistically significant differences in post-treatment binge frequency or body-mass index.

Venlafaxine. In a retrospective review of 35 consecutive obese women with BED, venlafaxine, mean 222 mg/d for 28 to 300 days (median 120 days), reduced binge eating, body weight, and depressive symptoms.27

 

 

Bupropion has been more effective than placebo for treating:

  • uncomplicated obesity (short- and long-term)
  • obesity associated with depressive symptoms
  • bulimia nervosa (although bupropion is contraindicated in these patients because of seizure risk).26,28,29

No controlled trials have studied bupropion for BED. When using dosages effective in depressive disorders, we find bupropion helpful in reducing binge eating, body weight, and depressive symptoms in BED patients.

Appetite suppressants decrease appetite and weight, may increase satiety, and may reduce depressive symptoms.

Sibutramine—a serotonin, norepinephrine, and dopamine reuptake inhibitor indicated for managing obesity—has been reported effective in BED in a 12-week, randomized, double-blind, placebo-controlled trial. A 15-mg/d dosage reduced binge frequency, body weight, and depressive symptoms more effectively than placebo in 60 obese patients with BED.30 Most-frequent adverse effects (dry mouth and constipation) were mild and benign, and no significant complications were observed.

Sibutramine’s mechanism of action in BED is unknown. However, it suppressed food intake during binge-eating episodes in patients with BED in a randomized, controlled, cross-over laboratory study.31

Orlistat. We know of no published controlled studies of the lipase inhibitor orlistat in treating BED. In our experience, some patients do well with this agent, though we have observed infrequent purging episodes with it in patients with BED.

With orlistat, 120 mg tid, our BED patients have experienced weight loss comparable to that seen in uncomplicated obesity at similar dosages. Orlistat seems most effective for:

  • patients whose binge eating is in remission
  • those who responded to behavioral weightloss treatment, a psychological treatment, or another medication.

Anticonvulsants such as topiramate and zonisamide have been shown effective in treating obesity32,33 and are sometimes used to treat BED. Obese BED patients with mood disorders often do best with psychotherapy plus medication

Topiramate at dosages of 50 to 600 mg/d (median 212 mg/d) reduced binge-eating frequency, obsessive-compulsive features of binge eating, and body weight more effectively than placebo in a 14-week study of 61 obese patients with BED. These effects were maintained across 48 weeks in an open-label extension trial.34

Zonisamide, mean 513 mg/d, produced similar results during a prospective, open-label, 12-week trial in 15 patients with BED.35 A controlled trial to replicate these findings is ongoing.

BED may respond to anticonvulsant therapy for several reasons:

  • Some anticonvulsants are effective in treating bipolar disorder, which may occur with BED.12
  • Some anticonvulsants have shown benefit in conditions associated with pathologic impulsivity, such as substance abuse, impulse-control, and cluster B disorders.10

Growing evidence shows that bulimia nervosa and BED may be associated with pathologic impulsivity.

Combination therapies are generally more effective than monotherapies in patients with mood disorders, uncomplicated obesity, and possibly bulimia nervosa. Even so, few trials have systematically studied combination therapy in managing patients with BED.

Two studies compared psychotherapy and antidepressants alone and in combination in treating BED.21,23 Both showed that CBT alone was more effective in decreasing binge frequency than desipramine alone,21 fluoxetine alone,23 and the combination of CBT and medication. On the other hand, patients who took desipramine either alone or in combination experienced a greater degree of weight loss than those who did not take desipramine.21

In another combination therapy, exercise has been shown to be an effective adjunct to CBT in maintenance treatment of obese women with BED.36

No studies have compared behavioral weight management or a specialized psychotherapy in combination with an antiobesity drug or a weight-loss anticonvulsant in treating BED.

Treatment recommendations

In our experience, BED patients—particularly those with obesity and psychopathology—often do best with some combination of psychological treatment and medication:

  • The psychological component may be behavioral weight-loss treatment, a specialized psychotherapy such as CBT or IPT, or some combination of behavioral weight-loss treatment and specialized psychotherapy.
  • The medication component may consist of an antidepressant, anticonvulsant, antiobesity drug, or multiple drugs (such as an SSRI or sibutramine with topiramate for BED with major depression, or topiramate with lithium for BED with bipolar disorder).

Although combination therapies may be optimal for some patients, this approach remains unproven in controlled trials.

Patient preference. In addition to comorbidities, patient preference is an important consideration when choosing BED treatments. We determine our patients’ preferences by educating them as much as possible about their options. We explain the benefits and weaknesses of all treatments and encourage them to participate in forming their individualized treatment plans.

Patients sometimes have strong treatment preferences. Some prefer psychological treatments, whereas others prefer medications. Working with patient preferences enhances treatment adherence. For example, patients who fail a preferred treatment are often more willing to adhere to another treatment modality about which they initially were skeptical.

 

 

Related resources

  • Bray GA, Bouchard C (eds). Handbook of obesity: clinical applications (2nd ed). New York, NY: Marcel Dekker, 2004.
  • Cooper Z, Zairburn CG, Hawker DM. Cognitive behavioral treatment of obesity. A clinician’s guide. New York: Guilford Press, 2003.
  • Carter WP, Hudson JI, Lalonde JK, et al. Pharmacologic treatment of binge eating disorder. Int J Eat Disord 2003;34(suppl):S74-88.

Drug brand names

  • Bupropion • Wellbutrin
  • Citalopram • Celexa
  • Desipramine • Norpramin
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Imipramine • Tofranil
  • Lamotrigine • Lamictal
  • Lithium • Eskalith, others
  • Orlistat • Xenical
  • Sertraline • Zoloft
  • Sibutramine • Meridia
  • Topiramate • Topamax
  • Venlafaxine • Effexor
  • Zonisamide • Zonegran

Disclosure

Dr. Kotwal receives grant support from Elan Corporation and is a speaker for Ortho-McNeil Pharmaceutical and Pfizer Inc.

Dr. Kaneria and Ms. Guerdjikova report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. McElroy is a consultant to Abbott Laboratories, Bristol-Myers Squibb Co., Elan Corporation, GlaxoSmithKline, Janssen Pharmaceutica, Eli Lilly and Co., and Ortho-McNeil Pharmaceutical. She receives grant/research support from Elan Pharmaceuticals, Forest Pharmaceuticals, Merck & Co., Ortho-McNeil Pharmaceutical, and Sanofi-Synthelabo and is a speaker for Eli Lilly and Co. and Ortho-McNeil Pharmaceutical.

References

1. Pope HG, Hudson JI. Bulimia nervosa: Persistent disorders requires equally persistent treatment. Current Psychiatry 2004;3(1):13-22.

2. Halmi KA. Anorexia nervosa: Dual therapy can bring patients back from the brink. Current Psychiatry 2004;3(3):39-56.

3. American Psychiatric Association. Diagnostic and statistical manual of mental disorders (4th ed). Washington, DC: American Psychiatric Association, 1994.

4. Agras WS. Treatment of binge eating disorder. In: Gabbard GO (ed). Treatments of psychiatric disorders (3rd ed). Washington, DC: American Psychiatric Press, 2001;2209-19.

5. de Zwaan M. Binge eating disorder and obesity. Int J Obes Relat Metab Disord 2001;25(suppl 1):S51-5.

6. Dingemans AE, Bruna MJ, van Furth EF. Binge eating disorder: a review. Int J Obes Relat Metab Disord 2002;26:299-307.

7. Fairburn CG, Harrison PJ. Eating disorders. Lancet 2003;361:407-16.

8. Walsh BT (ed). The current status of binge eating disorder. Int J Eat Disord 2003;34(suppl):S1-120.

9. Devlin MJ, Goldfein JA, Dobrow I. What is this thing called BED? Current status of binge eating disorder nosology. Int J Eat Disord 2003;34(suppl):S2-18.

10. McElroy SL, Kotwal R. Binge eating. In: Hollander E, Stein D (eds). Handbook of impulse control disorders Washington, DC: American Psychiatric Press (in press).

11. Smith DE, Marcus MD, Lewis CE, et al. Prevalence of binge eating disorder, obesity, and depression in a biracial cohort of young adults. Ann Behav Med 1998;20:227-32.

12. Kruger S, Shugar G, Cooke RG. Comorbidity of binge eating disorder and the partial binge eating syndrome with bipolar disorder. Int J Eat Disord 1996;19:45-52.

13. Fairburn CG, Cooper Z, Doll H, et al. The natural course of bulimia nervosa and binge eating disorder in young women. Arch Gen Psychiatry 2000;57:659-65.

14. Striegel-Moore RH, Franko DL. Epidemiology of binge eating disorder. Int J Eat Disord 2003;34(suppl):S19-29.

15. Tammela LI, Rissanen A, Kuikka JT, et al. Treatment improves serotonin transporter binding and reduces binge eating. Psychopharmacology (Berl) 2003;170:89-93.

16. Wang GJ, Volkow ND, Logan J, et al. Brain dopamine and obesity. Lancet 2001;357:354-7.

17. Branson R, Potoczna N, Kral JG, et al. Binge eating as a major phenotype of melanocortin 4 receptor gene mutations. N Engl J Med 2003;348:1096-103.

18. Fairburn CG, Doll HA, Welch SL, et al. Risk factors for binge eating disorder: a community-based, case-control study. Arch Gen Psychiatry 1998;55:425-32.

19. Striegel-Moore RH, Dohm FA, Pike KM, et al. Abuse, bullying, and discrimination as risk factors for binge eating disorder. Am J Psychiatry 2002;159:1902-7.

20. Wonderlich SA, de Zwaan M, Mitchell JE, et al. Psychological and dietary treatments of binge eating disorder: conceptual implications. Int J Eat Disord 2003;34(suppl):S58-78.

21. Agras WS, Telch DF, Arnow B, et al. Weight loss, cognitive-behavioral, and desipramine treatments in binge eating disorder. An additive design. Behav Ther 1994;25:225-38.

22. Wilfley DE, Welch RR, Stein RI, et al. A randomized comparison of group cognitive-behavioral therapy and group interpersonal psychotherapy for the treatment of overweight individuals with binge eating disorder. Arch Gen Psychiatry 2002;59:713-21.

23. Grilo CM. A controlled study of cognitive behavioral therapy and fluoxetine for binge eating disorder (presentation) Charleston, SC: Eating Disorders Research Society annual meeting, 2002.

24. Telch CF, Agras WS, Linehan MM. Dialectical behavior therapy for binge eating disorder. J Consult Clin Psychol 2001;69:1061-5.

25. Carter WP, Hudson JI, Lalonde JK, et al. Pharmacologic treatment of binge eating disorder. Int J Eat Disord 2003;34(suppl):S74-88.

26. Appolinario JC, McElroy SL. Pharmacologic approaches in the treatment of binge eating disorder. Curr Drug Targets (in press).

27. Malhotra S, King KH, Welge JA, et al. Venlafaxine treatment of binge-eating disorder associated with obesity: a series of 35 patients. J Clin Psychiatry 2002;63:802-6.

28. Anderson JW, Greenway FL, Fujioka K, et al. Bupropion SR enhances weight loss: a 48-week double-blind, placebo-controlled trial. Obes Res 2002;10:633-41.

29. McElroy SL, Kotwal R, Malhotra S, et al. Are mood disorders and obesity related? A review for the mental health professional. J Clin Psychiatry (in press).

30. Appolinario JC, Bacaltchuk J, Sichieri R, et al. A randomized, double-blind, placebo-controlled study of sibutramine in the treatment of binge-eating disorder. Arch Gen Psychiatry 2003;60:1109-16.

31. Mitchell JE, Gosnell BA, Roerig JL, et al. Effects of sibutramine on binge eating, hunger, and fullness in a laboratory human feeding paradigm. Obes Res 2003;11:599-602.

32. Bray GA, Hollander P, Klein S, et al. A 6-month randomized, placebo-controlled, dose-ranging trial of topiramate for weight loss in obesity. Obes Res 2003;11:722-33.

33. Gadde KM, Franciscy DM, Wagner HR, 2nd, Krishnan KR. Zonisamide for weight loss in obese adults: a randomized controlled trial. JAMA 2003;289:1820-5.

34. McElroy SL, Arnold LM, Shapira NA, et al. Topiramate in the treatment of binge eating disorder associated with obesity: a randomized, placebo-controlled trial. Am J Psychiatry 2003;160:255-61.

35. McElroy SL, Kotwal R, Hudson JI, et al. Zonisamide in the treatment of binge-eating disorder: an open-label, prospective trial. J Clin Psychiatry 2004;65:50-6.

36. Pendleton VR, Goodrick GK, Poston WSC, et al. Exercise augments the effects of cognitive-behavioral therapy in the treatment of binge eating. Int J Eat Disord 2002;31(2):172-84.

Author and Disclosure Information

Paul E. Keck, Jr, MD

Renu Kotwal, MD
Assistant professor of clinical psychiatry

Rakesh Kaneria, MD
Fourth-year resident in psychiatry

Anna Guerdjikova, MS
PhD candidate, neuroscience program

Susan L. McElroy, MD
Professor of psychiatry and neuroscience

Division of Psychopharmacology Research Department of Psychiatry University of Cincinnati College of Medicine

Issue
Current Psychiatry - 03(04)
Publications
Topics
Page Number
31-47
Sections
Author and Disclosure Information

Paul E. Keck, Jr, MD

Renu Kotwal, MD
Assistant professor of clinical psychiatry

Rakesh Kaneria, MD
Fourth-year resident in psychiatry

Anna Guerdjikova, MS
PhD candidate, neuroscience program

Susan L. McElroy, MD
Professor of psychiatry and neuroscience

Division of Psychopharmacology Research Department of Psychiatry University of Cincinnati College of Medicine

Author and Disclosure Information

Paul E. Keck, Jr, MD

Renu Kotwal, MD
Assistant professor of clinical psychiatry

Rakesh Kaneria, MD
Fourth-year resident in psychiatry

Anna Guerdjikova, MS
PhD candidate, neuroscience program

Susan L. McElroy, MD
Professor of psychiatry and neuroscience

Division of Psychopharmacology Research Department of Psychiatry University of Cincinnati College of Medicine

Clinical snapshot of BED

Managing patients with binge-eating disorder (BED) often requires behavioral, medical, and psychiatric interventions.

These patients suffer from recurrent episodes of distressing, uncontrollable overeating, but they do not purge or show other compensatory weight-loss behaviors common to bulimia nervosa1 and anorexia nervosa.2-10 As a result, they are often overweight or obese and may have obesity-related illnesses, such as hypertension or type 2 diabetes. Mild to severe depression—unipolar or bipolar—is a common psychopathology.

Because no one treatment fits all patients with binge eating disorder, their management usually requires an individualized program of:

  • behavioral weight control
  • psychotherapy
  • and sometimes medications.

In our weight management clinic, we consider medication options based on patient preference and whether BED is uncomplicated (Figure 1) or coexists with a mood disorder (Figure 2).

This article presents the evidence on which we base our comprehensive approach. General psychiatrists with knowledge of BED can treat patients with this eating disorder, although complicated cases may require referral for specialized treatment.

Figure 1 Medication options for uncomplicated BED


Clinical characteristics

Psychiatric comorbidity. BED often occurs in patients with mood, anxiety, substance-abuse, impulsecontrol, and personality disorders.4,6,10-12 Mood disorder—particularly depression—appears to be the most common comorbidity. BED can occur with bipolar disorder12—a comorbidity that in our experience is underrecognized both clinically and in the literature.

Patients with BED and bipolar disorder show increased impulsivity and mood lability. As bipolar II disorder and other “soft-spectrum” forms are more common than bipolar I disorder, BED is also more likely to occur with hypomania than mania.

Overweight. Not surprisingly, BED is associated with overweight and obesity.5,8,9,11 Not all patients with BED are overweight or obese, but most who participate in clinical trials of BED treatments are at least overweight. BED has been reported in up to:

  • 30% of participants in weight-loss programs7
  • 70% of participants in groups such as Overeaters Anonymous
  • 50% of patients who seek bariatric surgery.5

In our experience, patients are often more distressed by their weight than by their binge eating, depression, or anxiety. Indeed, overweight and obesity are the usual reasons patients with BED present for treatment at our center.

Diagnosis. BED’s validity as a clinical diagnosis has been controversial since the disorder was first included in DSM-IV (Table 1).3 Debate continues about some definitions in the DSM criteria, including what amount of food is “definitely larger” than most people would eat and what is “loss of control over eating.”

Nevertheless, screening for BED is relatively easy. Clinicians may use the eating disorder section of the Structured Clinical Interview for DSM-IV or the Eating Disorders Examination. Alternatively, simply ask patients if they have episodes of uncontrollable overeating, during which they eat unusually large amounts of food and their eating feels out of control.

Course. BED begins in adolescence or adulthood. Disease course is variable, with periods of remission, recurrence, and chronicity.6,7,10 Interestingly, one prospective study showed that even if the binge eating resolves, persons may still develop obesity.13

Prevalence. BED affects 1.5% to 3% of the U.S. population. It is more common in women than men, equally prevalent in whites and blacks, and more prevalent than anorexia nervosa and bulimia nervosa combined.11,14 Subthreshold BED—such as obesity with infrequent or nondistressing binge eating—appears to be much more common,10 although no data are available.

Theories of binge eating

BED’s cause is unknown, but biological, familial, and psychosocial factors have been implicated.

Biological factors. The neurotransmitters serotonin (5-HT) and dopamine—as well as various peptides—have been shown to help regulate feeding behavior.10

Table 1

Diagnostic criteria for binge-eating disorder*

  1. Recurrent episodes of binge eating are characterized by both of the following:
    • eating in a discrete period of time (as within any 2 hours) an amount of food that is definitely larger than what most people would eat in a similar period under similar circumstances
    • a sense of lack of control over eating during the episode (a feeling that one cannot stop eating or control what or how much one is eating)
  2. The binge-eating episodes are associated with three or more of the following:
    • eating much more rapidly than normal
    • eating until feeling uncomfortably full
    • eating large amounts of food when not feeling physically hungry
    • eating alone because of being embarrassed by how much one is eating
    • feeling disgusted with oneself, depressed, or very guilty after overeating
  3. Marked distress regarding binge eating is present.
  4. The binge eating occurs, on average, at least 2 days a week for 6 months.
  5. The binge eating is not associated with the regular use of inappropriate compensatory behaviors (purging, fasting, excessive exercise) and does not occur exclusively during the course of anorexia nervosa or bulimia nervosa.
* Research criteria, DSM-IV-TR appendix B.
Source: Reprinted with permission from the Diagnostic and statistical manual of mental disorders, 4th edition, text revision. Copyright 2000. American Psychiatric Association.
 

 

Serotonin. Reduced 5-HT transporter binding has been shown in obese women with BED.15 Their 5-HT binding improved and binge eating subsided with group psychotherapy and fluoxetine, although the women continued to gain weight.

Figure 2 Medication options for BED with obesity and a mood disorder*



Dopamine. Obese patients who compulsively overeat may have lower levels of dopamine D2 receptors than do normal-weight controls.16

Genetic factors. In severely obese patients (body mass index 44±2), those with a DSM-IV diagnosis of BED exhibited mutations of the melanocortin 4 receptor gene, which affects the anorectic properties of alpha melanocyte-stimulating hormone.17

Familial factors associated with BED include parental depression and obesity.18

Psychosocial correlates include physical and sexual abuse, bullying by peers, and discrimination because of being overweight.19

Treatment recommendations

Few systematic studies have examined BED treatment. Emerging research suggests that behavioral weight-loss treatment, specialized psychotherapies, and medications may be effective in some patients with BED.4,6,8

Behavioral weight-loss treatment’s main goal is to manage the patient’s weight with a lower-calorie, healthy diet and to increase exercise.20,21

Over the short term (<1 year), behavioral weight-loss treatment produces similar weight loss in obese patients with or without BED; long-term results in both groups, however, have not been satisfactory.20,21 No studies have examined the efficacy of specialized diets (such as low-carbohydrate regimens) in patients with BED.

Specialized psychotherapy’s goal is to modify bingeeating behavior with behavioral self-management strategies, reducing interpersonal dysfunction and stress, and/or managing affective dysregulation.

Cognitive-behavioral therapy (CBT) and interpersonal therapy (IPT) have been effective in reducing binge eating, both acutely and for up to 12 months4,20-24 but less effective in achieving and maintaining weight loss. Patients who achieve remission in binge eating after undergoing CBT or IPI often experience modest but stable weight loss.20-22 For example, in a comparison study of CBT and IPT:

  • After 20 weekly sessions, patients whose binge eating was in remission lost weight (mean body mass index [BMI] −0.5 ± 1.5 kg/m2), whereas those who continued to binge gained weight (mean BMI +0.4 ±2.0 kg/m2).
  • At 12 months’ follow-up, patients still in remission continued to lose weight (mean BMI −1.0 ± 3.0 kg/m2), whereas those no longer in remission gained weight (mean BMI +0.7 ±2.9 kg/m2[P = 0.01]).22

Self-help and dialectical behavioral therapy (DBT) may also help reduce binge eating in BED. As with CBT and IPT, they are less effective in weight loss. In the only controlled study of DBT,24 patients achieved an average 2.5-lb weight loss after 20 weeks of DBT, compared with an average 0.6-lb weight gain in the control group. This difference was not significant, and the report did not include data on weight loss maintenance.

In summary, CBT may be more effective than behavioral weight loss treatment for reducing binge eating, but behavioral weight loss is more effective for weight loss.

Medications for BED

Medications that have been tried for BED include antidepressants, appetite suppressants, and anticonvulsants.25,26 Antidepressants are used to treat BED because:

  • BED is often associated with depressive symptoms and disorders.
  • BED is related to bulimia nervosa, and placebo-controlled trials have shown that the binge eating of bulimia nervosa responds to several classes of antidepressants. The selective serotonin reuptake inhibitor (SSRI) fluoxetine is the only medication indicated for treating any eating disorder (bulimia nervosa).
  • Bupropion and venlafaxine—a serotonin-norepinephrine reuptake inhibitor (SNRI)—have weight-loss properties.

SSRIs are the most extensively studied antidepressants for treating BED. SSRIs have weightloss properties, but only short term.25-26 Citalopram, fluoxetine, fluvoxamine, and sertraline have reduced binge eating and body weight more effectively than placebo during 6 to 9 weeks of treatment (Table 2).25-26 However, one controlled study23 showed that fluoxetine was not significantly more effective than placebo in reducing binge frequency or body weight after 16 weeks.

TCAs. Studies of tricyclic antidepressants (TCAs) for BED are sparse, and results have been mixed. In one trial, imipramine was similar to placebo in reducing binge frequency and body weight. In a placebo-controlled study of patients with nonpurging bulimia nervosa, desipramine reduced binge eating but had no effect on body weight.25,26

Table 2

Drug therapies shown to be effective for BED*

MedicationBinge eatingWeightDepressionStudy sizeDuration (weeks)Dosage (mg/d)
Antidepressants
Citalopram++38620 to 60
Fluoxetine †+++60620 to 80
Fluvoxamine++85950 to 300
Sertraline++34650 to 200
Appetite suppressant
Sibutramine+++601215
Anticonvulsant
Topiramate++611450 to 600
+ Improvement
− No improvement
* Randomized, controlled trials. Antidepressants were studied in patients with BED; sibutramine and topiramate were studied in patients with BED and associated obesity.
† One 16-week trial of fluoxetine for BED (reference 23) did not show statistically significant differences in post-treatment binge frequency or body-mass index.

Venlafaxine. In a retrospective review of 35 consecutive obese women with BED, venlafaxine, mean 222 mg/d for 28 to 300 days (median 120 days), reduced binge eating, body weight, and depressive symptoms.27

 

 

Bupropion has been more effective than placebo for treating:

  • uncomplicated obesity (short- and long-term)
  • obesity associated with depressive symptoms
  • bulimia nervosa (although bupropion is contraindicated in these patients because of seizure risk).26,28,29

No controlled trials have studied bupropion for BED. When using dosages effective in depressive disorders, we find bupropion helpful in reducing binge eating, body weight, and depressive symptoms in BED patients.

Appetite suppressants decrease appetite and weight, may increase satiety, and may reduce depressive symptoms.

Sibutramine—a serotonin, norepinephrine, and dopamine reuptake inhibitor indicated for managing obesity—has been reported effective in BED in a 12-week, randomized, double-blind, placebo-controlled trial. A 15-mg/d dosage reduced binge frequency, body weight, and depressive symptoms more effectively than placebo in 60 obese patients with BED.30 Most-frequent adverse effects (dry mouth and constipation) were mild and benign, and no significant complications were observed.

Sibutramine’s mechanism of action in BED is unknown. However, it suppressed food intake during binge-eating episodes in patients with BED in a randomized, controlled, cross-over laboratory study.31

Orlistat. We know of no published controlled studies of the lipase inhibitor orlistat in treating BED. In our experience, some patients do well with this agent, though we have observed infrequent purging episodes with it in patients with BED.

With orlistat, 120 mg tid, our BED patients have experienced weight loss comparable to that seen in uncomplicated obesity at similar dosages. Orlistat seems most effective for:

  • patients whose binge eating is in remission
  • those who responded to behavioral weightloss treatment, a psychological treatment, or another medication.

Anticonvulsants such as topiramate and zonisamide have been shown effective in treating obesity32,33 and are sometimes used to treat BED. Obese BED patients with mood disorders often do best with psychotherapy plus medication

Topiramate at dosages of 50 to 600 mg/d (median 212 mg/d) reduced binge-eating frequency, obsessive-compulsive features of binge eating, and body weight more effectively than placebo in a 14-week study of 61 obese patients with BED. These effects were maintained across 48 weeks in an open-label extension trial.34

Zonisamide, mean 513 mg/d, produced similar results during a prospective, open-label, 12-week trial in 15 patients with BED.35 A controlled trial to replicate these findings is ongoing.

BED may respond to anticonvulsant therapy for several reasons:

  • Some anticonvulsants are effective in treating bipolar disorder, which may occur with BED.12
  • Some anticonvulsants have shown benefit in conditions associated with pathologic impulsivity, such as substance abuse, impulse-control, and cluster B disorders.10

Growing evidence shows that bulimia nervosa and BED may be associated with pathologic impulsivity.

Combination therapies are generally more effective than monotherapies in patients with mood disorders, uncomplicated obesity, and possibly bulimia nervosa. Even so, few trials have systematically studied combination therapy in managing patients with BED.

Two studies compared psychotherapy and antidepressants alone and in combination in treating BED.21,23 Both showed that CBT alone was more effective in decreasing binge frequency than desipramine alone,21 fluoxetine alone,23 and the combination of CBT and medication. On the other hand, patients who took desipramine either alone or in combination experienced a greater degree of weight loss than those who did not take desipramine.21

In another combination therapy, exercise has been shown to be an effective adjunct to CBT in maintenance treatment of obese women with BED.36

No studies have compared behavioral weight management or a specialized psychotherapy in combination with an antiobesity drug or a weight-loss anticonvulsant in treating BED.

Treatment recommendations

In our experience, BED patients—particularly those with obesity and psychopathology—often do best with some combination of psychological treatment and medication:

  • The psychological component may be behavioral weight-loss treatment, a specialized psychotherapy such as CBT or IPT, or some combination of behavioral weight-loss treatment and specialized psychotherapy.
  • The medication component may consist of an antidepressant, anticonvulsant, antiobesity drug, or multiple drugs (such as an SSRI or sibutramine with topiramate for BED with major depression, or topiramate with lithium for BED with bipolar disorder).

Although combination therapies may be optimal for some patients, this approach remains unproven in controlled trials.

Patient preference. In addition to comorbidities, patient preference is an important consideration when choosing BED treatments. We determine our patients’ preferences by educating them as much as possible about their options. We explain the benefits and weaknesses of all treatments and encourage them to participate in forming their individualized treatment plans.

Patients sometimes have strong treatment preferences. Some prefer psychological treatments, whereas others prefer medications. Working with patient preferences enhances treatment adherence. For example, patients who fail a preferred treatment are often more willing to adhere to another treatment modality about which they initially were skeptical.

 

 

Related resources

  • Bray GA, Bouchard C (eds). Handbook of obesity: clinical applications (2nd ed). New York, NY: Marcel Dekker, 2004.
  • Cooper Z, Zairburn CG, Hawker DM. Cognitive behavioral treatment of obesity. A clinician’s guide. New York: Guilford Press, 2003.
  • Carter WP, Hudson JI, Lalonde JK, et al. Pharmacologic treatment of binge eating disorder. Int J Eat Disord 2003;34(suppl):S74-88.

Drug brand names

  • Bupropion • Wellbutrin
  • Citalopram • Celexa
  • Desipramine • Norpramin
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Imipramine • Tofranil
  • Lamotrigine • Lamictal
  • Lithium • Eskalith, others
  • Orlistat • Xenical
  • Sertraline • Zoloft
  • Sibutramine • Meridia
  • Topiramate • Topamax
  • Venlafaxine • Effexor
  • Zonisamide • Zonegran

Disclosure

Dr. Kotwal receives grant support from Elan Corporation and is a speaker for Ortho-McNeil Pharmaceutical and Pfizer Inc.

Dr. Kaneria and Ms. Guerdjikova report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. McElroy is a consultant to Abbott Laboratories, Bristol-Myers Squibb Co., Elan Corporation, GlaxoSmithKline, Janssen Pharmaceutica, Eli Lilly and Co., and Ortho-McNeil Pharmaceutical. She receives grant/research support from Elan Pharmaceuticals, Forest Pharmaceuticals, Merck & Co., Ortho-McNeil Pharmaceutical, and Sanofi-Synthelabo and is a speaker for Eli Lilly and Co. and Ortho-McNeil Pharmaceutical.

Clinical snapshot of BED

Managing patients with binge-eating disorder (BED) often requires behavioral, medical, and psychiatric interventions.

These patients suffer from recurrent episodes of distressing, uncontrollable overeating, but they do not purge or show other compensatory weight-loss behaviors common to bulimia nervosa1 and anorexia nervosa.2-10 As a result, they are often overweight or obese and may have obesity-related illnesses, such as hypertension or type 2 diabetes. Mild to severe depression—unipolar or bipolar—is a common psychopathology.

Because no one treatment fits all patients with binge eating disorder, their management usually requires an individualized program of:

  • behavioral weight control
  • psychotherapy
  • and sometimes medications.

In our weight management clinic, we consider medication options based on patient preference and whether BED is uncomplicated (Figure 1) or coexists with a mood disorder (Figure 2).

This article presents the evidence on which we base our comprehensive approach. General psychiatrists with knowledge of BED can treat patients with this eating disorder, although complicated cases may require referral for specialized treatment.

Figure 1 Medication options for uncomplicated BED


Clinical characteristics

Psychiatric comorbidity. BED often occurs in patients with mood, anxiety, substance-abuse, impulsecontrol, and personality disorders.4,6,10-12 Mood disorder—particularly depression—appears to be the most common comorbidity. BED can occur with bipolar disorder12—a comorbidity that in our experience is underrecognized both clinically and in the literature.

Patients with BED and bipolar disorder show increased impulsivity and mood lability. As bipolar II disorder and other “soft-spectrum” forms are more common than bipolar I disorder, BED is also more likely to occur with hypomania than mania.

Overweight. Not surprisingly, BED is associated with overweight and obesity.5,8,9,11 Not all patients with BED are overweight or obese, but most who participate in clinical trials of BED treatments are at least overweight. BED has been reported in up to:

  • 30% of participants in weight-loss programs7
  • 70% of participants in groups such as Overeaters Anonymous
  • 50% of patients who seek bariatric surgery.5

In our experience, patients are often more distressed by their weight than by their binge eating, depression, or anxiety. Indeed, overweight and obesity are the usual reasons patients with BED present for treatment at our center.

Diagnosis. BED’s validity as a clinical diagnosis has been controversial since the disorder was first included in DSM-IV (Table 1).3 Debate continues about some definitions in the DSM criteria, including what amount of food is “definitely larger” than most people would eat and what is “loss of control over eating.”

Nevertheless, screening for BED is relatively easy. Clinicians may use the eating disorder section of the Structured Clinical Interview for DSM-IV or the Eating Disorders Examination. Alternatively, simply ask patients if they have episodes of uncontrollable overeating, during which they eat unusually large amounts of food and their eating feels out of control.

Course. BED begins in adolescence or adulthood. Disease course is variable, with periods of remission, recurrence, and chronicity.6,7,10 Interestingly, one prospective study showed that even if the binge eating resolves, persons may still develop obesity.13

Prevalence. BED affects 1.5% to 3% of the U.S. population. It is more common in women than men, equally prevalent in whites and blacks, and more prevalent than anorexia nervosa and bulimia nervosa combined.11,14 Subthreshold BED—such as obesity with infrequent or nondistressing binge eating—appears to be much more common,10 although no data are available.

Theories of binge eating

BED’s cause is unknown, but biological, familial, and psychosocial factors have been implicated.

Biological factors. The neurotransmitters serotonin (5-HT) and dopamine—as well as various peptides—have been shown to help regulate feeding behavior.10

Table 1

Diagnostic criteria for binge-eating disorder*

  1. Recurrent episodes of binge eating are characterized by both of the following:
    • eating in a discrete period of time (as within any 2 hours) an amount of food that is definitely larger than what most people would eat in a similar period under similar circumstances
    • a sense of lack of control over eating during the episode (a feeling that one cannot stop eating or control what or how much one is eating)
  2. The binge-eating episodes are associated with three or more of the following:
    • eating much more rapidly than normal
    • eating until feeling uncomfortably full
    • eating large amounts of food when not feeling physically hungry
    • eating alone because of being embarrassed by how much one is eating
    • feeling disgusted with oneself, depressed, or very guilty after overeating
  3. Marked distress regarding binge eating is present.
  4. The binge eating occurs, on average, at least 2 days a week for 6 months.
  5. The binge eating is not associated with the regular use of inappropriate compensatory behaviors (purging, fasting, excessive exercise) and does not occur exclusively during the course of anorexia nervosa or bulimia nervosa.
* Research criteria, DSM-IV-TR appendix B.
Source: Reprinted with permission from the Diagnostic and statistical manual of mental disorders, 4th edition, text revision. Copyright 2000. American Psychiatric Association.
 

 

Serotonin. Reduced 5-HT transporter binding has been shown in obese women with BED.15 Their 5-HT binding improved and binge eating subsided with group psychotherapy and fluoxetine, although the women continued to gain weight.

Figure 2 Medication options for BED with obesity and a mood disorder*



Dopamine. Obese patients who compulsively overeat may have lower levels of dopamine D2 receptors than do normal-weight controls.16

Genetic factors. In severely obese patients (body mass index 44±2), those with a DSM-IV diagnosis of BED exhibited mutations of the melanocortin 4 receptor gene, which affects the anorectic properties of alpha melanocyte-stimulating hormone.17

Familial factors associated with BED include parental depression and obesity.18

Psychosocial correlates include physical and sexual abuse, bullying by peers, and discrimination because of being overweight.19

Treatment recommendations

Few systematic studies have examined BED treatment. Emerging research suggests that behavioral weight-loss treatment, specialized psychotherapies, and medications may be effective in some patients with BED.4,6,8

Behavioral weight-loss treatment’s main goal is to manage the patient’s weight with a lower-calorie, healthy diet and to increase exercise.20,21

Over the short term (<1 year), behavioral weight-loss treatment produces similar weight loss in obese patients with or without BED; long-term results in both groups, however, have not been satisfactory.20,21 No studies have examined the efficacy of specialized diets (such as low-carbohydrate regimens) in patients with BED.

Specialized psychotherapy’s goal is to modify bingeeating behavior with behavioral self-management strategies, reducing interpersonal dysfunction and stress, and/or managing affective dysregulation.

Cognitive-behavioral therapy (CBT) and interpersonal therapy (IPT) have been effective in reducing binge eating, both acutely and for up to 12 months4,20-24 but less effective in achieving and maintaining weight loss. Patients who achieve remission in binge eating after undergoing CBT or IPI often experience modest but stable weight loss.20-22 For example, in a comparison study of CBT and IPT:

  • After 20 weekly sessions, patients whose binge eating was in remission lost weight (mean body mass index [BMI] −0.5 ± 1.5 kg/m2), whereas those who continued to binge gained weight (mean BMI +0.4 ±2.0 kg/m2).
  • At 12 months’ follow-up, patients still in remission continued to lose weight (mean BMI −1.0 ± 3.0 kg/m2), whereas those no longer in remission gained weight (mean BMI +0.7 ±2.9 kg/m2[P = 0.01]).22

Self-help and dialectical behavioral therapy (DBT) may also help reduce binge eating in BED. As with CBT and IPT, they are less effective in weight loss. In the only controlled study of DBT,24 patients achieved an average 2.5-lb weight loss after 20 weeks of DBT, compared with an average 0.6-lb weight gain in the control group. This difference was not significant, and the report did not include data on weight loss maintenance.

In summary, CBT may be more effective than behavioral weight loss treatment for reducing binge eating, but behavioral weight loss is more effective for weight loss.

Medications for BED

Medications that have been tried for BED include antidepressants, appetite suppressants, and anticonvulsants.25,26 Antidepressants are used to treat BED because:

  • BED is often associated with depressive symptoms and disorders.
  • BED is related to bulimia nervosa, and placebo-controlled trials have shown that the binge eating of bulimia nervosa responds to several classes of antidepressants. The selective serotonin reuptake inhibitor (SSRI) fluoxetine is the only medication indicated for treating any eating disorder (bulimia nervosa).
  • Bupropion and venlafaxine—a serotonin-norepinephrine reuptake inhibitor (SNRI)—have weight-loss properties.

SSRIs are the most extensively studied antidepressants for treating BED. SSRIs have weightloss properties, but only short term.25-26 Citalopram, fluoxetine, fluvoxamine, and sertraline have reduced binge eating and body weight more effectively than placebo during 6 to 9 weeks of treatment (Table 2).25-26 However, one controlled study23 showed that fluoxetine was not significantly more effective than placebo in reducing binge frequency or body weight after 16 weeks.

TCAs. Studies of tricyclic antidepressants (TCAs) for BED are sparse, and results have been mixed. In one trial, imipramine was similar to placebo in reducing binge frequency and body weight. In a placebo-controlled study of patients with nonpurging bulimia nervosa, desipramine reduced binge eating but had no effect on body weight.25,26

Table 2

Drug therapies shown to be effective for BED*

MedicationBinge eatingWeightDepressionStudy sizeDuration (weeks)Dosage (mg/d)
Antidepressants
Citalopram++38620 to 60
Fluoxetine †+++60620 to 80
Fluvoxamine++85950 to 300
Sertraline++34650 to 200
Appetite suppressant
Sibutramine+++601215
Anticonvulsant
Topiramate++611450 to 600
+ Improvement
− No improvement
* Randomized, controlled trials. Antidepressants were studied in patients with BED; sibutramine and topiramate were studied in patients with BED and associated obesity.
† One 16-week trial of fluoxetine for BED (reference 23) did not show statistically significant differences in post-treatment binge frequency or body-mass index.

Venlafaxine. In a retrospective review of 35 consecutive obese women with BED, venlafaxine, mean 222 mg/d for 28 to 300 days (median 120 days), reduced binge eating, body weight, and depressive symptoms.27

 

 

Bupropion has been more effective than placebo for treating:

  • uncomplicated obesity (short- and long-term)
  • obesity associated with depressive symptoms
  • bulimia nervosa (although bupropion is contraindicated in these patients because of seizure risk).26,28,29

No controlled trials have studied bupropion for BED. When using dosages effective in depressive disorders, we find bupropion helpful in reducing binge eating, body weight, and depressive symptoms in BED patients.

Appetite suppressants decrease appetite and weight, may increase satiety, and may reduce depressive symptoms.

Sibutramine—a serotonin, norepinephrine, and dopamine reuptake inhibitor indicated for managing obesity—has been reported effective in BED in a 12-week, randomized, double-blind, placebo-controlled trial. A 15-mg/d dosage reduced binge frequency, body weight, and depressive symptoms more effectively than placebo in 60 obese patients with BED.30 Most-frequent adverse effects (dry mouth and constipation) were mild and benign, and no significant complications were observed.

Sibutramine’s mechanism of action in BED is unknown. However, it suppressed food intake during binge-eating episodes in patients with BED in a randomized, controlled, cross-over laboratory study.31

Orlistat. We know of no published controlled studies of the lipase inhibitor orlistat in treating BED. In our experience, some patients do well with this agent, though we have observed infrequent purging episodes with it in patients with BED.

With orlistat, 120 mg tid, our BED patients have experienced weight loss comparable to that seen in uncomplicated obesity at similar dosages. Orlistat seems most effective for:

  • patients whose binge eating is in remission
  • those who responded to behavioral weightloss treatment, a psychological treatment, or another medication.

Anticonvulsants such as topiramate and zonisamide have been shown effective in treating obesity32,33 and are sometimes used to treat BED. Obese BED patients with mood disorders often do best with psychotherapy plus medication

Topiramate at dosages of 50 to 600 mg/d (median 212 mg/d) reduced binge-eating frequency, obsessive-compulsive features of binge eating, and body weight more effectively than placebo in a 14-week study of 61 obese patients with BED. These effects were maintained across 48 weeks in an open-label extension trial.34

Zonisamide, mean 513 mg/d, produced similar results during a prospective, open-label, 12-week trial in 15 patients with BED.35 A controlled trial to replicate these findings is ongoing.

BED may respond to anticonvulsant therapy for several reasons:

  • Some anticonvulsants are effective in treating bipolar disorder, which may occur with BED.12
  • Some anticonvulsants have shown benefit in conditions associated with pathologic impulsivity, such as substance abuse, impulse-control, and cluster B disorders.10

Growing evidence shows that bulimia nervosa and BED may be associated with pathologic impulsivity.

Combination therapies are generally more effective than monotherapies in patients with mood disorders, uncomplicated obesity, and possibly bulimia nervosa. Even so, few trials have systematically studied combination therapy in managing patients with BED.

Two studies compared psychotherapy and antidepressants alone and in combination in treating BED.21,23 Both showed that CBT alone was more effective in decreasing binge frequency than desipramine alone,21 fluoxetine alone,23 and the combination of CBT and medication. On the other hand, patients who took desipramine either alone or in combination experienced a greater degree of weight loss than those who did not take desipramine.21

In another combination therapy, exercise has been shown to be an effective adjunct to CBT in maintenance treatment of obese women with BED.36

No studies have compared behavioral weight management or a specialized psychotherapy in combination with an antiobesity drug or a weight-loss anticonvulsant in treating BED.

Treatment recommendations

In our experience, BED patients—particularly those with obesity and psychopathology—often do best with some combination of psychological treatment and medication:

  • The psychological component may be behavioral weight-loss treatment, a specialized psychotherapy such as CBT or IPT, or some combination of behavioral weight-loss treatment and specialized psychotherapy.
  • The medication component may consist of an antidepressant, anticonvulsant, antiobesity drug, or multiple drugs (such as an SSRI or sibutramine with topiramate for BED with major depression, or topiramate with lithium for BED with bipolar disorder).

Although combination therapies may be optimal for some patients, this approach remains unproven in controlled trials.

Patient preference. In addition to comorbidities, patient preference is an important consideration when choosing BED treatments. We determine our patients’ preferences by educating them as much as possible about their options. We explain the benefits and weaknesses of all treatments and encourage them to participate in forming their individualized treatment plans.

Patients sometimes have strong treatment preferences. Some prefer psychological treatments, whereas others prefer medications. Working with patient preferences enhances treatment adherence. For example, patients who fail a preferred treatment are often more willing to adhere to another treatment modality about which they initially were skeptical.

 

 

Related resources

  • Bray GA, Bouchard C (eds). Handbook of obesity: clinical applications (2nd ed). New York, NY: Marcel Dekker, 2004.
  • Cooper Z, Zairburn CG, Hawker DM. Cognitive behavioral treatment of obesity. A clinician’s guide. New York: Guilford Press, 2003.
  • Carter WP, Hudson JI, Lalonde JK, et al. Pharmacologic treatment of binge eating disorder. Int J Eat Disord 2003;34(suppl):S74-88.

Drug brand names

  • Bupropion • Wellbutrin
  • Citalopram • Celexa
  • Desipramine • Norpramin
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Imipramine • Tofranil
  • Lamotrigine • Lamictal
  • Lithium • Eskalith, others
  • Orlistat • Xenical
  • Sertraline • Zoloft
  • Sibutramine • Meridia
  • Topiramate • Topamax
  • Venlafaxine • Effexor
  • Zonisamide • Zonegran

Disclosure

Dr. Kotwal receives grant support from Elan Corporation and is a speaker for Ortho-McNeil Pharmaceutical and Pfizer Inc.

Dr. Kaneria and Ms. Guerdjikova report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. McElroy is a consultant to Abbott Laboratories, Bristol-Myers Squibb Co., Elan Corporation, GlaxoSmithKline, Janssen Pharmaceutica, Eli Lilly and Co., and Ortho-McNeil Pharmaceutical. She receives grant/research support from Elan Pharmaceuticals, Forest Pharmaceuticals, Merck & Co., Ortho-McNeil Pharmaceutical, and Sanofi-Synthelabo and is a speaker for Eli Lilly and Co. and Ortho-McNeil Pharmaceutical.

References

1. Pope HG, Hudson JI. Bulimia nervosa: Persistent disorders requires equally persistent treatment. Current Psychiatry 2004;3(1):13-22.

2. Halmi KA. Anorexia nervosa: Dual therapy can bring patients back from the brink. Current Psychiatry 2004;3(3):39-56.

3. American Psychiatric Association. Diagnostic and statistical manual of mental disorders (4th ed). Washington, DC: American Psychiatric Association, 1994.

4. Agras WS. Treatment of binge eating disorder. In: Gabbard GO (ed). Treatments of psychiatric disorders (3rd ed). Washington, DC: American Psychiatric Press, 2001;2209-19.

5. de Zwaan M. Binge eating disorder and obesity. Int J Obes Relat Metab Disord 2001;25(suppl 1):S51-5.

6. Dingemans AE, Bruna MJ, van Furth EF. Binge eating disorder: a review. Int J Obes Relat Metab Disord 2002;26:299-307.

7. Fairburn CG, Harrison PJ. Eating disorders. Lancet 2003;361:407-16.

8. Walsh BT (ed). The current status of binge eating disorder. Int J Eat Disord 2003;34(suppl):S1-120.

9. Devlin MJ, Goldfein JA, Dobrow I. What is this thing called BED? Current status of binge eating disorder nosology. Int J Eat Disord 2003;34(suppl):S2-18.

10. McElroy SL, Kotwal R. Binge eating. In: Hollander E, Stein D (eds). Handbook of impulse control disorders Washington, DC: American Psychiatric Press (in press).

11. Smith DE, Marcus MD, Lewis CE, et al. Prevalence of binge eating disorder, obesity, and depression in a biracial cohort of young adults. Ann Behav Med 1998;20:227-32.

12. Kruger S, Shugar G, Cooke RG. Comorbidity of binge eating disorder and the partial binge eating syndrome with bipolar disorder. Int J Eat Disord 1996;19:45-52.

13. Fairburn CG, Cooper Z, Doll H, et al. The natural course of bulimia nervosa and binge eating disorder in young women. Arch Gen Psychiatry 2000;57:659-65.

14. Striegel-Moore RH, Franko DL. Epidemiology of binge eating disorder. Int J Eat Disord 2003;34(suppl):S19-29.

15. Tammela LI, Rissanen A, Kuikka JT, et al. Treatment improves serotonin transporter binding and reduces binge eating. Psychopharmacology (Berl) 2003;170:89-93.

16. Wang GJ, Volkow ND, Logan J, et al. Brain dopamine and obesity. Lancet 2001;357:354-7.

17. Branson R, Potoczna N, Kral JG, et al. Binge eating as a major phenotype of melanocortin 4 receptor gene mutations. N Engl J Med 2003;348:1096-103.

18. Fairburn CG, Doll HA, Welch SL, et al. Risk factors for binge eating disorder: a community-based, case-control study. Arch Gen Psychiatry 1998;55:425-32.

19. Striegel-Moore RH, Dohm FA, Pike KM, et al. Abuse, bullying, and discrimination as risk factors for binge eating disorder. Am J Psychiatry 2002;159:1902-7.

20. Wonderlich SA, de Zwaan M, Mitchell JE, et al. Psychological and dietary treatments of binge eating disorder: conceptual implications. Int J Eat Disord 2003;34(suppl):S58-78.

21. Agras WS, Telch DF, Arnow B, et al. Weight loss, cognitive-behavioral, and desipramine treatments in binge eating disorder. An additive design. Behav Ther 1994;25:225-38.

22. Wilfley DE, Welch RR, Stein RI, et al. A randomized comparison of group cognitive-behavioral therapy and group interpersonal psychotherapy for the treatment of overweight individuals with binge eating disorder. Arch Gen Psychiatry 2002;59:713-21.

23. Grilo CM. A controlled study of cognitive behavioral therapy and fluoxetine for binge eating disorder (presentation) Charleston, SC: Eating Disorders Research Society annual meeting, 2002.

24. Telch CF, Agras WS, Linehan MM. Dialectical behavior therapy for binge eating disorder. J Consult Clin Psychol 2001;69:1061-5.

25. Carter WP, Hudson JI, Lalonde JK, et al. Pharmacologic treatment of binge eating disorder. Int J Eat Disord 2003;34(suppl):S74-88.

26. Appolinario JC, McElroy SL. Pharmacologic approaches in the treatment of binge eating disorder. Curr Drug Targets (in press).

27. Malhotra S, King KH, Welge JA, et al. Venlafaxine treatment of binge-eating disorder associated with obesity: a series of 35 patients. J Clin Psychiatry 2002;63:802-6.

28. Anderson JW, Greenway FL, Fujioka K, et al. Bupropion SR enhances weight loss: a 48-week double-blind, placebo-controlled trial. Obes Res 2002;10:633-41.

29. McElroy SL, Kotwal R, Malhotra S, et al. Are mood disorders and obesity related? A review for the mental health professional. J Clin Psychiatry (in press).

30. Appolinario JC, Bacaltchuk J, Sichieri R, et al. A randomized, double-blind, placebo-controlled study of sibutramine in the treatment of binge-eating disorder. Arch Gen Psychiatry 2003;60:1109-16.

31. Mitchell JE, Gosnell BA, Roerig JL, et al. Effects of sibutramine on binge eating, hunger, and fullness in a laboratory human feeding paradigm. Obes Res 2003;11:599-602.

32. Bray GA, Hollander P, Klein S, et al. A 6-month randomized, placebo-controlled, dose-ranging trial of topiramate for weight loss in obesity. Obes Res 2003;11:722-33.

33. Gadde KM, Franciscy DM, Wagner HR, 2nd, Krishnan KR. Zonisamide for weight loss in obese adults: a randomized controlled trial. JAMA 2003;289:1820-5.

34. McElroy SL, Arnold LM, Shapira NA, et al. Topiramate in the treatment of binge eating disorder associated with obesity: a randomized, placebo-controlled trial. Am J Psychiatry 2003;160:255-61.

35. McElroy SL, Kotwal R, Hudson JI, et al. Zonisamide in the treatment of binge-eating disorder: an open-label, prospective trial. J Clin Psychiatry 2004;65:50-6.

36. Pendleton VR, Goodrick GK, Poston WSC, et al. Exercise augments the effects of cognitive-behavioral therapy in the treatment of binge eating. Int J Eat Disord 2002;31(2):172-84.

References

1. Pope HG, Hudson JI. Bulimia nervosa: Persistent disorders requires equally persistent treatment. Current Psychiatry 2004;3(1):13-22.

2. Halmi KA. Anorexia nervosa: Dual therapy can bring patients back from the brink. Current Psychiatry 2004;3(3):39-56.

3. American Psychiatric Association. Diagnostic and statistical manual of mental disorders (4th ed). Washington, DC: American Psychiatric Association, 1994.

4. Agras WS. Treatment of binge eating disorder. In: Gabbard GO (ed). Treatments of psychiatric disorders (3rd ed). Washington, DC: American Psychiatric Press, 2001;2209-19.

5. de Zwaan M. Binge eating disorder and obesity. Int J Obes Relat Metab Disord 2001;25(suppl 1):S51-5.

6. Dingemans AE, Bruna MJ, van Furth EF. Binge eating disorder: a review. Int J Obes Relat Metab Disord 2002;26:299-307.

7. Fairburn CG, Harrison PJ. Eating disorders. Lancet 2003;361:407-16.

8. Walsh BT (ed). The current status of binge eating disorder. Int J Eat Disord 2003;34(suppl):S1-120.

9. Devlin MJ, Goldfein JA, Dobrow I. What is this thing called BED? Current status of binge eating disorder nosology. Int J Eat Disord 2003;34(suppl):S2-18.

10. McElroy SL, Kotwal R. Binge eating. In: Hollander E, Stein D (eds). Handbook of impulse control disorders Washington, DC: American Psychiatric Press (in press).

11. Smith DE, Marcus MD, Lewis CE, et al. Prevalence of binge eating disorder, obesity, and depression in a biracial cohort of young adults. Ann Behav Med 1998;20:227-32.

12. Kruger S, Shugar G, Cooke RG. Comorbidity of binge eating disorder and the partial binge eating syndrome with bipolar disorder. Int J Eat Disord 1996;19:45-52.

13. Fairburn CG, Cooper Z, Doll H, et al. The natural course of bulimia nervosa and binge eating disorder in young women. Arch Gen Psychiatry 2000;57:659-65.

14. Striegel-Moore RH, Franko DL. Epidemiology of binge eating disorder. Int J Eat Disord 2003;34(suppl):S19-29.

15. Tammela LI, Rissanen A, Kuikka JT, et al. Treatment improves serotonin transporter binding and reduces binge eating. Psychopharmacology (Berl) 2003;170:89-93.

16. Wang GJ, Volkow ND, Logan J, et al. Brain dopamine and obesity. Lancet 2001;357:354-7.

17. Branson R, Potoczna N, Kral JG, et al. Binge eating as a major phenotype of melanocortin 4 receptor gene mutations. N Engl J Med 2003;348:1096-103.

18. Fairburn CG, Doll HA, Welch SL, et al. Risk factors for binge eating disorder: a community-based, case-control study. Arch Gen Psychiatry 1998;55:425-32.

19. Striegel-Moore RH, Dohm FA, Pike KM, et al. Abuse, bullying, and discrimination as risk factors for binge eating disorder. Am J Psychiatry 2002;159:1902-7.

20. Wonderlich SA, de Zwaan M, Mitchell JE, et al. Psychological and dietary treatments of binge eating disorder: conceptual implications. Int J Eat Disord 2003;34(suppl):S58-78.

21. Agras WS, Telch DF, Arnow B, et al. Weight loss, cognitive-behavioral, and desipramine treatments in binge eating disorder. An additive design. Behav Ther 1994;25:225-38.

22. Wilfley DE, Welch RR, Stein RI, et al. A randomized comparison of group cognitive-behavioral therapy and group interpersonal psychotherapy for the treatment of overweight individuals with binge eating disorder. Arch Gen Psychiatry 2002;59:713-21.

23. Grilo CM. A controlled study of cognitive behavioral therapy and fluoxetine for binge eating disorder (presentation) Charleston, SC: Eating Disorders Research Society annual meeting, 2002.

24. Telch CF, Agras WS, Linehan MM. Dialectical behavior therapy for binge eating disorder. J Consult Clin Psychol 2001;69:1061-5.

25. Carter WP, Hudson JI, Lalonde JK, et al. Pharmacologic treatment of binge eating disorder. Int J Eat Disord 2003;34(suppl):S74-88.

26. Appolinario JC, McElroy SL. Pharmacologic approaches in the treatment of binge eating disorder. Curr Drug Targets (in press).

27. Malhotra S, King KH, Welge JA, et al. Venlafaxine treatment of binge-eating disorder associated with obesity: a series of 35 patients. J Clin Psychiatry 2002;63:802-6.

28. Anderson JW, Greenway FL, Fujioka K, et al. Bupropion SR enhances weight loss: a 48-week double-blind, placebo-controlled trial. Obes Res 2002;10:633-41.

29. McElroy SL, Kotwal R, Malhotra S, et al. Are mood disorders and obesity related? A review for the mental health professional. J Clin Psychiatry (in press).

30. Appolinario JC, Bacaltchuk J, Sichieri R, et al. A randomized, double-blind, placebo-controlled study of sibutramine in the treatment of binge-eating disorder. Arch Gen Psychiatry 2003;60:1109-16.

31. Mitchell JE, Gosnell BA, Roerig JL, et al. Effects of sibutramine on binge eating, hunger, and fullness in a laboratory human feeding paradigm. Obes Res 2003;11:599-602.

32. Bray GA, Hollander P, Klein S, et al. A 6-month randomized, placebo-controlled, dose-ranging trial of topiramate for weight loss in obesity. Obes Res 2003;11:722-33.

33. Gadde KM, Franciscy DM, Wagner HR, 2nd, Krishnan KR. Zonisamide for weight loss in obese adults: a randomized controlled trial. JAMA 2003;289:1820-5.

34. McElroy SL, Arnold LM, Shapira NA, et al. Topiramate in the treatment of binge eating disorder associated with obesity: a randomized, placebo-controlled trial. Am J Psychiatry 2003;160:255-61.

35. McElroy SL, Kotwal R, Hudson JI, et al. Zonisamide in the treatment of binge-eating disorder: an open-label, prospective trial. J Clin Psychiatry 2004;65:50-6.

36. Pendleton VR, Goodrick GK, Poston WSC, et al. Exercise augments the effects of cognitive-behavioral therapy in the treatment of binge eating. Int J Eat Disord 2002;31(2):172-84.

Issue
Current Psychiatry - 03(04)
Issue
Current Psychiatry - 03(04)
Page Number
31-47
Page Number
31-47
Publications
Publications
Topics
Article Type
Display Headline
Update on eating disorders: Binge-eating disorder
Display Headline
Update on eating disorders: Binge-eating disorder
Sections
Article Source

PURLs Copyright

Inside the Article

Bipolar I vs. Bipolar II

Article Type
Changed
Mon, 04/16/2018 - 14:16
Display Headline
Bipolar I vs. Bipolar II

I agree with most of the points in Dr. Shivakumar and Dr. Suppes’ article on the Texas Medical Algorithm Project (TMAP) and with the algorithms they mentioned (Current Psychiatry, February 2004).

However, the article does not address the difference between bipolar type I and bipolar type II disorder. While this may at first seem trivial, recognizing the difference is crucial to planning treatment. Since rapid cycling and depression are more prevalent than hypomania in bipolar type II, patients with this form of the disorder often require different medication(s) than do those with bipolar type I.

Also, some psychotropics are appropriate for outpatient treatment but not for inpatients and vice-versa. For example, lamotrigine takes time to work up to an effective dosage without significantly increasing the risk for rash; this would be reasonable treatment for an outpatient with bipolar type II but is not practical for an inpatient, especially with bipolar type I.

Michael S. Wilson, II, MD
Louisiana State University Health Sciences Center
New Orleans

Dr. Suppes responds

Dr. Wilson raises the issue of treatment recommendations for bipolar I versus bipolar II disorder.

All treatment guidelines—including the American Psychiatric Association Guidelines, Texas Algorithms, and others—are based on evidence gathered from studies of bipolar I patients. The full article from the TMAP consensus conference discusses this issue as well as the paucity of data available to make treatment recommendations for patients with bipolar II disorder.1

Unfortunately, this has not changed dramatically over the last 4 years. The good news is that numerous ongoing studies will reveal how best to treat bipolar II patients.

Dr. Wilson also notes that time to response makes a medication appropriate for use in one setting but not in another. Given today’s brief inpatient stays, any antidepressant or maintenance medication started during hospitalization will not begin to work until after discharge. Following titration guidelines with lamotrigine is critical, but as with antidepressants the time to response is a few weeks. Thus, these medications will require outpatient monitoring to assess efficacy and tolerability.

Delineating treatment for patients with bipolar II disorder is important. No matter how the prevalence is evaluated, bipolar II disorder affects many individuals. We recently reviewed the evidence in this area2 and were struck by how little attention this patient group has received to date.

Trisha Suppes, MD, PhD
Associate professor, department of psychiatry
Director, Bipolar Disorder Research Program
University of Texas Southwestern Medical Center
Dallas

References

  1. Suppes T, Dennehy EB, Swann AC, et al. Report of the Texas consensus conference panel on medication treatment of bipolar disorder 2000. J Clin Psychiatry 2002;63:288–99.
  2. Suppes T, Dennehy EB. Evidence-based long term treatment of bipolar II disorder. J Clin Psychiatry 2002;63(suppl 10):29–33.
Article PDF
Author and Disclosure Information

Issue
Current Psychiatry - 03(04)
Publications
Topics
Page Number
08-13
Sections
Author and Disclosure Information

Author and Disclosure Information

Article PDF
Article PDF

I agree with most of the points in Dr. Shivakumar and Dr. Suppes’ article on the Texas Medical Algorithm Project (TMAP) and with the algorithms they mentioned (Current Psychiatry, February 2004).

However, the article does not address the difference between bipolar type I and bipolar type II disorder. While this may at first seem trivial, recognizing the difference is crucial to planning treatment. Since rapid cycling and depression are more prevalent than hypomania in bipolar type II, patients with this form of the disorder often require different medication(s) than do those with bipolar type I.

Also, some psychotropics are appropriate for outpatient treatment but not for inpatients and vice-versa. For example, lamotrigine takes time to work up to an effective dosage without significantly increasing the risk for rash; this would be reasonable treatment for an outpatient with bipolar type II but is not practical for an inpatient, especially with bipolar type I.

Michael S. Wilson, II, MD
Louisiana State University Health Sciences Center
New Orleans

Dr. Suppes responds

Dr. Wilson raises the issue of treatment recommendations for bipolar I versus bipolar II disorder.

All treatment guidelines—including the American Psychiatric Association Guidelines, Texas Algorithms, and others—are based on evidence gathered from studies of bipolar I patients. The full article from the TMAP consensus conference discusses this issue as well as the paucity of data available to make treatment recommendations for patients with bipolar II disorder.1

Unfortunately, this has not changed dramatically over the last 4 years. The good news is that numerous ongoing studies will reveal how best to treat bipolar II patients.

Dr. Wilson also notes that time to response makes a medication appropriate for use in one setting but not in another. Given today’s brief inpatient stays, any antidepressant or maintenance medication started during hospitalization will not begin to work until after discharge. Following titration guidelines with lamotrigine is critical, but as with antidepressants the time to response is a few weeks. Thus, these medications will require outpatient monitoring to assess efficacy and tolerability.

Delineating treatment for patients with bipolar II disorder is important. No matter how the prevalence is evaluated, bipolar II disorder affects many individuals. We recently reviewed the evidence in this area2 and were struck by how little attention this patient group has received to date.

Trisha Suppes, MD, PhD
Associate professor, department of psychiatry
Director, Bipolar Disorder Research Program
University of Texas Southwestern Medical Center
Dallas

References

  1. Suppes T, Dennehy EB, Swann AC, et al. Report of the Texas consensus conference panel on medication treatment of bipolar disorder 2000. J Clin Psychiatry 2002;63:288–99.
  2. Suppes T, Dennehy EB. Evidence-based long term treatment of bipolar II disorder. J Clin Psychiatry 2002;63(suppl 10):29–33.

I agree with most of the points in Dr. Shivakumar and Dr. Suppes’ article on the Texas Medical Algorithm Project (TMAP) and with the algorithms they mentioned (Current Psychiatry, February 2004).

However, the article does not address the difference between bipolar type I and bipolar type II disorder. While this may at first seem trivial, recognizing the difference is crucial to planning treatment. Since rapid cycling and depression are more prevalent than hypomania in bipolar type II, patients with this form of the disorder often require different medication(s) than do those with bipolar type I.

Also, some psychotropics are appropriate for outpatient treatment but not for inpatients and vice-versa. For example, lamotrigine takes time to work up to an effective dosage without significantly increasing the risk for rash; this would be reasonable treatment for an outpatient with bipolar type II but is not practical for an inpatient, especially with bipolar type I.

Michael S. Wilson, II, MD
Louisiana State University Health Sciences Center
New Orleans

Dr. Suppes responds

Dr. Wilson raises the issue of treatment recommendations for bipolar I versus bipolar II disorder.

All treatment guidelines—including the American Psychiatric Association Guidelines, Texas Algorithms, and others—are based on evidence gathered from studies of bipolar I patients. The full article from the TMAP consensus conference discusses this issue as well as the paucity of data available to make treatment recommendations for patients with bipolar II disorder.1

Unfortunately, this has not changed dramatically over the last 4 years. The good news is that numerous ongoing studies will reveal how best to treat bipolar II patients.

Dr. Wilson also notes that time to response makes a medication appropriate for use in one setting but not in another. Given today’s brief inpatient stays, any antidepressant or maintenance medication started during hospitalization will not begin to work until after discharge. Following titration guidelines with lamotrigine is critical, but as with antidepressants the time to response is a few weeks. Thus, these medications will require outpatient monitoring to assess efficacy and tolerability.

Delineating treatment for patients with bipolar II disorder is important. No matter how the prevalence is evaluated, bipolar II disorder affects many individuals. We recently reviewed the evidence in this area2 and were struck by how little attention this patient group has received to date.

Trisha Suppes, MD, PhD
Associate professor, department of psychiatry
Director, Bipolar Disorder Research Program
University of Texas Southwestern Medical Center
Dallas

References

  1. Suppes T, Dennehy EB, Swann AC, et al. Report of the Texas consensus conference panel on medication treatment of bipolar disorder 2000. J Clin Psychiatry 2002;63:288–99.
  2. Suppes T, Dennehy EB. Evidence-based long term treatment of bipolar II disorder. J Clin Psychiatry 2002;63(suppl 10):29–33.
Issue
Current Psychiatry - 03(04)
Issue
Current Psychiatry - 03(04)
Page Number
08-13
Page Number
08-13
Publications
Publications
Topics
Article Type
Display Headline
Bipolar I vs. Bipolar II
Display Headline
Bipolar I vs. Bipolar II
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media