Allowed Publications
Slot System
Featured Buckets
Featured Buckets Admin

Clinical guide to countertransference: Help medical colleagues deal with ‘difficult’ patients

Article Type
Changed
Tue, 12/11/2018 - 15:06
Display Headline
Clinical guide to countertransference: Help medical colleagues deal with ‘difficult’ patients

WEB AUDIO
Listen to Dr. Muskin discuss the patient-physician dynamic

Two strangers meet in the hospital cafeteria. Mrs. R, an elderly woman, asks Dr. W, a first-year medical resident, for help in getting a bottle of soda from the cooler. Afterward, Dr. W comments to a colleague with whom she is having lunch, “That woman reminds me of my grandmother.”

What does that comment reflect about Dr. W? It is a statement about the doctor’s transference. That is, she is aware of elements about Mrs. R that evoke internal responses appropriate to a prior important relationship.

What if Mrs. R was to subsequently faint, require admission to the hospital, and become Dr. W’s patient? If Dr. W’s comment indicates transference, would the same reaction to Mrs. R now be countertransference? Does that change if the doctor is unaware of emotions Mrs. R evokes? Is it still countertransference whether Dr. W is caring and compassionate, overly involved with Mrs. R, or—unaware of negative feelings associated with “grandmothers”—avoids the patient?

This article explores how complex internal experiences play out in the general medical setting and discusses how psychiatric consultants can help medical/surgical colleagues understand and manage difficult patient-physician relationships.

The therapeutic dyad

Countertransference and transference are concepts embedded in psychodynamic thinking. They are part of how many people think about interpersonal relations, whether or not they use these terms. Countertransference and transference may be conscious, but they always have an unconscious component. Factors that influence what will be transference and countertransference in adult life have both:

  • a biological component because part of personality is genetic
  • a psychological component based upon experiences throughout life ( Box 1 ).1
Box 1

Roots of transference: From a child’s experience of the world

Genetic factors play a role in personality formation. A child’s personality, which emerges early in life, shapes interactions with people who are significant during childhood. Predispositions shape those experiences and influence what people internalize from those relationships.

In adults, many aspects of what we understand as transference—the experience someone has of a figure from the past—originate from the limitations with which children perceive and interpret their experiences. Transference is not truth about a significant past relationship; it is truth as the person experienced other people and now remembers or reacts to individuals who are reminiscent of those from the past.1

Not all psychotherapeutic treatments—and thus not all therapists—use the concept of transference as a therapeutic component. Some therapists who employ transference in treatment will discuss how the patient interacts with the therapist only when the phenomenon interferes with therapy. Interpretation of transference is a therapeutic modality of psychoanalysis and psychodynamic psychotherapy. Discussion of how the patient interacts with the therapist is not the same as a transference interpretation. Because transference exists in all human relationships, transferential aspects in a relationship may have positive or negative effects on interactions outside the therapeutic environment. Whether acknowledged or ignored, transference—and thus countertransference—is present.

Transference is experiencing and/or relating to someone in the present as if that person was a significant individual from the past. The concept implies that all personal relationships contain elements of transference(s). That is, we all have the potential to displace or transfer to current situations infantile and internal conflicts that are out of place and thus not appropriate to the present person and/or situation.

Countertransference is a dimensional concept, not an all-or-nothing experience. Some reactions to patients are based entirely upon their transference to us and have nothing to do with us (therapists) as people. Others derive mostly from psychodynamics within the therapist ( Box 2 ). Countertransference has evolved to incorporate responses evoked by a combination of:

  • the patient’s transference
  • the therapist’s unique psychodynamics
  • the real relationship in the therapeutic dyad.2
Box 2

Reactions to other people: When are they countertransference?

In the therapeutic setting, some reactions to the patient are experienced as unusually powerful, out of keeping with our self-image, or as consciously disturbing. Such reactions to a patient—while still countertransference—might result from projective identification. This type of countertransference is most commonly, but not exclusively, encountered in therapy of patients with borderline personality organization.3

We suggest that the term countertransference be restricted to therapeutic situations (any relationship in which one person has the role of treating or helping the other person), including all patient-physician or patient-provider relationships. They have a transferential component because the physician occupies a role of authority/knowledge/power from which the patient seeks to benefit.

Outside of therapeutic situations, reactions to other people are our transferences to them, evoked by our internalized past relationships. We may have an emotional response to how someone behaves toward us (their transference), but that is a counter-transference, not countertransference.

 

 

Patients with medical illness

Psychiatrists think of countertransference as a psychological situation occurring in the office or on an inpatient psychiatric unit. We focus our attention on how we feel and what we think while working with patients. We talk about our reactions to patients in supervision, rounds, case conferences, and other situations where mental health professionals discuss patients.

Our medical/surgical colleagues’ reactions to patients often correlate with certain patient presentations and may have little to do with the actual person who is the patient.4 The medical setting provides an opportunity for countertransference to occur in the absence of apparent transference.

Somatic illness imposes on patients some degree of regression. This regression and attempts to cope with it are inherent to somatic illness and hospitalization. Several schemas5 describe basic coping mechanisms common to most patients ( Table ).6,7 Recognizing a patient’s character style or personality type may help clinicians predict their countertransference when interacting with that patient. Uncooperative patients and those perceived as “difficult” are particularly likely to evoke negative countertransference.8

Table

Patients’ response to illness,
with common countertransference by medical staff

Patient’s coping mechanismsStaff’s countertransference
Dependent personality
• Unconsciously wishes for unlimited care
• Depends on others to feel secure
• May make excessive requests of staff
• Gratification at being able to take care of patient’s needs
• Resentment if patient’s needs seem insatiable
Obsessional personality
• Meticulous self-discipline
• Illness represents loss of control
• Will try to gain mastery over illness by focusing on details, information
• Relief at patient’s willingness to actively participate
• Power struggle is possible
Histrionic personality
• Outgoing, colorful, lively
• Attractiveness and sexuality important
• Needs to feel the center of attention
• Illness represents defect, loss of physical beauty
• Warm initial engagement
• Fear of crossing boundaries
• Wonder about veracity of complaints
Masochistic personality
• Satisfies unconscious needs by suffering
• Needs to play victim role
• Frustration when reassurance does not help
• May unconsciously play into patient’s need for punishment
Paranoid personality
• Pervasive doubt of others’ motivations
• Often questions motives for interventions
• Illness represents threat to safety
• Wary of lack of alliance
• Anger that patient questions treatment motives
• Frustrated at inability to form a trusting relationship with patient
• Unsettled by lack of connection
Narcissistic personality
• Grandiose sense of self, which protects against shame, humiliation
• May demand superior care, insult junior team members
• May feel flattered by ability to treat patient as VIP
• May alternately feel devalued, wonder about competence
Source: References 6,7

CASE CONTINUED: No longer ‘grandmotherly’

Mrs. R and Dr. W are now in a patient-physician relationship. Dr. W is no longer handing Mrs. R a bottle of soda but is inquiring about her life, use of alcohol and other drugs, intimate activities, etc. Mrs. R reacts with anger at the “personal questions.” In addition, Dr. W orders tests that are uncomfortable for Mrs. R, who refuses to cooperate with some procedures.

Dr. W’s memories of her grandmother (who was encouraging, supportive, and loving) color her experience of Mrs. R. She ignores nursing staff’s complaints about Mrs. R being demanding and difficult as the patient becomes aggressive and increasingly confused.

Unable to see the patient as she really is, Dr. W becomes angry and defends Mrs. R’s behavior. The nurses feel Dr. W is unrealistic and ignore her at the nursing station. Late on a Friday night, Mrs. R becomes paranoid, hallucinating that “demons” are in her room. She tries to elope from the hospital. Dr. W is off for the weekend, and the staff requests an emergency psychiatric consultation.

Mrs. R evokes a reaction from the nurses because of how she interacts with them. Dr. W’s response—based on her experience of her grandmother—has nothing to do with the way Mrs. R relates interpersonally but reflects a reaction to the patient’s gender and age. Both reactions would be countertransference, using the modern definition.

If reactions to a patient such as Mrs. R are positive, no one seems to notice and the reactions might or might not influence her care. If the reactions are negative, they might influence her care and generate a request for a psychiatric consultation.

Countertransference might have a negative effect on patient care. For example, if a physician were to avoid Mrs. R because she is uncooperative, and if the nursing staff is intolerant of the patient’s confusion and agitation, she might be labeled as “demented” and be given medication without anyone exploring the etiology of her behavior.
 

 


Some patients cannot communicate because of neurologic disorders, intubation, language barriers, or because they are unconscious when admitted. Without information from the patient, medical staff may form ideas about the patient based on their unconscious fantasies. These fantasies may influence the patient’s care.9 Psychiatric consultants are not immune to countertransference, but we come into situations with the opportunity to experience all participants from the outside.

CASE CONTINUED: The psychiatric consultation

During the interview, the psychiatrist asks Mrs. R if she takes any medications. She retorts that she always takes “Centrum” at bed-time and demands to know why she is not getting her “vitamins.” She is given oxazepam and falls asleep.

The psychiatrist recommends benzodiazepine detoxification, suspecting Mrs. R is taking prazepam at home from an old prescription (when the medication was a brand called “Centrax”). This suspicion is confirmed when Mrs. R’s family brings in a large shopping bag of medications she has collected over decades, and Mrs. R identifies her nighttime “vitamin.”

A full evaluation for delirium is completed over the next 2 days. Mrs. R’s confusion and aggressive behavior respond to oxazepam.

Patients with particular character styles evoke predictable reactions from others, including psychotherapists. Discussing these reactions has been a part of psychiatric training for decades. A subset of patients has been described as “hateful,” as they routinely evoke extremely negative responses.10 Whether their primary disorder is psychiatric, medical, or some of both, these patients evoke strong countertransference reactions.

Psychiatrists may be comfortable discussing a “narcissistic patient, a dependent clinger with borderline features,” but our medical colleagues might not share our comfort with psychiatric jargon.11 It may be more useful to say to medical staff that the patient “thinks of himself as very important, cannot accept his need to be taken care of, and tends to see things in black and white.”

Managing difficult patients

The characterizations that follow describe unconscious reactions to types of individuals who are routinely experienced as “difficult” patients. Some patients may exhibit a mixture of character styles ( Table ) and do not easily fall into 1 category. The concepts can be useful in clarifying the reactions that patients evoke in medical staff.

‘Dependent’ patients. Some patients demand continuous attention but are unaware of their insatiable neediness. Early in treatment, they may evoke positive countertransference because they are intensely grateful for attention. They can be enticing, unconsciously seductive, and gratifying to their doctors. Over time, they drain and exhaust their physicians, who resort to avoidance and wish to get rid of these patients.

Recommendation. Set limits to prevent the patient from feeling rejected or an actual rejection when he or she is transferred to another doctor’s care. Coach physicians to:

  • ask patients to “Tell me what is most important for us to discuss today”
  • be clear how long the visit will last.
Reassuring the patient that other issues will be addressed in the next visit prevents the physician from feeling overwhelmed by the patient consuming too much time.

‘Entitled’ patients. Another type of “difficult” patient projects an air of entitlement, which typically reflects an underlying insatiable neediness. They may use intimidation, guilt, and threats of punishment to get their doctors to provide the care they demand. These patients appear powerful (even though they may possess no special status), and they may be overtly devaluing of the physician while simultaneously demanding special attention.

The doctor resents the patient’s entitlement but develops an expectable countertransference fear that he or she will get in trouble if the demands are not met. Wishes to retaliate and “put the patient in his or her place” are common.

Recommendation. Saying, “It is understandable that you want the best care, and I plan to give you the best care,” makes it clear to the patient that the physician hears the patient’s concerns. Advise the physician to request the patient’s “understanding and compassion” for other patients who also need the physician’s time and attention.

‘Help-rejecting’ patients. “Help-rejecting” patients demand care but show little faith in treatment and do not follow treatment plans. The harder the physician tries to help, the less likely the plan will succeed. For these patients, treatment success evokes a fear of abandonment; thus, treatment must fail to maintain the relationship.

Common countertransference reactions are initial anxiety that the treatment plan was not adequate, followed by anger and depression as the physician feels stuck with a patient for whom nothing works.

 

 

Recommendation. Setting realistic goals for treatment helps the physician guide the patient, who expects to be told not to return the moment he or she gets better. Telling the patient that medical care does not stop when a particular malady is treated speaks to the patient’s fear of being abandoned.

When the patient adheres only partially to the plan and a psychiatric consultant is called for an “uncooperative” patient, help the doctor understand how the patient sees the world. It is the patient’s psychological needs—not the physician’s failure—that control the outcome of the care.

‘Self-destructive’ patients may appear unaware of their dangerous actions. They evoke malice from their doctors, who feel the patients are purposely engaging in life-threatening behaviors. The patients’ unconscious dependence remains unknown as their denial of the consequences of their behavior frightens and angers those involved in their care. Some of these patients cannot be stopped before their actions cause them permanent harm or death.

Recommendation. You might remind the physician that we all are entitled to live our lives as we choose. To decompress intense feelings, advise the physician to share, without blaming the patient, what medical staff can realistically do. Saying “We’ll do the best we can” (rather than “Treatment is useless for someone like you”) permits the patient to receive the degree of care he or she can accept without the physician feeling helpless. Understanding our limitations and obligations is part of using our countertransference to aid in patient care.

CASE CONTINUED: Feeling better

When Dr. W returns on Monday, she angrily calls the psychiatrist to complain that her patient has been placed on a benzodiazepine and at the “implication” that Mrs. R was abusing medication. When they talk in person, the psychiatrist explains the situation to Dr. W and suggests they meet with Mrs. R together.

Mrs. R is embarrassed when told about her behavior, identifies the pill, and admits taking prazepam for several weeks prior to hospitalization. She says she never understood how a vitamin could help her sleep so well. No longer delirious, Mrs. R is pleasant and asks many questions. She is surprised that “so young” a doctor was assigned to her case and asks if the chief of medicine could be brought in, as she is on the board of directors of another hospital. “No offense, dear,” she says to Dr. W; “I’m sure you did an excellent job, but usually only senior doctors take care of me.”

Later, Dr. W talks with the psychiatric consultant about her chance meeting with Mrs. R in the cafeteria and the discord with the nursing staff. She notes that she was doing an elective in another country when her grandmother died. She realizes that her feelings about her grandmother are superimposed on the patient, resulting in an inability to see the patient as she really is.

Dr. W accepts the psychiatrist’s suggestion to repair her relationship with the nurses with an apology. She now notes that Mrs. R is nothing like her grandmother and seems “pretty stuck up.” She is glad to be off the case and accepts the psychiatrist’s idea that Mrs. R’s need to feel important should not make Dr. W feel bad about herself.

Related resources

  • Gabbard GO, ed. Countertransference issues in psychiatric treatment. In: Oldham JM, Riba MB, eds. Review of psychiatry series. Washington, DC: American Psychiatric Publishing, Inc.; 1999.
  • Blumenfield M, Strain JJ, Grossman S. Psychodynamic approach. In: Blumenfield M, Strain JJ, eds. Psychosomatic medicine. Philadelphia, PA: Lippincott, Williams and Wilkins; 2006:817-828.
Drug brand names

  • Oxazepam • Serax
  • Prazepam • Centrax
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. Gabbard GO. Basic principles of psychodynamic psychotherapy. In: Gabbard GO, ed. Psychodynamic psychiatry in clinical practice. Washington, DC: American Psychiatric Publishing, Inc.; 2005:1-30.

2. Harris A. Transference, countertransference, and the real relationship. In: Person ES, Cooper AM, Gabbard GO, eds. Textbook of psychoanalysis. Washington, DC: American Psychiatric Publishing, Inc.; 2005:201-216.

3. Goldstein WN. Clarification of projective identification. Am J Psychiatry. 1991;148:153-161.

4. Kuchariski A, Groves JE. The so-called “inappropriate” psychiatric consultation request on a medical or surgical ward. Int J Psychiatry Med. 1976;7(3):209-220.

5. Groves MA, Muskin PR. Psychological responses to illness. In: Levenson JL, ed. Textbook of psychosomatic medicine. Washington, DC: American Psychiatric Publishing, Inc.; 2005:67-90.

6. Kahana RJ, Bibring GL. Personality types in medical management. In: Zinberg N, ed. Psychiatry and medical practice in a general hospital. New York, NY: International Universities Press; 1964:108-123.

7. Geringer ES, Stern TA. Coping with medical illness: the impact of personality types. Psychosomatics. 1986;27:251-261.

8. Mozian SA, Muskin PR. The difficult patient. In: Barnhill JW, ed. The approach to the psychiatric patient. Washington, DC: American Psychiatric Publishing, Inc.; 2008:192-196.

9. Groves JE. Management of the borderline patient on a medical or surgical ward: the psychiatric consultant’s role. Int J Psychiatry Med. 1975;6:337-348.

10. Groves JE. Taking care of the hateful patient. N Engl J Med. 1978;298:883-887.

11. Pasnau RO. Ten Commandments of medical etiquette for psychiatrists. Psychosomatics. 1985;26(2):128-132.

Article PDF
Author and Disclosure Information

Philip R. Muskin, MD
Professor of clinical psychiatry, Columbia University College of Physicians and Surgeons, chief, consultation-liaison psychiatry, Columbia University Medical Center, faculty, Columbia University Psychoanalytic Center for Training and Research, New York, NY

Lucy A. Epstein, MD
Postdoctoral clinical fellow in psychosomatic medicine, Columbia University College of Physicians and Surgeons, New York, NY

Issue
Current Psychiatry - 08(04)
Publications
Page Number
25-32
Legacy Keywords
countertransference;transference;difficult patients;Philip Muskin;Lucy Epstein
Sections
Author and Disclosure Information

Philip R. Muskin, MD
Professor of clinical psychiatry, Columbia University College of Physicians and Surgeons, chief, consultation-liaison psychiatry, Columbia University Medical Center, faculty, Columbia University Psychoanalytic Center for Training and Research, New York, NY

Lucy A. Epstein, MD
Postdoctoral clinical fellow in psychosomatic medicine, Columbia University College of Physicians and Surgeons, New York, NY

Author and Disclosure Information

Philip R. Muskin, MD
Professor of clinical psychiatry, Columbia University College of Physicians and Surgeons, chief, consultation-liaison psychiatry, Columbia University Medical Center, faculty, Columbia University Psychoanalytic Center for Training and Research, New York, NY

Lucy A. Epstein, MD
Postdoctoral clinical fellow in psychosomatic medicine, Columbia University College of Physicians and Surgeons, New York, NY

Article PDF
Article PDF

WEB AUDIO
Listen to Dr. Muskin discuss the patient-physician dynamic

Two strangers meet in the hospital cafeteria. Mrs. R, an elderly woman, asks Dr. W, a first-year medical resident, for help in getting a bottle of soda from the cooler. Afterward, Dr. W comments to a colleague with whom she is having lunch, “That woman reminds me of my grandmother.”

What does that comment reflect about Dr. W? It is a statement about the doctor’s transference. That is, she is aware of elements about Mrs. R that evoke internal responses appropriate to a prior important relationship.

What if Mrs. R was to subsequently faint, require admission to the hospital, and become Dr. W’s patient? If Dr. W’s comment indicates transference, would the same reaction to Mrs. R now be countertransference? Does that change if the doctor is unaware of emotions Mrs. R evokes? Is it still countertransference whether Dr. W is caring and compassionate, overly involved with Mrs. R, or—unaware of negative feelings associated with “grandmothers”—avoids the patient?

This article explores how complex internal experiences play out in the general medical setting and discusses how psychiatric consultants can help medical/surgical colleagues understand and manage difficult patient-physician relationships.

The therapeutic dyad

Countertransference and transference are concepts embedded in psychodynamic thinking. They are part of how many people think about interpersonal relations, whether or not they use these terms. Countertransference and transference may be conscious, but they always have an unconscious component. Factors that influence what will be transference and countertransference in adult life have both:

  • a biological component because part of personality is genetic
  • a psychological component based upon experiences throughout life ( Box 1 ).1
Box 1

Roots of transference: From a child’s experience of the world

Genetic factors play a role in personality formation. A child’s personality, which emerges early in life, shapes interactions with people who are significant during childhood. Predispositions shape those experiences and influence what people internalize from those relationships.

In adults, many aspects of what we understand as transference—the experience someone has of a figure from the past—originate from the limitations with which children perceive and interpret their experiences. Transference is not truth about a significant past relationship; it is truth as the person experienced other people and now remembers or reacts to individuals who are reminiscent of those from the past.1

Not all psychotherapeutic treatments—and thus not all therapists—use the concept of transference as a therapeutic component. Some therapists who employ transference in treatment will discuss how the patient interacts with the therapist only when the phenomenon interferes with therapy. Interpretation of transference is a therapeutic modality of psychoanalysis and psychodynamic psychotherapy. Discussion of how the patient interacts with the therapist is not the same as a transference interpretation. Because transference exists in all human relationships, transferential aspects in a relationship may have positive or negative effects on interactions outside the therapeutic environment. Whether acknowledged or ignored, transference—and thus countertransference—is present.

Transference is experiencing and/or relating to someone in the present as if that person was a significant individual from the past. The concept implies that all personal relationships contain elements of transference(s). That is, we all have the potential to displace or transfer to current situations infantile and internal conflicts that are out of place and thus not appropriate to the present person and/or situation.

Countertransference is a dimensional concept, not an all-or-nothing experience. Some reactions to patients are based entirely upon their transference to us and have nothing to do with us (therapists) as people. Others derive mostly from psychodynamics within the therapist ( Box 2 ). Countertransference has evolved to incorporate responses evoked by a combination of:

  • the patient’s transference
  • the therapist’s unique psychodynamics
  • the real relationship in the therapeutic dyad.2
Box 2

Reactions to other people: When are they countertransference?

In the therapeutic setting, some reactions to the patient are experienced as unusually powerful, out of keeping with our self-image, or as consciously disturbing. Such reactions to a patient—while still countertransference—might result from projective identification. This type of countertransference is most commonly, but not exclusively, encountered in therapy of patients with borderline personality organization.3

We suggest that the term countertransference be restricted to therapeutic situations (any relationship in which one person has the role of treating or helping the other person), including all patient-physician or patient-provider relationships. They have a transferential component because the physician occupies a role of authority/knowledge/power from which the patient seeks to benefit.

Outside of therapeutic situations, reactions to other people are our transferences to them, evoked by our internalized past relationships. We may have an emotional response to how someone behaves toward us (their transference), but that is a counter-transference, not countertransference.

 

 

Patients with medical illness

Psychiatrists think of countertransference as a psychological situation occurring in the office or on an inpatient psychiatric unit. We focus our attention on how we feel and what we think while working with patients. We talk about our reactions to patients in supervision, rounds, case conferences, and other situations where mental health professionals discuss patients.

Our medical/surgical colleagues’ reactions to patients often correlate with certain patient presentations and may have little to do with the actual person who is the patient.4 The medical setting provides an opportunity for countertransference to occur in the absence of apparent transference.

Somatic illness imposes on patients some degree of regression. This regression and attempts to cope with it are inherent to somatic illness and hospitalization. Several schemas5 describe basic coping mechanisms common to most patients ( Table ).6,7 Recognizing a patient’s character style or personality type may help clinicians predict their countertransference when interacting with that patient. Uncooperative patients and those perceived as “difficult” are particularly likely to evoke negative countertransference.8

Table

Patients’ response to illness,
with common countertransference by medical staff

Patient’s coping mechanismsStaff’s countertransference
Dependent personality
• Unconsciously wishes for unlimited care
• Depends on others to feel secure
• May make excessive requests of staff
• Gratification at being able to take care of patient’s needs
• Resentment if patient’s needs seem insatiable
Obsessional personality
• Meticulous self-discipline
• Illness represents loss of control
• Will try to gain mastery over illness by focusing on details, information
• Relief at patient’s willingness to actively participate
• Power struggle is possible
Histrionic personality
• Outgoing, colorful, lively
• Attractiveness and sexuality important
• Needs to feel the center of attention
• Illness represents defect, loss of physical beauty
• Warm initial engagement
• Fear of crossing boundaries
• Wonder about veracity of complaints
Masochistic personality
• Satisfies unconscious needs by suffering
• Needs to play victim role
• Frustration when reassurance does not help
• May unconsciously play into patient’s need for punishment
Paranoid personality
• Pervasive doubt of others’ motivations
• Often questions motives for interventions
• Illness represents threat to safety
• Wary of lack of alliance
• Anger that patient questions treatment motives
• Frustrated at inability to form a trusting relationship with patient
• Unsettled by lack of connection
Narcissistic personality
• Grandiose sense of self, which protects against shame, humiliation
• May demand superior care, insult junior team members
• May feel flattered by ability to treat patient as VIP
• May alternately feel devalued, wonder about competence
Source: References 6,7

CASE CONTINUED: No longer ‘grandmotherly’

Mrs. R and Dr. W are now in a patient-physician relationship. Dr. W is no longer handing Mrs. R a bottle of soda but is inquiring about her life, use of alcohol and other drugs, intimate activities, etc. Mrs. R reacts with anger at the “personal questions.” In addition, Dr. W orders tests that are uncomfortable for Mrs. R, who refuses to cooperate with some procedures.

Dr. W’s memories of her grandmother (who was encouraging, supportive, and loving) color her experience of Mrs. R. She ignores nursing staff’s complaints about Mrs. R being demanding and difficult as the patient becomes aggressive and increasingly confused.

Unable to see the patient as she really is, Dr. W becomes angry and defends Mrs. R’s behavior. The nurses feel Dr. W is unrealistic and ignore her at the nursing station. Late on a Friday night, Mrs. R becomes paranoid, hallucinating that “demons” are in her room. She tries to elope from the hospital. Dr. W is off for the weekend, and the staff requests an emergency psychiatric consultation.

Mrs. R evokes a reaction from the nurses because of how she interacts with them. Dr. W’s response—based on her experience of her grandmother—has nothing to do with the way Mrs. R relates interpersonally but reflects a reaction to the patient’s gender and age. Both reactions would be countertransference, using the modern definition.

If reactions to a patient such as Mrs. R are positive, no one seems to notice and the reactions might or might not influence her care. If the reactions are negative, they might influence her care and generate a request for a psychiatric consultation.

Countertransference might have a negative effect on patient care. For example, if a physician were to avoid Mrs. R because she is uncooperative, and if the nursing staff is intolerant of the patient’s confusion and agitation, she might be labeled as “demented” and be given medication without anyone exploring the etiology of her behavior.
 

 


Some patients cannot communicate because of neurologic disorders, intubation, language barriers, or because they are unconscious when admitted. Without information from the patient, medical staff may form ideas about the patient based on their unconscious fantasies. These fantasies may influence the patient’s care.9 Psychiatric consultants are not immune to countertransference, but we come into situations with the opportunity to experience all participants from the outside.

CASE CONTINUED: The psychiatric consultation

During the interview, the psychiatrist asks Mrs. R if she takes any medications. She retorts that she always takes “Centrum” at bed-time and demands to know why she is not getting her “vitamins.” She is given oxazepam and falls asleep.

The psychiatrist recommends benzodiazepine detoxification, suspecting Mrs. R is taking prazepam at home from an old prescription (when the medication was a brand called “Centrax”). This suspicion is confirmed when Mrs. R’s family brings in a large shopping bag of medications she has collected over decades, and Mrs. R identifies her nighttime “vitamin.”

A full evaluation for delirium is completed over the next 2 days. Mrs. R’s confusion and aggressive behavior respond to oxazepam.

Patients with particular character styles evoke predictable reactions from others, including psychotherapists. Discussing these reactions has been a part of psychiatric training for decades. A subset of patients has been described as “hateful,” as they routinely evoke extremely negative responses.10 Whether their primary disorder is psychiatric, medical, or some of both, these patients evoke strong countertransference reactions.

Psychiatrists may be comfortable discussing a “narcissistic patient, a dependent clinger with borderline features,” but our medical colleagues might not share our comfort with psychiatric jargon.11 It may be more useful to say to medical staff that the patient “thinks of himself as very important, cannot accept his need to be taken care of, and tends to see things in black and white.”

Managing difficult patients

The characterizations that follow describe unconscious reactions to types of individuals who are routinely experienced as “difficult” patients. Some patients may exhibit a mixture of character styles ( Table ) and do not easily fall into 1 category. The concepts can be useful in clarifying the reactions that patients evoke in medical staff.

‘Dependent’ patients. Some patients demand continuous attention but are unaware of their insatiable neediness. Early in treatment, they may evoke positive countertransference because they are intensely grateful for attention. They can be enticing, unconsciously seductive, and gratifying to their doctors. Over time, they drain and exhaust their physicians, who resort to avoidance and wish to get rid of these patients.

Recommendation. Set limits to prevent the patient from feeling rejected or an actual rejection when he or she is transferred to another doctor’s care. Coach physicians to:

  • ask patients to “Tell me what is most important for us to discuss today”
  • be clear how long the visit will last.
Reassuring the patient that other issues will be addressed in the next visit prevents the physician from feeling overwhelmed by the patient consuming too much time.

‘Entitled’ patients. Another type of “difficult” patient projects an air of entitlement, which typically reflects an underlying insatiable neediness. They may use intimidation, guilt, and threats of punishment to get their doctors to provide the care they demand. These patients appear powerful (even though they may possess no special status), and they may be overtly devaluing of the physician while simultaneously demanding special attention.

The doctor resents the patient’s entitlement but develops an expectable countertransference fear that he or she will get in trouble if the demands are not met. Wishes to retaliate and “put the patient in his or her place” are common.

Recommendation. Saying, “It is understandable that you want the best care, and I plan to give you the best care,” makes it clear to the patient that the physician hears the patient’s concerns. Advise the physician to request the patient’s “understanding and compassion” for other patients who also need the physician’s time and attention.

‘Help-rejecting’ patients. “Help-rejecting” patients demand care but show little faith in treatment and do not follow treatment plans. The harder the physician tries to help, the less likely the plan will succeed. For these patients, treatment success evokes a fear of abandonment; thus, treatment must fail to maintain the relationship.

Common countertransference reactions are initial anxiety that the treatment plan was not adequate, followed by anger and depression as the physician feels stuck with a patient for whom nothing works.

 

 

Recommendation. Setting realistic goals for treatment helps the physician guide the patient, who expects to be told not to return the moment he or she gets better. Telling the patient that medical care does not stop when a particular malady is treated speaks to the patient’s fear of being abandoned.

When the patient adheres only partially to the plan and a psychiatric consultant is called for an “uncooperative” patient, help the doctor understand how the patient sees the world. It is the patient’s psychological needs—not the physician’s failure—that control the outcome of the care.

‘Self-destructive’ patients may appear unaware of their dangerous actions. They evoke malice from their doctors, who feel the patients are purposely engaging in life-threatening behaviors. The patients’ unconscious dependence remains unknown as their denial of the consequences of their behavior frightens and angers those involved in their care. Some of these patients cannot be stopped before their actions cause them permanent harm or death.

Recommendation. You might remind the physician that we all are entitled to live our lives as we choose. To decompress intense feelings, advise the physician to share, without blaming the patient, what medical staff can realistically do. Saying “We’ll do the best we can” (rather than “Treatment is useless for someone like you”) permits the patient to receive the degree of care he or she can accept without the physician feeling helpless. Understanding our limitations and obligations is part of using our countertransference to aid in patient care.

CASE CONTINUED: Feeling better

When Dr. W returns on Monday, she angrily calls the psychiatrist to complain that her patient has been placed on a benzodiazepine and at the “implication” that Mrs. R was abusing medication. When they talk in person, the psychiatrist explains the situation to Dr. W and suggests they meet with Mrs. R together.

Mrs. R is embarrassed when told about her behavior, identifies the pill, and admits taking prazepam for several weeks prior to hospitalization. She says she never understood how a vitamin could help her sleep so well. No longer delirious, Mrs. R is pleasant and asks many questions. She is surprised that “so young” a doctor was assigned to her case and asks if the chief of medicine could be brought in, as she is on the board of directors of another hospital. “No offense, dear,” she says to Dr. W; “I’m sure you did an excellent job, but usually only senior doctors take care of me.”

Later, Dr. W talks with the psychiatric consultant about her chance meeting with Mrs. R in the cafeteria and the discord with the nursing staff. She notes that she was doing an elective in another country when her grandmother died. She realizes that her feelings about her grandmother are superimposed on the patient, resulting in an inability to see the patient as she really is.

Dr. W accepts the psychiatrist’s suggestion to repair her relationship with the nurses with an apology. She now notes that Mrs. R is nothing like her grandmother and seems “pretty stuck up.” She is glad to be off the case and accepts the psychiatrist’s idea that Mrs. R’s need to feel important should not make Dr. W feel bad about herself.

Related resources

  • Gabbard GO, ed. Countertransference issues in psychiatric treatment. In: Oldham JM, Riba MB, eds. Review of psychiatry series. Washington, DC: American Psychiatric Publishing, Inc.; 1999.
  • Blumenfield M, Strain JJ, Grossman S. Psychodynamic approach. In: Blumenfield M, Strain JJ, eds. Psychosomatic medicine. Philadelphia, PA: Lippincott, Williams and Wilkins; 2006:817-828.
Drug brand names

  • Oxazepam • Serax
  • Prazepam • Centrax
Disclosure

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

WEB AUDIO
Listen to Dr. Muskin discuss the patient-physician dynamic

Two strangers meet in the hospital cafeteria. Mrs. R, an elderly woman, asks Dr. W, a first-year medical resident, for help in getting a bottle of soda from the cooler. Afterward, Dr. W comments to a colleague with whom she is having lunch, “That woman reminds me of my grandmother.”

What does that comment reflect about Dr. W? It is a statement about the doctor’s transference. That is, she is aware of elements about Mrs. R that evoke internal responses appropriate to a prior important relationship.

What if Mrs. R was to subsequently faint, require admission to the hospital, and become Dr. W’s patient? If Dr. W’s comment indicates transference, would the same reaction to Mrs. R now be countertransference? Does that change if the doctor is unaware of emotions Mrs. R evokes? Is it still countertransference whether Dr. W is caring and compassionate, overly involved with Mrs. R, or—unaware of negative feelings associated with “grandmothers”—avoids the patient?

This article explores how complex internal experiences play out in the general medical setting and discusses how psychiatric consultants can help medical/surgical colleagues understand and manage difficult patient-physician relationships.

The therapeutic dyad

Countertransference and transference are concepts embedded in psychodynamic thinking. They are part of how many people think about interpersonal relations, whether or not they use these terms. Countertransference and transference may be conscious, but they always have an unconscious component. Factors that influence what will be transference and countertransference in adult life have both:

  • a biological component because part of personality is genetic
  • a psychological component based upon experiences throughout life ( Box 1 ).1
Box 1

Roots of transference: From a child’s experience of the world

Genetic factors play a role in personality formation. A child’s personality, which emerges early in life, shapes interactions with people who are significant during childhood. Predispositions shape those experiences and influence what people internalize from those relationships.

In adults, many aspects of what we understand as transference—the experience someone has of a figure from the past—originate from the limitations with which children perceive and interpret their experiences. Transference is not truth about a significant past relationship; it is truth as the person experienced other people and now remembers or reacts to individuals who are reminiscent of those from the past.1

Not all psychotherapeutic treatments—and thus not all therapists—use the concept of transference as a therapeutic component. Some therapists who employ transference in treatment will discuss how the patient interacts with the therapist only when the phenomenon interferes with therapy. Interpretation of transference is a therapeutic modality of psychoanalysis and psychodynamic psychotherapy. Discussion of how the patient interacts with the therapist is not the same as a transference interpretation. Because transference exists in all human relationships, transferential aspects in a relationship may have positive or negative effects on interactions outside the therapeutic environment. Whether acknowledged or ignored, transference—and thus countertransference—is present.

Transference is experiencing and/or relating to someone in the present as if that person was a significant individual from the past. The concept implies that all personal relationships contain elements of transference(s). That is, we all have the potential to displace or transfer to current situations infantile and internal conflicts that are out of place and thus not appropriate to the present person and/or situation.

Countertransference is a dimensional concept, not an all-or-nothing experience. Some reactions to patients are based entirely upon their transference to us and have nothing to do with us (therapists) as people. Others derive mostly from psychodynamics within the therapist ( Box 2 ). Countertransference has evolved to incorporate responses evoked by a combination of:

  • the patient’s transference
  • the therapist’s unique psychodynamics
  • the real relationship in the therapeutic dyad.2
Box 2

Reactions to other people: When are they countertransference?

In the therapeutic setting, some reactions to the patient are experienced as unusually powerful, out of keeping with our self-image, or as consciously disturbing. Such reactions to a patient—while still countertransference—might result from projective identification. This type of countertransference is most commonly, but not exclusively, encountered in therapy of patients with borderline personality organization.3

We suggest that the term countertransference be restricted to therapeutic situations (any relationship in which one person has the role of treating or helping the other person), including all patient-physician or patient-provider relationships. They have a transferential component because the physician occupies a role of authority/knowledge/power from which the patient seeks to benefit.

Outside of therapeutic situations, reactions to other people are our transferences to them, evoked by our internalized past relationships. We may have an emotional response to how someone behaves toward us (their transference), but that is a counter-transference, not countertransference.

 

 

Patients with medical illness

Psychiatrists think of countertransference as a psychological situation occurring in the office or on an inpatient psychiatric unit. We focus our attention on how we feel and what we think while working with patients. We talk about our reactions to patients in supervision, rounds, case conferences, and other situations where mental health professionals discuss patients.

Our medical/surgical colleagues’ reactions to patients often correlate with certain patient presentations and may have little to do with the actual person who is the patient.4 The medical setting provides an opportunity for countertransference to occur in the absence of apparent transference.

Somatic illness imposes on patients some degree of regression. This regression and attempts to cope with it are inherent to somatic illness and hospitalization. Several schemas5 describe basic coping mechanisms common to most patients ( Table ).6,7 Recognizing a patient’s character style or personality type may help clinicians predict their countertransference when interacting with that patient. Uncooperative patients and those perceived as “difficult” are particularly likely to evoke negative countertransference.8

Table

Patients’ response to illness,
with common countertransference by medical staff

Patient’s coping mechanismsStaff’s countertransference
Dependent personality
• Unconsciously wishes for unlimited care
• Depends on others to feel secure
• May make excessive requests of staff
• Gratification at being able to take care of patient’s needs
• Resentment if patient’s needs seem insatiable
Obsessional personality
• Meticulous self-discipline
• Illness represents loss of control
• Will try to gain mastery over illness by focusing on details, information
• Relief at patient’s willingness to actively participate
• Power struggle is possible
Histrionic personality
• Outgoing, colorful, lively
• Attractiveness and sexuality important
• Needs to feel the center of attention
• Illness represents defect, loss of physical beauty
• Warm initial engagement
• Fear of crossing boundaries
• Wonder about veracity of complaints
Masochistic personality
• Satisfies unconscious needs by suffering
• Needs to play victim role
• Frustration when reassurance does not help
• May unconsciously play into patient’s need for punishment
Paranoid personality
• Pervasive doubt of others’ motivations
• Often questions motives for interventions
• Illness represents threat to safety
• Wary of lack of alliance
• Anger that patient questions treatment motives
• Frustrated at inability to form a trusting relationship with patient
• Unsettled by lack of connection
Narcissistic personality
• Grandiose sense of self, which protects against shame, humiliation
• May demand superior care, insult junior team members
• May feel flattered by ability to treat patient as VIP
• May alternately feel devalued, wonder about competence
Source: References 6,7

CASE CONTINUED: No longer ‘grandmotherly’

Mrs. R and Dr. W are now in a patient-physician relationship. Dr. W is no longer handing Mrs. R a bottle of soda but is inquiring about her life, use of alcohol and other drugs, intimate activities, etc. Mrs. R reacts with anger at the “personal questions.” In addition, Dr. W orders tests that are uncomfortable for Mrs. R, who refuses to cooperate with some procedures.

Dr. W’s memories of her grandmother (who was encouraging, supportive, and loving) color her experience of Mrs. R. She ignores nursing staff’s complaints about Mrs. R being demanding and difficult as the patient becomes aggressive and increasingly confused.

Unable to see the patient as she really is, Dr. W becomes angry and defends Mrs. R’s behavior. The nurses feel Dr. W is unrealistic and ignore her at the nursing station. Late on a Friday night, Mrs. R becomes paranoid, hallucinating that “demons” are in her room. She tries to elope from the hospital. Dr. W is off for the weekend, and the staff requests an emergency psychiatric consultation.

Mrs. R evokes a reaction from the nurses because of how she interacts with them. Dr. W’s response—based on her experience of her grandmother—has nothing to do with the way Mrs. R relates interpersonally but reflects a reaction to the patient’s gender and age. Both reactions would be countertransference, using the modern definition.

If reactions to a patient such as Mrs. R are positive, no one seems to notice and the reactions might or might not influence her care. If the reactions are negative, they might influence her care and generate a request for a psychiatric consultation.

Countertransference might have a negative effect on patient care. For example, if a physician were to avoid Mrs. R because she is uncooperative, and if the nursing staff is intolerant of the patient’s confusion and agitation, she might be labeled as “demented” and be given medication without anyone exploring the etiology of her behavior.
 

 


Some patients cannot communicate because of neurologic disorders, intubation, language barriers, or because they are unconscious when admitted. Without information from the patient, medical staff may form ideas about the patient based on their unconscious fantasies. These fantasies may influence the patient’s care.9 Psychiatric consultants are not immune to countertransference, but we come into situations with the opportunity to experience all participants from the outside.

CASE CONTINUED: The psychiatric consultation

During the interview, the psychiatrist asks Mrs. R if she takes any medications. She retorts that she always takes “Centrum” at bed-time and demands to know why she is not getting her “vitamins.” She is given oxazepam and falls asleep.

The psychiatrist recommends benzodiazepine detoxification, suspecting Mrs. R is taking prazepam at home from an old prescription (when the medication was a brand called “Centrax”). This suspicion is confirmed when Mrs. R’s family brings in a large shopping bag of medications she has collected over decades, and Mrs. R identifies her nighttime “vitamin.”

A full evaluation for delirium is completed over the next 2 days. Mrs. R’s confusion and aggressive behavior respond to oxazepam.

Patients with particular character styles evoke predictable reactions from others, including psychotherapists. Discussing these reactions has been a part of psychiatric training for decades. A subset of patients has been described as “hateful,” as they routinely evoke extremely negative responses.10 Whether their primary disorder is psychiatric, medical, or some of both, these patients evoke strong countertransference reactions.

Psychiatrists may be comfortable discussing a “narcissistic patient, a dependent clinger with borderline features,” but our medical colleagues might not share our comfort with psychiatric jargon.11 It may be more useful to say to medical staff that the patient “thinks of himself as very important, cannot accept his need to be taken care of, and tends to see things in black and white.”

Managing difficult patients

The characterizations that follow describe unconscious reactions to types of individuals who are routinely experienced as “difficult” patients. Some patients may exhibit a mixture of character styles ( Table ) and do not easily fall into 1 category. The concepts can be useful in clarifying the reactions that patients evoke in medical staff.

‘Dependent’ patients. Some patients demand continuous attention but are unaware of their insatiable neediness. Early in treatment, they may evoke positive countertransference because they are intensely grateful for attention. They can be enticing, unconsciously seductive, and gratifying to their doctors. Over time, they drain and exhaust their physicians, who resort to avoidance and wish to get rid of these patients.

Recommendation. Set limits to prevent the patient from feeling rejected or an actual rejection when he or she is transferred to another doctor’s care. Coach physicians to:

  • ask patients to “Tell me what is most important for us to discuss today”
  • be clear how long the visit will last.
Reassuring the patient that other issues will be addressed in the next visit prevents the physician from feeling overwhelmed by the patient consuming too much time.

‘Entitled’ patients. Another type of “difficult” patient projects an air of entitlement, which typically reflects an underlying insatiable neediness. They may use intimidation, guilt, and threats of punishment to get their doctors to provide the care they demand. These patients appear powerful (even though they may possess no special status), and they may be overtly devaluing of the physician while simultaneously demanding special attention.

The doctor resents the patient’s entitlement but develops an expectable countertransference fear that he or she will get in trouble if the demands are not met. Wishes to retaliate and “put the patient in his or her place” are common.

Recommendation. Saying, “It is understandable that you want the best care, and I plan to give you the best care,” makes it clear to the patient that the physician hears the patient’s concerns. Advise the physician to request the patient’s “understanding and compassion” for other patients who also need the physician’s time and attention.

‘Help-rejecting’ patients. “Help-rejecting” patients demand care but show little faith in treatment and do not follow treatment plans. The harder the physician tries to help, the less likely the plan will succeed. For these patients, treatment success evokes a fear of abandonment; thus, treatment must fail to maintain the relationship.

Common countertransference reactions are initial anxiety that the treatment plan was not adequate, followed by anger and depression as the physician feels stuck with a patient for whom nothing works.

 

 

Recommendation. Setting realistic goals for treatment helps the physician guide the patient, who expects to be told not to return the moment he or she gets better. Telling the patient that medical care does not stop when a particular malady is treated speaks to the patient’s fear of being abandoned.

When the patient adheres only partially to the plan and a psychiatric consultant is called for an “uncooperative” patient, help the doctor understand how the patient sees the world. It is the patient’s psychological needs—not the physician’s failure—that control the outcome of the care.

‘Self-destructive’ patients may appear unaware of their dangerous actions. They evoke malice from their doctors, who feel the patients are purposely engaging in life-threatening behaviors. The patients’ unconscious dependence remains unknown as their denial of the consequences of their behavior frightens and angers those involved in their care. Some of these patients cannot be stopped before their actions cause them permanent harm or death.

Recommendation. You might remind the physician that we all are entitled to live our lives as we choose. To decompress intense feelings, advise the physician to share, without blaming the patient, what medical staff can realistically do. Saying “We’ll do the best we can” (rather than “Treatment is useless for someone like you”) permits the patient to receive the degree of care he or she can accept without the physician feeling helpless. Understanding our limitations and obligations is part of using our countertransference to aid in patient care.

CASE CONTINUED: Feeling better

When Dr. W returns on Monday, she angrily calls the psychiatrist to complain that her patient has been placed on a benzodiazepine and at the “implication” that Mrs. R was abusing medication. When they talk in person, the psychiatrist explains the situation to Dr. W and suggests they meet with Mrs. R together.

Mrs. R is embarrassed when told about her behavior, identifies the pill, and admits taking prazepam for several weeks prior to hospitalization. She says she never understood how a vitamin could help her sleep so well. No longer delirious, Mrs. R is pleasant and asks many questions. She is surprised that “so young” a doctor was assigned to her case and asks if the chief of medicine could be brought in, as she is on the board of directors of another hospital. “No offense, dear,” she says to Dr. W; “I’m sure you did an excellent job, but usually only senior doctors take care of me.”

Later, Dr. W talks with the psychiatric consultant about her chance meeting with Mrs. R in the cafeteria and the discord with the nursing staff. She notes that she was doing an elective in another country when her grandmother died. She realizes that her feelings about her grandmother are superimposed on the patient, resulting in an inability to see the patient as she really is.

Dr. W accepts the psychiatrist’s suggestion to repair her relationship with the nurses with an apology. She now notes that Mrs. R is nothing like her grandmother and seems “pretty stuck up.” She is glad to be off the case and accepts the psychiatrist’s idea that Mrs. R’s need to feel important should not make Dr. W feel bad about herself.

Related resources

  • Gabbard GO, ed. Countertransference issues in psychiatric treatment. In: Oldham JM, Riba MB, eds. Review of psychiatry series. Washington, DC: American Psychiatric Publishing, Inc.; 1999.
  • Blumenfield M, Strain JJ, Grossman S. Psychodynamic approach. In: Blumenfield M, Strain JJ, eds. Psychosomatic medicine. Philadelphia, PA: Lippincott, Williams and Wilkins; 2006:817-828.
Drug brand names

  • Oxazepam • Serax
  • Prazepam • Centrax
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. Gabbard GO. Basic principles of psychodynamic psychotherapy. In: Gabbard GO, ed. Psychodynamic psychiatry in clinical practice. Washington, DC: American Psychiatric Publishing, Inc.; 2005:1-30.

2. Harris A. Transference, countertransference, and the real relationship. In: Person ES, Cooper AM, Gabbard GO, eds. Textbook of psychoanalysis. Washington, DC: American Psychiatric Publishing, Inc.; 2005:201-216.

3. Goldstein WN. Clarification of projective identification. Am J Psychiatry. 1991;148:153-161.

4. Kuchariski A, Groves JE. The so-called “inappropriate” psychiatric consultation request on a medical or surgical ward. Int J Psychiatry Med. 1976;7(3):209-220.

5. Groves MA, Muskin PR. Psychological responses to illness. In: Levenson JL, ed. Textbook of psychosomatic medicine. Washington, DC: American Psychiatric Publishing, Inc.; 2005:67-90.

6. Kahana RJ, Bibring GL. Personality types in medical management. In: Zinberg N, ed. Psychiatry and medical practice in a general hospital. New York, NY: International Universities Press; 1964:108-123.

7. Geringer ES, Stern TA. Coping with medical illness: the impact of personality types. Psychosomatics. 1986;27:251-261.

8. Mozian SA, Muskin PR. The difficult patient. In: Barnhill JW, ed. The approach to the psychiatric patient. Washington, DC: American Psychiatric Publishing, Inc.; 2008:192-196.

9. Groves JE. Management of the borderline patient on a medical or surgical ward: the psychiatric consultant’s role. Int J Psychiatry Med. 1975;6:337-348.

10. Groves JE. Taking care of the hateful patient. N Engl J Med. 1978;298:883-887.

11. Pasnau RO. Ten Commandments of medical etiquette for psychiatrists. Psychosomatics. 1985;26(2):128-132.

References

1. Gabbard GO. Basic principles of psychodynamic psychotherapy. In: Gabbard GO, ed. Psychodynamic psychiatry in clinical practice. Washington, DC: American Psychiatric Publishing, Inc.; 2005:1-30.

2. Harris A. Transference, countertransference, and the real relationship. In: Person ES, Cooper AM, Gabbard GO, eds. Textbook of psychoanalysis. Washington, DC: American Psychiatric Publishing, Inc.; 2005:201-216.

3. Goldstein WN. Clarification of projective identification. Am J Psychiatry. 1991;148:153-161.

4. Kuchariski A, Groves JE. The so-called “inappropriate” psychiatric consultation request on a medical or surgical ward. Int J Psychiatry Med. 1976;7(3):209-220.

5. Groves MA, Muskin PR. Psychological responses to illness. In: Levenson JL, ed. Textbook of psychosomatic medicine. Washington, DC: American Psychiatric Publishing, Inc.; 2005:67-90.

6. Kahana RJ, Bibring GL. Personality types in medical management. In: Zinberg N, ed. Psychiatry and medical practice in a general hospital. New York, NY: International Universities Press; 1964:108-123.

7. Geringer ES, Stern TA. Coping with medical illness: the impact of personality types. Psychosomatics. 1986;27:251-261.

8. Mozian SA, Muskin PR. The difficult patient. In: Barnhill JW, ed. The approach to the psychiatric patient. Washington, DC: American Psychiatric Publishing, Inc.; 2008:192-196.

9. Groves JE. Management of the borderline patient on a medical or surgical ward: the psychiatric consultant’s role. Int J Psychiatry Med. 1975;6:337-348.

10. Groves JE. Taking care of the hateful patient. N Engl J Med. 1978;298:883-887.

11. Pasnau RO. Ten Commandments of medical etiquette for psychiatrists. Psychosomatics. 1985;26(2):128-132.

Issue
Current Psychiatry - 08(04)
Issue
Current Psychiatry - 08(04)
Page Number
25-32
Page Number
25-32
Publications
Publications
Article Type
Display Headline
Clinical guide to countertransference: Help medical colleagues deal with ‘difficult’ patients
Display Headline
Clinical guide to countertransference: Help medical colleagues deal with ‘difficult’ patients
Legacy Keywords
countertransference;transference;difficult patients;Philip Muskin;Lucy Epstein
Legacy Keywords
countertransference;transference;difficult patients;Philip Muskin;Lucy Epstein
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Worried about high-dose prescribing? Manage risk for you and your patient

Article Type
Changed
Tue, 12/11/2018 - 15:06
Display Headline
Worried about high-dose prescribing? Manage risk for you and your patient

Comment on this article

Mr. B, age 35, is admitted for the fourth time to the inpatient service with hallucinations and delusions related to chronic schizophrenia. After appropriate attempts to control his symptoms, he has begun to respond to usual treatment with an atypical antipsychotic. He remains a “partial responder,” however, at the maximum FDA-approved dosage listed in the package insert (PI). What do you do next?

Because of this author’s (NSK) dual training in medicine and forensic psychiatry, other clinicians often ask me about patients such as Mr. B. Prescribing for patients who do not respond to standard dosages can create anxiety about going “off-label.” This article describes how to manage potential risk to yourself and your patient by communicating effectively and documenting informed consent.

What are the options?

To effectively treat Mr. B’s symptoms, you could:

  • change medications and start over
  • augment with a second atypical antipsychotic
  • stay with the antipsychotic to which he has shown partial response, but go above the PI dosing.
Each strategy could pose problems, but most psychopharmacologists would choose the third option—the most logical one.

Changing medications is attractive, but the choice of an atypical antipsychotic with relative metabolic neutrality is limited, and “switching” is time-consuming. When a drug begins to show efficacy, most clinicians won’t opt to “change horses midstream”—especially if managed care is pressuring for rapid discharge.

Augmentation introduces polypharmacy and potential drug-drug interactions. Very little evidence guides us in combining antipsychotics, as most manufacturers will never study the coadministration of 2 branded medications with the same indication.

Only a few case reports have described combining atypical antipsychotics.1-4 Moreover, many managed care providers and governmental payers/regulators will not pay for polypharmacy with 2 atypical antipsychotics or will allow it only during cross-tapering from one agent to another.

‘High-dose’ monotherapy is the choice most often taken by clinicians and experts. Pharmaceutical manufacturers study a wide range of doses during medication development. Two pivotal trials form the basis of the New Drug Application for FDA approval and largely dictate the PI language.

Don’t misconstrue the PI dosing as optimal for a specific medication or patient. Historically, FDA-approved dosing for atypical antipsychotics has been too high (risperidone, aripiprazole) or too low (ziprasidone, quetiapine) for many patients we treat, even when the medications are used as indicated. This problem is magnified when clinicians try to make individual patients (N=1) resemble the average pooled analysis of the clinical trial group (N>200) and find that the individual patient may be a low-dose, average-dose, or high-dose responder (Table 1).

Informed prescribing. Polypharmacy is a complex issue because essentially no pharmacokinetic or pharmacodynamic studies have examined the simultaneous use of ≥3 psychotropics. When a pharmacist or drug interaction computer alerts you to a potential drug-drug interaction, the warning is almost always theoretical. No real data exist about coadministering most medications.

Physicians may query a manufacturer about off-label, above-PI dosing data by contacting the company’s medical information department or asking a pharmaceutical representative. What you receive will vary by manufacturer, but in almost every case you will get the safety data you want. Occasionally you also will get efficacy data, which is nice but not crucial. An online literature search of MEDLINE is another way to obtain this information.

Table 1

Patient factors that influence response to medication

Patient body mass, age, race, ethnicity, and gender
Variability in medication absorption
Hepatic metabolizing factors
How ‘sick’ the patient is, compared with those in pivotal clinical trials
Patient’s behavior, lifestyle, habits, and diet
Comorbid medical conditions
Other psychiatric and nonpsychiatric pharmacotherapy

Liability risk?

Every clinician I’ve met prescribes drugs off-label, whether in terms of dose, indication, or age limits in the PI as published in the Physicians’ Desk Reference (PDR).5 Still, nearly all describe to me the following nightmare, in which they “violated” the PI and “something bad” happens.

They are sitting in court on the witness stand, white-knuckled and sweaty, as a plaintiff’s attorney strolls up to them, PDR in hand, and says: “Doctor, isn’t this the Bible, and you violated the Bible?” And thus is born the fear of a malpractice claim, predicated on off-label dosing.

Off-label prescribing is rarely the only issue in a lawsuit, according to Denny Rodriguez, assistant vice president, claims, Professional Risk Management Services (PRMS), Inc.—manager of The Psychiatrists’ Program endorsed by the American Psychiatric Association. When raised, allegations related to off-label prescribing are among many presented by the plaintiff under the rubric of treatment that violated the standard of care.

 

 

Contrary to the plaintiff’s allegations, off-label prescribing rarely violates the standard of care because it has valid clinical and scientific bases. And don’t acknowledge the PDR as “the Bible,” which it is not; it’s a compilation of PIs. The FDA affirms that once a product is approved for marketing, a physician may choose to prescribe it for off-label use (Box).5

Box

FDA statement on off-label prescribing

The FDA acknowledges that doctors need to treat patients and may prescribe medications off-label. As stated in the foreword to the Physicians’ Desk Reference:

The FDA has also recognized that the [Federal Food, Drug, and Cosmetic] Act does not, however, limit the manner in which a physician may use an approved drug. Once a product is approved for marketing, a physician may choose to prescribe it for uses or in treatment regimens or patient populations that are not included in approved drug labeling. The FDA also observes that accepted medical practice includes drug use that is not reflected in approved drug labeling.5

Standard of care

The real issue for practitioners is the “standard of care.” Violating the standard of care—what a similarly trained clinician would do under similar circumstances—is the first step on the slippery slope to malpractice. Here we can be quite sure that the standard of care and evidence-based medicine are in sync and support the use of off-label, high-dose monotherapy.

Properly documenting your reasoning helps to demonstrate that your prescribing meets the standard of care. Always document and obtain informed consent. Also stay up-to-date about:

  • medications you prescribe
  • emerging evidence and safety information
  • appropriate patient monitoring for clinical response and adverse effects.8

Black boxes and bold lettering

The FDA may mandate that a manufacturer highlight certain information on a PI in 3 ways—bold lettering, black-box warning, and red lettering, in order of presumed increasing seriousness. This system is meant to draw prescribers’ attention to potential safety problems with pharmaceutical agents. No psychiatric medications carry red-letter warnings, a classification usually reserved for antineoplastic agents.

At one time the FDA relied on evidenced-based data to determine the need for warnings. Recently, however, when a problem has been identified with one agent, the FDA has tended to require all drugs in that agent’s class to carry similar—if not identical—PI warnings. In psychiatry, the FDA has ordered suicide precautions on all antidepressants and metabolic syndrome/hyperglycemia warnings on all atypical antipsychotics, despite evidence of differences in potential risks associated with medications within classes. For example, clinical trials have shown a higher risk of obesity and diabetes among patients receiving olanzapine compared with those receiving ziprasidone.7

The FDA’s action appears to “level the playing field,” giving patients the misperception that any treatment would carry an equal risk. Therefore, when you prescribe a drug that carries a class-wide warning in its PI, present the evidence in a balanced, objective manner so that the patient can make an informed decision.

Managing risk

Your best protection against liability is to communicate effectively with the patient and document that communication—including informed consent—in the medical record.8 Obtain and document informed consent whenever:

  • you initiate a drug or other treatment
  • treatment extends beyond the PI-recommended maximum dose.
Similarly, when a new side-effect warning or safety information about a medication emerges, update the informed consent discussion and re-obtain and re-document the patient’s consent. When warnings are discussed on the nightly news or the Internet, patients prescribed that medication will expect you to address this. Informed consent discussions are an excellent way to discover and address patients’ concerns and ensure that they have realistic expectations about treatment.

Potential benefits for patients from updating informed consent include:

  • changes in medications or dosages based on the new information
  • closer monitoring of potential side effects and other actions
  • empowerment to make decisions about stopping a medication or trying alternate medications or treatments.
Documentation also reflects individualization of care, the patient’s involvement, and your clinical judgment and decision-making—all critical elements of a record that supports good patient care and protects both patient and clinician.

Documenting informed consent

What to include. View informed consent as an ongoing discussion, not a document that needs to be put into a chart to comply with a legal mandate. Documenting informed consent may be as simple as going through the process and then including pertinent points in the medical record (Table 2). The following is an example of a medical record entry used by one author (NSK) to document an initial informed consent discussion:

 

 

“I have explained to the patient the reasons for prescribing the above medication, the expected benefits and potential side effects, the treatment alternatives and possible risks and benefits of the alternatives, and the expected course w/o treatment. The patient asked appropriate questions and appeared to understand the answers. (I discussed off-label use.) I provided information from the manufacturer (or some other source). The patient has decided to try this medication and to be followed.”

Caveats. Avoid “cutting and pasting” language for each informed consent discussion into each medical record. Make your discussion and its documentation reflect each individual’s treatment plan. If you use a preprinted consent/medications side-effect form (as required by many institutions and clinics), consider entering a personalized notation into the progress notes as needed, such as when:

  • you prescribe medications with high risk for serious side effects
  • you use off-label prescribing that is not customary
  • a patient needs extra assistance to follow the treatment plan.8
The procedure’s formality helps a patient focus on the consent process, making it less likely that he/she will later believe he/she was not adequately informed. The signed form supports the assertion that the consent process took place and establishes at least some of what was disclosed. The signed form and the clinician’s entry in the record documenting the informed consent discussion will be beneficial should malpractice litigation allege consent issues.

Table 2

Informed consent: Pertinent points to document

Proposed treatment
Potential side effects (most common)
Potential side effects (most dangerous)
Potential side effects that might make a patient anxious, such as those included in recent FDA statements, changes in labeling, or advertisers’ consumer marketing messages
Alternatives, including their potential side effects
Course without treatment
Demonstration of patient’s comprehension of warnings and opportunity to ask questions
Preprinted forms. A disadvantage of preprinted forms is the difficulty in knowing what information to include. If the form’s content is very broad, then important information may not be disclosed. If the form is very specific and attempts to list all possible complications, one could presume that any complication not listed was not disclosed. If you incorporate an informed consent form into your practice:

  • include all significant and material risks on the form
  • state on the form that the risks “include, but are not limited to” those listed on the form
  • have thorough informed consent discussions with patients
  • enter into the medical record your discussion and a copy of the form signed by the patient.

What to disclose. Clinicians often struggle with how much information to disclose to patients. In general, include what a reasonable person would need to know to make an informed decision. A practical way to think about this is to ask yourself the following questions:

  • What information would I want a physician to disclose to my loved one (parent, child, spouse, etc.) if I was not present and my loved one needed to give consent to a treatment recommendation?
  • Is this information of the type that a reasonable person could say: “I wouldn’t have consented if the doctor had told me that”? If you think so, then provide this information to your patient.
Patient resources. Medication information sheets can enhance informed consent and patients’ understanding and retention of information about medications you prescribe. The FDA’s Web site (www.fda.gov) offers printable patient education sheets on hundreds of medications, medication guides, and other resources (see Related Resources).6 Many manufacturers also offer patient education information at their Web sites, via pharmaceutical representatives, and as part of the PI.

Related resources

Drug brand names

  • Aripiprazole • Abilify
  • Olanzapine • Zyprexa
  • Quetiapine • Seroquel
  • Risperidone • Risperdal
  • Ziprasidone • Geodon
Disclosures

Dr. Kaye receives research support from Pfizer Inc. and Takeda Pharmaceutical and is a consultant to and speaker for Pfizer Inc., AstraZeneca, and GlaxoSmithKline.

Jacqueline Melonas is a full-time employee of PRMS, Inc. PRMS, Inc. contracts with Eli Lilly and Company to provide risk management content related to the management of medical malpractice liability.

References

1. Kaye NS. Ziprasidone augmentation of clozapine in 11 patients. J Clin Psychiatry. 2003;64:215-216.

2. Freudenreich O, Goff DC. Antipsychotic combination therapy in schizophrenia: a review of efficacy and risks of current combinations. Acta Psychiatr Scand. 2002;106:323-330.

3. Pies R. Combining antipsychotics: risks and benefits. Psychopharm Rev. 2001;36(2):9-13.

4. Stahl SM. Antipsychotic polypharmacy, part 1: therapeutic option or dirty little secret? J Clin Psychiatry. 1999;60:425-426.

5. Physicians’ Desk Reference. 62nd ed. Montvale, NJ: Thompson Publishing, 2008.

6. Medwatch. Medical product safety information. Food and Drug Administration. Available at: http://www.fda.gov/medwatch/safety.htm. Accessed July 30, 2008.

7. American Diabetes Association, American Psychiatric Association, American Association of Clinical Endocrinologists, North American Association for the Study of Obesity. Consensus development conference on antipsychotic drugs and obesity and diabetes. Diabetes Care. 2004;27:596-601.

8. Melonas JM. Preventing and reducing professional liability risk related to psychopharmacology. Psychiatric Times. 2005;23(14).-Available at http://www.psychiatrictimes.com/display/article/10168/48416. Accessed September 3, 2008.

Article PDF
Author and Disclosure Information

Neil S. Kaye, MD, DFAPA
Assistant clinical professor of psychiatry and human behavior, assistant clinical professor of family medicine, Jefferson Medical College, Philadelphia, PA

Jacqueline M. Melonas, RN, MS, JD
Vice president, risk management, Professional Risk Management Services, Inc., Arlington, VA

Issue
Current Psychiatry - 08(04)
Publications
Page Number
17-23
Legacy Keywords
high dose prescribing;off-label prescribing; offlabel prescribing;informed consent;liability risk;managing risk;standard of care;documentation;Neil Kaye;Jacqueline Melonas
Sections
Author and Disclosure Information

Neil S. Kaye, MD, DFAPA
Assistant clinical professor of psychiatry and human behavior, assistant clinical professor of family medicine, Jefferson Medical College, Philadelphia, PA

Jacqueline M. Melonas, RN, MS, JD
Vice president, risk management, Professional Risk Management Services, Inc., Arlington, VA

Author and Disclosure Information

Neil S. Kaye, MD, DFAPA
Assistant clinical professor of psychiatry and human behavior, assistant clinical professor of family medicine, Jefferson Medical College, Philadelphia, PA

Jacqueline M. Melonas, RN, MS, JD
Vice president, risk management, Professional Risk Management Services, Inc., Arlington, VA

Article PDF
Article PDF

Comment on this article

Mr. B, age 35, is admitted for the fourth time to the inpatient service with hallucinations and delusions related to chronic schizophrenia. After appropriate attempts to control his symptoms, he has begun to respond to usual treatment with an atypical antipsychotic. He remains a “partial responder,” however, at the maximum FDA-approved dosage listed in the package insert (PI). What do you do next?

Because of this author’s (NSK) dual training in medicine and forensic psychiatry, other clinicians often ask me about patients such as Mr. B. Prescribing for patients who do not respond to standard dosages can create anxiety about going “off-label.” This article describes how to manage potential risk to yourself and your patient by communicating effectively and documenting informed consent.

What are the options?

To effectively treat Mr. B’s symptoms, you could:

  • change medications and start over
  • augment with a second atypical antipsychotic
  • stay with the antipsychotic to which he has shown partial response, but go above the PI dosing.
Each strategy could pose problems, but most psychopharmacologists would choose the third option—the most logical one.

Changing medications is attractive, but the choice of an atypical antipsychotic with relative metabolic neutrality is limited, and “switching” is time-consuming. When a drug begins to show efficacy, most clinicians won’t opt to “change horses midstream”—especially if managed care is pressuring for rapid discharge.

Augmentation introduces polypharmacy and potential drug-drug interactions. Very little evidence guides us in combining antipsychotics, as most manufacturers will never study the coadministration of 2 branded medications with the same indication.

Only a few case reports have described combining atypical antipsychotics.1-4 Moreover, many managed care providers and governmental payers/regulators will not pay for polypharmacy with 2 atypical antipsychotics or will allow it only during cross-tapering from one agent to another.

‘High-dose’ monotherapy is the choice most often taken by clinicians and experts. Pharmaceutical manufacturers study a wide range of doses during medication development. Two pivotal trials form the basis of the New Drug Application for FDA approval and largely dictate the PI language.

Don’t misconstrue the PI dosing as optimal for a specific medication or patient. Historically, FDA-approved dosing for atypical antipsychotics has been too high (risperidone, aripiprazole) or too low (ziprasidone, quetiapine) for many patients we treat, even when the medications are used as indicated. This problem is magnified when clinicians try to make individual patients (N=1) resemble the average pooled analysis of the clinical trial group (N>200) and find that the individual patient may be a low-dose, average-dose, or high-dose responder (Table 1).

Informed prescribing. Polypharmacy is a complex issue because essentially no pharmacokinetic or pharmacodynamic studies have examined the simultaneous use of ≥3 psychotropics. When a pharmacist or drug interaction computer alerts you to a potential drug-drug interaction, the warning is almost always theoretical. No real data exist about coadministering most medications.

Physicians may query a manufacturer about off-label, above-PI dosing data by contacting the company’s medical information department or asking a pharmaceutical representative. What you receive will vary by manufacturer, but in almost every case you will get the safety data you want. Occasionally you also will get efficacy data, which is nice but not crucial. An online literature search of MEDLINE is another way to obtain this information.

Table 1

Patient factors that influence response to medication

Patient body mass, age, race, ethnicity, and gender
Variability in medication absorption
Hepatic metabolizing factors
How ‘sick’ the patient is, compared with those in pivotal clinical trials
Patient’s behavior, lifestyle, habits, and diet
Comorbid medical conditions
Other psychiatric and nonpsychiatric pharmacotherapy

Liability risk?

Every clinician I’ve met prescribes drugs off-label, whether in terms of dose, indication, or age limits in the PI as published in the Physicians’ Desk Reference (PDR).5 Still, nearly all describe to me the following nightmare, in which they “violated” the PI and “something bad” happens.

They are sitting in court on the witness stand, white-knuckled and sweaty, as a plaintiff’s attorney strolls up to them, PDR in hand, and says: “Doctor, isn’t this the Bible, and you violated the Bible?” And thus is born the fear of a malpractice claim, predicated on off-label dosing.

Off-label prescribing is rarely the only issue in a lawsuit, according to Denny Rodriguez, assistant vice president, claims, Professional Risk Management Services (PRMS), Inc.—manager of The Psychiatrists’ Program endorsed by the American Psychiatric Association. When raised, allegations related to off-label prescribing are among many presented by the plaintiff under the rubric of treatment that violated the standard of care.

 

 

Contrary to the plaintiff’s allegations, off-label prescribing rarely violates the standard of care because it has valid clinical and scientific bases. And don’t acknowledge the PDR as “the Bible,” which it is not; it’s a compilation of PIs. The FDA affirms that once a product is approved for marketing, a physician may choose to prescribe it for off-label use (Box).5

Box

FDA statement on off-label prescribing

The FDA acknowledges that doctors need to treat patients and may prescribe medications off-label. As stated in the foreword to the Physicians’ Desk Reference:

The FDA has also recognized that the [Federal Food, Drug, and Cosmetic] Act does not, however, limit the manner in which a physician may use an approved drug. Once a product is approved for marketing, a physician may choose to prescribe it for uses or in treatment regimens or patient populations that are not included in approved drug labeling. The FDA also observes that accepted medical practice includes drug use that is not reflected in approved drug labeling.5

Standard of care

The real issue for practitioners is the “standard of care.” Violating the standard of care—what a similarly trained clinician would do under similar circumstances—is the first step on the slippery slope to malpractice. Here we can be quite sure that the standard of care and evidence-based medicine are in sync and support the use of off-label, high-dose monotherapy.

Properly documenting your reasoning helps to demonstrate that your prescribing meets the standard of care. Always document and obtain informed consent. Also stay up-to-date about:

  • medications you prescribe
  • emerging evidence and safety information
  • appropriate patient monitoring for clinical response and adverse effects.8

Black boxes and bold lettering

The FDA may mandate that a manufacturer highlight certain information on a PI in 3 ways—bold lettering, black-box warning, and red lettering, in order of presumed increasing seriousness. This system is meant to draw prescribers’ attention to potential safety problems with pharmaceutical agents. No psychiatric medications carry red-letter warnings, a classification usually reserved for antineoplastic agents.

At one time the FDA relied on evidenced-based data to determine the need for warnings. Recently, however, when a problem has been identified with one agent, the FDA has tended to require all drugs in that agent’s class to carry similar—if not identical—PI warnings. In psychiatry, the FDA has ordered suicide precautions on all antidepressants and metabolic syndrome/hyperglycemia warnings on all atypical antipsychotics, despite evidence of differences in potential risks associated with medications within classes. For example, clinical trials have shown a higher risk of obesity and diabetes among patients receiving olanzapine compared with those receiving ziprasidone.7

The FDA’s action appears to “level the playing field,” giving patients the misperception that any treatment would carry an equal risk. Therefore, when you prescribe a drug that carries a class-wide warning in its PI, present the evidence in a balanced, objective manner so that the patient can make an informed decision.

Managing risk

Your best protection against liability is to communicate effectively with the patient and document that communication—including informed consent—in the medical record.8 Obtain and document informed consent whenever:

  • you initiate a drug or other treatment
  • treatment extends beyond the PI-recommended maximum dose.
Similarly, when a new side-effect warning or safety information about a medication emerges, update the informed consent discussion and re-obtain and re-document the patient’s consent. When warnings are discussed on the nightly news or the Internet, patients prescribed that medication will expect you to address this. Informed consent discussions are an excellent way to discover and address patients’ concerns and ensure that they have realistic expectations about treatment.

Potential benefits for patients from updating informed consent include:

  • changes in medications or dosages based on the new information
  • closer monitoring of potential side effects and other actions
  • empowerment to make decisions about stopping a medication or trying alternate medications or treatments.
Documentation also reflects individualization of care, the patient’s involvement, and your clinical judgment and decision-making—all critical elements of a record that supports good patient care and protects both patient and clinician.

Documenting informed consent

What to include. View informed consent as an ongoing discussion, not a document that needs to be put into a chart to comply with a legal mandate. Documenting informed consent may be as simple as going through the process and then including pertinent points in the medical record (Table 2). The following is an example of a medical record entry used by one author (NSK) to document an initial informed consent discussion:

 

 

“I have explained to the patient the reasons for prescribing the above medication, the expected benefits and potential side effects, the treatment alternatives and possible risks and benefits of the alternatives, and the expected course w/o treatment. The patient asked appropriate questions and appeared to understand the answers. (I discussed off-label use.) I provided information from the manufacturer (or some other source). The patient has decided to try this medication and to be followed.”

Caveats. Avoid “cutting and pasting” language for each informed consent discussion into each medical record. Make your discussion and its documentation reflect each individual’s treatment plan. If you use a preprinted consent/medications side-effect form (as required by many institutions and clinics), consider entering a personalized notation into the progress notes as needed, such as when:

  • you prescribe medications with high risk for serious side effects
  • you use off-label prescribing that is not customary
  • a patient needs extra assistance to follow the treatment plan.8
The procedure’s formality helps a patient focus on the consent process, making it less likely that he/she will later believe he/she was not adequately informed. The signed form supports the assertion that the consent process took place and establishes at least some of what was disclosed. The signed form and the clinician’s entry in the record documenting the informed consent discussion will be beneficial should malpractice litigation allege consent issues.

Table 2

Informed consent: Pertinent points to document

Proposed treatment
Potential side effects (most common)
Potential side effects (most dangerous)
Potential side effects that might make a patient anxious, such as those included in recent FDA statements, changes in labeling, or advertisers’ consumer marketing messages
Alternatives, including their potential side effects
Course without treatment
Demonstration of patient’s comprehension of warnings and opportunity to ask questions
Preprinted forms. A disadvantage of preprinted forms is the difficulty in knowing what information to include. If the form’s content is very broad, then important information may not be disclosed. If the form is very specific and attempts to list all possible complications, one could presume that any complication not listed was not disclosed. If you incorporate an informed consent form into your practice:

  • include all significant and material risks on the form
  • state on the form that the risks “include, but are not limited to” those listed on the form
  • have thorough informed consent discussions with patients
  • enter into the medical record your discussion and a copy of the form signed by the patient.

What to disclose. Clinicians often struggle with how much information to disclose to patients. In general, include what a reasonable person would need to know to make an informed decision. A practical way to think about this is to ask yourself the following questions:

  • What information would I want a physician to disclose to my loved one (parent, child, spouse, etc.) if I was not present and my loved one needed to give consent to a treatment recommendation?
  • Is this information of the type that a reasonable person could say: “I wouldn’t have consented if the doctor had told me that”? If you think so, then provide this information to your patient.
Patient resources. Medication information sheets can enhance informed consent and patients’ understanding and retention of information about medications you prescribe. The FDA’s Web site (www.fda.gov) offers printable patient education sheets on hundreds of medications, medication guides, and other resources (see Related Resources).6 Many manufacturers also offer patient education information at their Web sites, via pharmaceutical representatives, and as part of the PI.

Related resources

Drug brand names

  • Aripiprazole • Abilify
  • Olanzapine • Zyprexa
  • Quetiapine • Seroquel
  • Risperidone • Risperdal
  • Ziprasidone • Geodon
Disclosures

Dr. Kaye receives research support from Pfizer Inc. and Takeda Pharmaceutical and is a consultant to and speaker for Pfizer Inc., AstraZeneca, and GlaxoSmithKline.

Jacqueline Melonas is a full-time employee of PRMS, Inc. PRMS, Inc. contracts with Eli Lilly and Company to provide risk management content related to the management of medical malpractice liability.

Comment on this article

Mr. B, age 35, is admitted for the fourth time to the inpatient service with hallucinations and delusions related to chronic schizophrenia. After appropriate attempts to control his symptoms, he has begun to respond to usual treatment with an atypical antipsychotic. He remains a “partial responder,” however, at the maximum FDA-approved dosage listed in the package insert (PI). What do you do next?

Because of this author’s (NSK) dual training in medicine and forensic psychiatry, other clinicians often ask me about patients such as Mr. B. Prescribing for patients who do not respond to standard dosages can create anxiety about going “off-label.” This article describes how to manage potential risk to yourself and your patient by communicating effectively and documenting informed consent.

What are the options?

To effectively treat Mr. B’s symptoms, you could:

  • change medications and start over
  • augment with a second atypical antipsychotic
  • stay with the antipsychotic to which he has shown partial response, but go above the PI dosing.
Each strategy could pose problems, but most psychopharmacologists would choose the third option—the most logical one.

Changing medications is attractive, but the choice of an atypical antipsychotic with relative metabolic neutrality is limited, and “switching” is time-consuming. When a drug begins to show efficacy, most clinicians won’t opt to “change horses midstream”—especially if managed care is pressuring for rapid discharge.

Augmentation introduces polypharmacy and potential drug-drug interactions. Very little evidence guides us in combining antipsychotics, as most manufacturers will never study the coadministration of 2 branded medications with the same indication.

Only a few case reports have described combining atypical antipsychotics.1-4 Moreover, many managed care providers and governmental payers/regulators will not pay for polypharmacy with 2 atypical antipsychotics or will allow it only during cross-tapering from one agent to another.

‘High-dose’ monotherapy is the choice most often taken by clinicians and experts. Pharmaceutical manufacturers study a wide range of doses during medication development. Two pivotal trials form the basis of the New Drug Application for FDA approval and largely dictate the PI language.

Don’t misconstrue the PI dosing as optimal for a specific medication or patient. Historically, FDA-approved dosing for atypical antipsychotics has been too high (risperidone, aripiprazole) or too low (ziprasidone, quetiapine) for many patients we treat, even when the medications are used as indicated. This problem is magnified when clinicians try to make individual patients (N=1) resemble the average pooled analysis of the clinical trial group (N>200) and find that the individual patient may be a low-dose, average-dose, or high-dose responder (Table 1).

Informed prescribing. Polypharmacy is a complex issue because essentially no pharmacokinetic or pharmacodynamic studies have examined the simultaneous use of ≥3 psychotropics. When a pharmacist or drug interaction computer alerts you to a potential drug-drug interaction, the warning is almost always theoretical. No real data exist about coadministering most medications.

Physicians may query a manufacturer about off-label, above-PI dosing data by contacting the company’s medical information department or asking a pharmaceutical representative. What you receive will vary by manufacturer, but in almost every case you will get the safety data you want. Occasionally you also will get efficacy data, which is nice but not crucial. An online literature search of MEDLINE is another way to obtain this information.

Table 1

Patient factors that influence response to medication

Patient body mass, age, race, ethnicity, and gender
Variability in medication absorption
Hepatic metabolizing factors
How ‘sick’ the patient is, compared with those in pivotal clinical trials
Patient’s behavior, lifestyle, habits, and diet
Comorbid medical conditions
Other psychiatric and nonpsychiatric pharmacotherapy

Liability risk?

Every clinician I’ve met prescribes drugs off-label, whether in terms of dose, indication, or age limits in the PI as published in the Physicians’ Desk Reference (PDR).5 Still, nearly all describe to me the following nightmare, in which they “violated” the PI and “something bad” happens.

They are sitting in court on the witness stand, white-knuckled and sweaty, as a plaintiff’s attorney strolls up to them, PDR in hand, and says: “Doctor, isn’t this the Bible, and you violated the Bible?” And thus is born the fear of a malpractice claim, predicated on off-label dosing.

Off-label prescribing is rarely the only issue in a lawsuit, according to Denny Rodriguez, assistant vice president, claims, Professional Risk Management Services (PRMS), Inc.—manager of The Psychiatrists’ Program endorsed by the American Psychiatric Association. When raised, allegations related to off-label prescribing are among many presented by the plaintiff under the rubric of treatment that violated the standard of care.

 

 

Contrary to the plaintiff’s allegations, off-label prescribing rarely violates the standard of care because it has valid clinical and scientific bases. And don’t acknowledge the PDR as “the Bible,” which it is not; it’s a compilation of PIs. The FDA affirms that once a product is approved for marketing, a physician may choose to prescribe it for off-label use (Box).5

Box

FDA statement on off-label prescribing

The FDA acknowledges that doctors need to treat patients and may prescribe medications off-label. As stated in the foreword to the Physicians’ Desk Reference:

The FDA has also recognized that the [Federal Food, Drug, and Cosmetic] Act does not, however, limit the manner in which a physician may use an approved drug. Once a product is approved for marketing, a physician may choose to prescribe it for uses or in treatment regimens or patient populations that are not included in approved drug labeling. The FDA also observes that accepted medical practice includes drug use that is not reflected in approved drug labeling.5

Standard of care

The real issue for practitioners is the “standard of care.” Violating the standard of care—what a similarly trained clinician would do under similar circumstances—is the first step on the slippery slope to malpractice. Here we can be quite sure that the standard of care and evidence-based medicine are in sync and support the use of off-label, high-dose monotherapy.

Properly documenting your reasoning helps to demonstrate that your prescribing meets the standard of care. Always document and obtain informed consent. Also stay up-to-date about:

  • medications you prescribe
  • emerging evidence and safety information
  • appropriate patient monitoring for clinical response and adverse effects.8

Black boxes and bold lettering

The FDA may mandate that a manufacturer highlight certain information on a PI in 3 ways—bold lettering, black-box warning, and red lettering, in order of presumed increasing seriousness. This system is meant to draw prescribers’ attention to potential safety problems with pharmaceutical agents. No psychiatric medications carry red-letter warnings, a classification usually reserved for antineoplastic agents.

At one time the FDA relied on evidenced-based data to determine the need for warnings. Recently, however, when a problem has been identified with one agent, the FDA has tended to require all drugs in that agent’s class to carry similar—if not identical—PI warnings. In psychiatry, the FDA has ordered suicide precautions on all antidepressants and metabolic syndrome/hyperglycemia warnings on all atypical antipsychotics, despite evidence of differences in potential risks associated with medications within classes. For example, clinical trials have shown a higher risk of obesity and diabetes among patients receiving olanzapine compared with those receiving ziprasidone.7

The FDA’s action appears to “level the playing field,” giving patients the misperception that any treatment would carry an equal risk. Therefore, when you prescribe a drug that carries a class-wide warning in its PI, present the evidence in a balanced, objective manner so that the patient can make an informed decision.

Managing risk

Your best protection against liability is to communicate effectively with the patient and document that communication—including informed consent—in the medical record.8 Obtain and document informed consent whenever:

  • you initiate a drug or other treatment
  • treatment extends beyond the PI-recommended maximum dose.
Similarly, when a new side-effect warning or safety information about a medication emerges, update the informed consent discussion and re-obtain and re-document the patient’s consent. When warnings are discussed on the nightly news or the Internet, patients prescribed that medication will expect you to address this. Informed consent discussions are an excellent way to discover and address patients’ concerns and ensure that they have realistic expectations about treatment.

Potential benefits for patients from updating informed consent include:

  • changes in medications or dosages based on the new information
  • closer monitoring of potential side effects and other actions
  • empowerment to make decisions about stopping a medication or trying alternate medications or treatments.
Documentation also reflects individualization of care, the patient’s involvement, and your clinical judgment and decision-making—all critical elements of a record that supports good patient care and protects both patient and clinician.

Documenting informed consent

What to include. View informed consent as an ongoing discussion, not a document that needs to be put into a chart to comply with a legal mandate. Documenting informed consent may be as simple as going through the process and then including pertinent points in the medical record (Table 2). The following is an example of a medical record entry used by one author (NSK) to document an initial informed consent discussion:

 

 

“I have explained to the patient the reasons for prescribing the above medication, the expected benefits and potential side effects, the treatment alternatives and possible risks and benefits of the alternatives, and the expected course w/o treatment. The patient asked appropriate questions and appeared to understand the answers. (I discussed off-label use.) I provided information from the manufacturer (or some other source). The patient has decided to try this medication and to be followed.”

Caveats. Avoid “cutting and pasting” language for each informed consent discussion into each medical record. Make your discussion and its documentation reflect each individual’s treatment plan. If you use a preprinted consent/medications side-effect form (as required by many institutions and clinics), consider entering a personalized notation into the progress notes as needed, such as when:

  • you prescribe medications with high risk for serious side effects
  • you use off-label prescribing that is not customary
  • a patient needs extra assistance to follow the treatment plan.8
The procedure’s formality helps a patient focus on the consent process, making it less likely that he/she will later believe he/she was not adequately informed. The signed form supports the assertion that the consent process took place and establishes at least some of what was disclosed. The signed form and the clinician’s entry in the record documenting the informed consent discussion will be beneficial should malpractice litigation allege consent issues.

Table 2

Informed consent: Pertinent points to document

Proposed treatment
Potential side effects (most common)
Potential side effects (most dangerous)
Potential side effects that might make a patient anxious, such as those included in recent FDA statements, changes in labeling, or advertisers’ consumer marketing messages
Alternatives, including their potential side effects
Course without treatment
Demonstration of patient’s comprehension of warnings and opportunity to ask questions
Preprinted forms. A disadvantage of preprinted forms is the difficulty in knowing what information to include. If the form’s content is very broad, then important information may not be disclosed. If the form is very specific and attempts to list all possible complications, one could presume that any complication not listed was not disclosed. If you incorporate an informed consent form into your practice:

  • include all significant and material risks on the form
  • state on the form that the risks “include, but are not limited to” those listed on the form
  • have thorough informed consent discussions with patients
  • enter into the medical record your discussion and a copy of the form signed by the patient.

What to disclose. Clinicians often struggle with how much information to disclose to patients. In general, include what a reasonable person would need to know to make an informed decision. A practical way to think about this is to ask yourself the following questions:

  • What information would I want a physician to disclose to my loved one (parent, child, spouse, etc.) if I was not present and my loved one needed to give consent to a treatment recommendation?
  • Is this information of the type that a reasonable person could say: “I wouldn’t have consented if the doctor had told me that”? If you think so, then provide this information to your patient.
Patient resources. Medication information sheets can enhance informed consent and patients’ understanding and retention of information about medications you prescribe. The FDA’s Web site (www.fda.gov) offers printable patient education sheets on hundreds of medications, medication guides, and other resources (see Related Resources).6 Many manufacturers also offer patient education information at their Web sites, via pharmaceutical representatives, and as part of the PI.

Related resources

Drug brand names

  • Aripiprazole • Abilify
  • Olanzapine • Zyprexa
  • Quetiapine • Seroquel
  • Risperidone • Risperdal
  • Ziprasidone • Geodon
Disclosures

Dr. Kaye receives research support from Pfizer Inc. and Takeda Pharmaceutical and is a consultant to and speaker for Pfizer Inc., AstraZeneca, and GlaxoSmithKline.

Jacqueline Melonas is a full-time employee of PRMS, Inc. PRMS, Inc. contracts with Eli Lilly and Company to provide risk management content related to the management of medical malpractice liability.

References

1. Kaye NS. Ziprasidone augmentation of clozapine in 11 patients. J Clin Psychiatry. 2003;64:215-216.

2. Freudenreich O, Goff DC. Antipsychotic combination therapy in schizophrenia: a review of efficacy and risks of current combinations. Acta Psychiatr Scand. 2002;106:323-330.

3. Pies R. Combining antipsychotics: risks and benefits. Psychopharm Rev. 2001;36(2):9-13.

4. Stahl SM. Antipsychotic polypharmacy, part 1: therapeutic option or dirty little secret? J Clin Psychiatry. 1999;60:425-426.

5. Physicians’ Desk Reference. 62nd ed. Montvale, NJ: Thompson Publishing, 2008.

6. Medwatch. Medical product safety information. Food and Drug Administration. Available at: http://www.fda.gov/medwatch/safety.htm. Accessed July 30, 2008.

7. American Diabetes Association, American Psychiatric Association, American Association of Clinical Endocrinologists, North American Association for the Study of Obesity. Consensus development conference on antipsychotic drugs and obesity and diabetes. Diabetes Care. 2004;27:596-601.

8. Melonas JM. Preventing and reducing professional liability risk related to psychopharmacology. Psychiatric Times. 2005;23(14).-Available at http://www.psychiatrictimes.com/display/article/10168/48416. Accessed September 3, 2008.

References

1. Kaye NS. Ziprasidone augmentation of clozapine in 11 patients. J Clin Psychiatry. 2003;64:215-216.

2. Freudenreich O, Goff DC. Antipsychotic combination therapy in schizophrenia: a review of efficacy and risks of current combinations. Acta Psychiatr Scand. 2002;106:323-330.

3. Pies R. Combining antipsychotics: risks and benefits. Psychopharm Rev. 2001;36(2):9-13.

4. Stahl SM. Antipsychotic polypharmacy, part 1: therapeutic option or dirty little secret? J Clin Psychiatry. 1999;60:425-426.

5. Physicians’ Desk Reference. 62nd ed. Montvale, NJ: Thompson Publishing, 2008.

6. Medwatch. Medical product safety information. Food and Drug Administration. Available at: http://www.fda.gov/medwatch/safety.htm. Accessed July 30, 2008.

7. American Diabetes Association, American Psychiatric Association, American Association of Clinical Endocrinologists, North American Association for the Study of Obesity. Consensus development conference on antipsychotic drugs and obesity and diabetes. Diabetes Care. 2004;27:596-601.

8. Melonas JM. Preventing and reducing professional liability risk related to psychopharmacology. Psychiatric Times. 2005;23(14).-Available at http://www.psychiatrictimes.com/display/article/10168/48416. Accessed September 3, 2008.

Issue
Current Psychiatry - 08(04)
Issue
Current Psychiatry - 08(04)
Page Number
17-23
Page Number
17-23
Publications
Publications
Article Type
Display Headline
Worried about high-dose prescribing? Manage risk for you and your patient
Display Headline
Worried about high-dose prescribing? Manage risk for you and your patient
Legacy Keywords
high dose prescribing;off-label prescribing; offlabel prescribing;informed consent;liability risk;managing risk;standard of care;documentation;Neil Kaye;Jacqueline Melonas
Legacy Keywords
high dose prescribing;off-label prescribing; offlabel prescribing;informed consent;liability risk;managing risk;standard of care;documentation;Neil Kaye;Jacqueline Melonas
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Diabetes screening: Which patients, what tests, and how often?

Article Type
Changed
Tue, 12/11/2018 - 15:06
Display Headline
Diabetes screening: Which patients, what tests, and how often?

Dr. Keenan is associate professor, department of medicine, University of California, Davis.

Principal Source: U.S. Preventive Services Task Force. Screening for type 2 diabetes mellitus in adults: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2008;148(11):846-854.—Discussant: Craig R. Keenan, MD

Practice Points

  • Screen annually for type 2 diabetes mellitus (T2DM), prediabetes, weight gain, and lipid abnormalities in all patients taking atypical antipsychotics.
  • Screen annually psychiatric patients age ≥30 who do not take atypicals for T2DM and prediabetes.
  • For patients age <30, regularly review your patients’ risk factors for diabetes to determine whom to screen for T2DM or prediabetes.
  • Screening is done most simply by ordering a fasting plasma glucose test.

Psychiatric patients—especially those with schizophrenia or taking atypical antipsychotics—are at risk for developing type 2 diabetes mellitus (T2DM) and prediabetes conditions. T2DM can be present for years without significant symptoms and even asymptomatic conditions increase the risk of cardiovascular, renal, retinal, and neurologic complications.

Despite a need for T2DM screening and treatment, expert guidelines disagree on who and how to screen (Table 1). Although testing patients who have diabetes symptoms—including polyuria, polydipsia, and weight loss—is indicated, some medical groups advocate screening asymptomatic persons for T2DM.

Screening recommendations

Consensus guidelines. In 2004, the American Diabetes Association (ADA), American Psychiatric Association (APA), American Association of Clinical Endocrinologists (AACE), and North American Association for the Study of Obesity (NAASO) created consensus guidelines for screening psychiatric patients receiving atypical antipsychotics. In addition to diabetes risk, psychiatric patients are at higher risk for metabolic syndrome, dyslipidemia, obesity, and hypertension.1 The ADA, APA, AACE, and NAASO recommend regularly screening for weight gain and dyslipidemia, obtaining baseline values of fasting plasma glucose (FPG), rechecking FPG after 3 months, and then screening annually for diabetes or prediabetes. For patients with risk factors for diabetes and those who develop diabetes or prediabetes while taking an atypical antipsychotic, consider an atypical with a lower risk of diabetes—specifically aripiprazole or ziprasidone.1 For psychiatric patients who do not take atypicals, there is no consensus on who and how to screen for T2DM.

The U.S. Preventive Services Task Force (USPSTF) recommends screening only adults with hypertension.2 Its review found insufficient evidence that early detection and treatment leads to improved clinical outcomes in asymptomatic adults.

The ADA recommends more liberal screening, including individuals age ≥45 or anyone age <45 who is overweight and has any other diabetes risk factors.3 The ADA admits that no trials show a benefit of screening asymptomatic patients but notes that the duration of glycemic burden predicts adverse outcomes and effective interventions for diabetes and prediabetes are available.

AACE guidelines recommend screening starting at age 30 if the patient has risk factors for T2DM. This is the only group that includes psychiatric illness as a risk factor.4

European Association for the Study of Diabetes (EASD) guidelines calculate a risk score based on common risk factors to determine who should be screened and recommend using the oral glucose tolerance test (OGTT) rather the FPG.5 The OGTT identifies more cases of diabetes and pre-diabetes but takes >2 hours to administer.

Table 1

General population screening recommendations for type 2 diabetes mellitus or prediabetes

OrganizationYearWhom to screenHow to screen
U.S. Preventive Services Task Force (USPSTF)2008Asymptomatic adults with sustained blood pressure >135/80 mmHg (treated or untreated)FPG or OGTT every 3 years
American Diabetes Association (ADA)2009All adults age ≥45
Adults of any age with BMI >25 kg/m2 and ≥1 risk factors for diabetes (Table 2)
FPG or 2-hour OGTT every 3 years or more frequently, depending on initial results and risks
American Association of Clinical Endocrinologists (AACE)2007All adults age ≥30 with risk factors for diabetes (Table 2)FPG or 2-hour OGTT (frequency not specified)
European Association for the Study of Diabetes (EASD) and European Society of Cardiology (ESC)2007All adults with elevated risk score*OGTT (frequency not indicated)
FPG: fasting plasma glucose; OGTT: oral glucose tolerance test (75 gm glucose load); BMI: body mass index
*Risk scoring tool available at www.diabetes.fi/english/risktest

Discussion

Despite a lack evidence showing benefit to the screened population, treating diabetes and its comorbidities improves outcomes, and the potential risks of therapy are low. Therefore, it seems reasonable to screen more patients than the USPSTF recommends.

Using the EASD risk score is intriguing, but difficult to implement in a busy practice. Therefore, I recommend following the AACE guidelines, which recognize psychiatric illness as a risk factor, for screening psychiatric patients who are not receiving atypicals.

Annually screen psychiatric patients age ≥30, especially those with schizophrenia or affective disorders. I also follow the ADA guidelines and screen overweight adults age ≤30 if they have any of the other risk factors listed in Table 2. The most common risk factors seen in practice are being a member of a high-risk ethnic group, hypertension, lipid abnormalities, and cardiovascular disease. For overweight adults without other risk factors, I start screening at age 30.

 

 

Other practitioners can be more or less conservative and still be within accepted guidelines. The FPG—glucose level drawn from a vein after at least 8 hours of fasting—is probably the easiest screening test in practice. Any patient with a value >100mg/dL should be referred to the patient’s primary care physician. Any patient who develops diabetes symptoms—including polyuria, polydipsia, and weight loss—should be tested immediately. The hemoglobin A1C test is not recommended for screening.

Table 2

Risk factors identified for diabetes or prediabetes

American Diabetes Association (ADA)
  • BMI >25 kg/m2
  • physical inactivity
  • first-degree relative with diabetes
  • members of high-risk ethnic populations (African-American, Latino, Native American, Asian, Pacific Islander)
  • women who delivered a baby >9 lb or had gestational diabetes
  • hypertension
  • high-density lipoproteins cholesterol <35 mg/dL and/or triglyceride level >250 mg/dL
  • women with polycystic ovarian syndrome
  • impaired glucose tolerance or impaired fasting glucose on previous testing
  • conditions associated with insulin resistance, such as severe obesity or acanthosis nigricans
  • history of cardiovascular disease
American Association of Clinical Endocrinologists
  • All of the risk factors identified by the ADA, except for conditions associated with insulin resistance, such as severe obesity or acanthosis nigricans
  • psychiatric illness

Clinical presentation

Screening detects overt diabetes and can identify prediabetes. Prediabetes includes conditions of impaired fasting glucose (IFG) or impaired glucose tolerance (IGT). IFG is defined as a fasting glucose of 100 to 125 mg/dL, and IGT is defined as having a 2-hour glucose of 140 to 199 mg/dL on an OGTT.

Approximately one-quarter of the adult population has prediabetes, and interventions can prevent the progression of prediabetes to overt diabetes and reverse prediabetes. The Diabetes Prevention Trial found that lifestyle measures—including exercise and diet—were most effective, with a 53% reduction in the rate of progression to diabetes.6 Metformin also was effective, but less so than lifestyle measures alone.

Treatment slows the development or progression of microvascular complications, such as retinopathy, nephropathy, and neuropathy. Aggressive treatment of comorbid conditions, including hyperlipidemia and hypertension, also reduces the risk of cardiovascular events.

Drug brand names

  • Aripiprazole • Abilify
  • Metformin • Glucophage
  • Ziprasidone • Geodon

Related resources

  • American Diabetes Association. Diabetes risk calculator. www.diabetes.org/risk-test.jsp.
  • Dagogo-Jack S. The role of antipsychotic agents in the development of diabetes mellitus. Nat Clin Pract Endocrinol Metab. 2009;5(1):22-23. Quick, up-to-date review of the association between atypical antipsychotics and diabetes mellitus.

Disclosure

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

References

1. American Diabetes Association, American Psychiatric Association, American Association of Clinical Endocrinologists, et al. Consensus development conference on antipsychotic drugs and obesity and diabetes. Diabetes Care. 2004;27(2):596-601.

2. U.S. Preventive Services Task Force. Screening for type 2 diabetes mellitus in adults: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2008;148(11):846-854.

3. American Diabetes Association. Standards of medical care in diabetes—2009. Diabetes Care. 2009;32(suppl 1):S13-61.

4. Rodbard HW, Blonde L, Braithwaite SS, et al. American Association of Clinical Endocrinologists medical guidelines for clinical practice for the management of diabetes mellitus. Endocr Pract. 2007;13(suppl 1):1-68.

5. Rydén L, Standl E, Bartnik M, et al. Guidelines on diabetes, pre-diabetes and cardiovascular diseases: executive summary. The Task Force on Diabetes and Cardio-vascular Diseases of the European Society of Cardiology (ESC) and of the European Association for the Study of Diabetes (EASD). Eur Heart J. 2007;28(1):88-136.

6. Knowler WC, Barrett-Connor E, Fowler SE, et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med. 2002;346(6):393-403.

Article PDF
Author and Disclosure Information


Robert M. McCarron, DO
Series Editor

Issue
Current Psychiatry - 08(03)
Publications
Page Number
19-23
Legacy Keywords
diabetes screening;diabetes risk factors;diabetes guidelines;antipsychotics;Craig Keenan;Robert McCarron
Sections
Author and Disclosure Information


Robert M. McCarron, DO
Series Editor

Author and Disclosure Information


Robert M. McCarron, DO
Series Editor

Article PDF
Article PDF

Dr. Keenan is associate professor, department of medicine, University of California, Davis.

Principal Source: U.S. Preventive Services Task Force. Screening for type 2 diabetes mellitus in adults: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2008;148(11):846-854.—Discussant: Craig R. Keenan, MD

Practice Points

  • Screen annually for type 2 diabetes mellitus (T2DM), prediabetes, weight gain, and lipid abnormalities in all patients taking atypical antipsychotics.
  • Screen annually psychiatric patients age ≥30 who do not take atypicals for T2DM and prediabetes.
  • For patients age <30, regularly review your patients’ risk factors for diabetes to determine whom to screen for T2DM or prediabetes.
  • Screening is done most simply by ordering a fasting plasma glucose test.

Psychiatric patients—especially those with schizophrenia or taking atypical antipsychotics—are at risk for developing type 2 diabetes mellitus (T2DM) and prediabetes conditions. T2DM can be present for years without significant symptoms and even asymptomatic conditions increase the risk of cardiovascular, renal, retinal, and neurologic complications.

Despite a need for T2DM screening and treatment, expert guidelines disagree on who and how to screen (Table 1). Although testing patients who have diabetes symptoms—including polyuria, polydipsia, and weight loss—is indicated, some medical groups advocate screening asymptomatic persons for T2DM.

Screening recommendations

Consensus guidelines. In 2004, the American Diabetes Association (ADA), American Psychiatric Association (APA), American Association of Clinical Endocrinologists (AACE), and North American Association for the Study of Obesity (NAASO) created consensus guidelines for screening psychiatric patients receiving atypical antipsychotics. In addition to diabetes risk, psychiatric patients are at higher risk for metabolic syndrome, dyslipidemia, obesity, and hypertension.1 The ADA, APA, AACE, and NAASO recommend regularly screening for weight gain and dyslipidemia, obtaining baseline values of fasting plasma glucose (FPG), rechecking FPG after 3 months, and then screening annually for diabetes or prediabetes. For patients with risk factors for diabetes and those who develop diabetes or prediabetes while taking an atypical antipsychotic, consider an atypical with a lower risk of diabetes—specifically aripiprazole or ziprasidone.1 For psychiatric patients who do not take atypicals, there is no consensus on who and how to screen for T2DM.

The U.S. Preventive Services Task Force (USPSTF) recommends screening only adults with hypertension.2 Its review found insufficient evidence that early detection and treatment leads to improved clinical outcomes in asymptomatic adults.

The ADA recommends more liberal screening, including individuals age ≥45 or anyone age <45 who is overweight and has any other diabetes risk factors.3 The ADA admits that no trials show a benefit of screening asymptomatic patients but notes that the duration of glycemic burden predicts adverse outcomes and effective interventions for diabetes and prediabetes are available.

AACE guidelines recommend screening starting at age 30 if the patient has risk factors for T2DM. This is the only group that includes psychiatric illness as a risk factor.4

European Association for the Study of Diabetes (EASD) guidelines calculate a risk score based on common risk factors to determine who should be screened and recommend using the oral glucose tolerance test (OGTT) rather the FPG.5 The OGTT identifies more cases of diabetes and pre-diabetes but takes >2 hours to administer.

Table 1

General population screening recommendations for type 2 diabetes mellitus or prediabetes

OrganizationYearWhom to screenHow to screen
U.S. Preventive Services Task Force (USPSTF)2008Asymptomatic adults with sustained blood pressure >135/80 mmHg (treated or untreated)FPG or OGTT every 3 years
American Diabetes Association (ADA)2009All adults age ≥45
Adults of any age with BMI >25 kg/m2 and ≥1 risk factors for diabetes (Table 2)
FPG or 2-hour OGTT every 3 years or more frequently, depending on initial results and risks
American Association of Clinical Endocrinologists (AACE)2007All adults age ≥30 with risk factors for diabetes (Table 2)FPG or 2-hour OGTT (frequency not specified)
European Association for the Study of Diabetes (EASD) and European Society of Cardiology (ESC)2007All adults with elevated risk score*OGTT (frequency not indicated)
FPG: fasting plasma glucose; OGTT: oral glucose tolerance test (75 gm glucose load); BMI: body mass index
*Risk scoring tool available at www.diabetes.fi/english/risktest

Discussion

Despite a lack evidence showing benefit to the screened population, treating diabetes and its comorbidities improves outcomes, and the potential risks of therapy are low. Therefore, it seems reasonable to screen more patients than the USPSTF recommends.

Using the EASD risk score is intriguing, but difficult to implement in a busy practice. Therefore, I recommend following the AACE guidelines, which recognize psychiatric illness as a risk factor, for screening psychiatric patients who are not receiving atypicals.

Annually screen psychiatric patients age ≥30, especially those with schizophrenia or affective disorders. I also follow the ADA guidelines and screen overweight adults age ≤30 if they have any of the other risk factors listed in Table 2. The most common risk factors seen in practice are being a member of a high-risk ethnic group, hypertension, lipid abnormalities, and cardiovascular disease. For overweight adults without other risk factors, I start screening at age 30.

 

 

Other practitioners can be more or less conservative and still be within accepted guidelines. The FPG—glucose level drawn from a vein after at least 8 hours of fasting—is probably the easiest screening test in practice. Any patient with a value >100mg/dL should be referred to the patient’s primary care physician. Any patient who develops diabetes symptoms—including polyuria, polydipsia, and weight loss—should be tested immediately. The hemoglobin A1C test is not recommended for screening.

Table 2

Risk factors identified for diabetes or prediabetes

American Diabetes Association (ADA)
  • BMI >25 kg/m2
  • physical inactivity
  • first-degree relative with diabetes
  • members of high-risk ethnic populations (African-American, Latino, Native American, Asian, Pacific Islander)
  • women who delivered a baby >9 lb or had gestational diabetes
  • hypertension
  • high-density lipoproteins cholesterol <35 mg/dL and/or triglyceride level >250 mg/dL
  • women with polycystic ovarian syndrome
  • impaired glucose tolerance or impaired fasting glucose on previous testing
  • conditions associated with insulin resistance, such as severe obesity or acanthosis nigricans
  • history of cardiovascular disease
American Association of Clinical Endocrinologists
  • All of the risk factors identified by the ADA, except for conditions associated with insulin resistance, such as severe obesity or acanthosis nigricans
  • psychiatric illness

Clinical presentation

Screening detects overt diabetes and can identify prediabetes. Prediabetes includes conditions of impaired fasting glucose (IFG) or impaired glucose tolerance (IGT). IFG is defined as a fasting glucose of 100 to 125 mg/dL, and IGT is defined as having a 2-hour glucose of 140 to 199 mg/dL on an OGTT.

Approximately one-quarter of the adult population has prediabetes, and interventions can prevent the progression of prediabetes to overt diabetes and reverse prediabetes. The Diabetes Prevention Trial found that lifestyle measures—including exercise and diet—were most effective, with a 53% reduction in the rate of progression to diabetes.6 Metformin also was effective, but less so than lifestyle measures alone.

Treatment slows the development or progression of microvascular complications, such as retinopathy, nephropathy, and neuropathy. Aggressive treatment of comorbid conditions, including hyperlipidemia and hypertension, also reduces the risk of cardiovascular events.

Drug brand names

  • Aripiprazole • Abilify
  • Metformin • Glucophage
  • Ziprasidone • Geodon

Related resources

  • American Diabetes Association. Diabetes risk calculator. www.diabetes.org/risk-test.jsp.
  • Dagogo-Jack S. The role of antipsychotic agents in the development of diabetes mellitus. Nat Clin Pract Endocrinol Metab. 2009;5(1):22-23. Quick, up-to-date review of the association between atypical antipsychotics and diabetes mellitus.

Disclosure

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

Dr. Keenan is associate professor, department of medicine, University of California, Davis.

Principal Source: U.S. Preventive Services Task Force. Screening for type 2 diabetes mellitus in adults: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2008;148(11):846-854.—Discussant: Craig R. Keenan, MD

Practice Points

  • Screen annually for type 2 diabetes mellitus (T2DM), prediabetes, weight gain, and lipid abnormalities in all patients taking atypical antipsychotics.
  • Screen annually psychiatric patients age ≥30 who do not take atypicals for T2DM and prediabetes.
  • For patients age <30, regularly review your patients’ risk factors for diabetes to determine whom to screen for T2DM or prediabetes.
  • Screening is done most simply by ordering a fasting plasma glucose test.

Psychiatric patients—especially those with schizophrenia or taking atypical antipsychotics—are at risk for developing type 2 diabetes mellitus (T2DM) and prediabetes conditions. T2DM can be present for years without significant symptoms and even asymptomatic conditions increase the risk of cardiovascular, renal, retinal, and neurologic complications.

Despite a need for T2DM screening and treatment, expert guidelines disagree on who and how to screen (Table 1). Although testing patients who have diabetes symptoms—including polyuria, polydipsia, and weight loss—is indicated, some medical groups advocate screening asymptomatic persons for T2DM.

Screening recommendations

Consensus guidelines. In 2004, the American Diabetes Association (ADA), American Psychiatric Association (APA), American Association of Clinical Endocrinologists (AACE), and North American Association for the Study of Obesity (NAASO) created consensus guidelines for screening psychiatric patients receiving atypical antipsychotics. In addition to diabetes risk, psychiatric patients are at higher risk for metabolic syndrome, dyslipidemia, obesity, and hypertension.1 The ADA, APA, AACE, and NAASO recommend regularly screening for weight gain and dyslipidemia, obtaining baseline values of fasting plasma glucose (FPG), rechecking FPG after 3 months, and then screening annually for diabetes or prediabetes. For patients with risk factors for diabetes and those who develop diabetes or prediabetes while taking an atypical antipsychotic, consider an atypical with a lower risk of diabetes—specifically aripiprazole or ziprasidone.1 For psychiatric patients who do not take atypicals, there is no consensus on who and how to screen for T2DM.

The U.S. Preventive Services Task Force (USPSTF) recommends screening only adults with hypertension.2 Its review found insufficient evidence that early detection and treatment leads to improved clinical outcomes in asymptomatic adults.

The ADA recommends more liberal screening, including individuals age ≥45 or anyone age <45 who is overweight and has any other diabetes risk factors.3 The ADA admits that no trials show a benefit of screening asymptomatic patients but notes that the duration of glycemic burden predicts adverse outcomes and effective interventions for diabetes and prediabetes are available.

AACE guidelines recommend screening starting at age 30 if the patient has risk factors for T2DM. This is the only group that includes psychiatric illness as a risk factor.4

European Association for the Study of Diabetes (EASD) guidelines calculate a risk score based on common risk factors to determine who should be screened and recommend using the oral glucose tolerance test (OGTT) rather the FPG.5 The OGTT identifies more cases of diabetes and pre-diabetes but takes >2 hours to administer.

Table 1

General population screening recommendations for type 2 diabetes mellitus or prediabetes

OrganizationYearWhom to screenHow to screen
U.S. Preventive Services Task Force (USPSTF)2008Asymptomatic adults with sustained blood pressure >135/80 mmHg (treated or untreated)FPG or OGTT every 3 years
American Diabetes Association (ADA)2009All adults age ≥45
Adults of any age with BMI >25 kg/m2 and ≥1 risk factors for diabetes (Table 2)
FPG or 2-hour OGTT every 3 years or more frequently, depending on initial results and risks
American Association of Clinical Endocrinologists (AACE)2007All adults age ≥30 with risk factors for diabetes (Table 2)FPG or 2-hour OGTT (frequency not specified)
European Association for the Study of Diabetes (EASD) and European Society of Cardiology (ESC)2007All adults with elevated risk score*OGTT (frequency not indicated)
FPG: fasting plasma glucose; OGTT: oral glucose tolerance test (75 gm glucose load); BMI: body mass index
*Risk scoring tool available at www.diabetes.fi/english/risktest

Discussion

Despite a lack evidence showing benefit to the screened population, treating diabetes and its comorbidities improves outcomes, and the potential risks of therapy are low. Therefore, it seems reasonable to screen more patients than the USPSTF recommends.

Using the EASD risk score is intriguing, but difficult to implement in a busy practice. Therefore, I recommend following the AACE guidelines, which recognize psychiatric illness as a risk factor, for screening psychiatric patients who are not receiving atypicals.

Annually screen psychiatric patients age ≥30, especially those with schizophrenia or affective disorders. I also follow the ADA guidelines and screen overweight adults age ≤30 if they have any of the other risk factors listed in Table 2. The most common risk factors seen in practice are being a member of a high-risk ethnic group, hypertension, lipid abnormalities, and cardiovascular disease. For overweight adults without other risk factors, I start screening at age 30.

 

 

Other practitioners can be more or less conservative and still be within accepted guidelines. The FPG—glucose level drawn from a vein after at least 8 hours of fasting—is probably the easiest screening test in practice. Any patient with a value >100mg/dL should be referred to the patient’s primary care physician. Any patient who develops diabetes symptoms—including polyuria, polydipsia, and weight loss—should be tested immediately. The hemoglobin A1C test is not recommended for screening.

Table 2

Risk factors identified for diabetes or prediabetes

American Diabetes Association (ADA)
  • BMI >25 kg/m2
  • physical inactivity
  • first-degree relative with diabetes
  • members of high-risk ethnic populations (African-American, Latino, Native American, Asian, Pacific Islander)
  • women who delivered a baby >9 lb or had gestational diabetes
  • hypertension
  • high-density lipoproteins cholesterol <35 mg/dL and/or triglyceride level >250 mg/dL
  • women with polycystic ovarian syndrome
  • impaired glucose tolerance or impaired fasting glucose on previous testing
  • conditions associated with insulin resistance, such as severe obesity or acanthosis nigricans
  • history of cardiovascular disease
American Association of Clinical Endocrinologists
  • All of the risk factors identified by the ADA, except for conditions associated with insulin resistance, such as severe obesity or acanthosis nigricans
  • psychiatric illness

Clinical presentation

Screening detects overt diabetes and can identify prediabetes. Prediabetes includes conditions of impaired fasting glucose (IFG) or impaired glucose tolerance (IGT). IFG is defined as a fasting glucose of 100 to 125 mg/dL, and IGT is defined as having a 2-hour glucose of 140 to 199 mg/dL on an OGTT.

Approximately one-quarter of the adult population has prediabetes, and interventions can prevent the progression of prediabetes to overt diabetes and reverse prediabetes. The Diabetes Prevention Trial found that lifestyle measures—including exercise and diet—were most effective, with a 53% reduction in the rate of progression to diabetes.6 Metformin also was effective, but less so than lifestyle measures alone.

Treatment slows the development or progression of microvascular complications, such as retinopathy, nephropathy, and neuropathy. Aggressive treatment of comorbid conditions, including hyperlipidemia and hypertension, also reduces the risk of cardiovascular events.

Drug brand names

  • Aripiprazole • Abilify
  • Metformin • Glucophage
  • Ziprasidone • Geodon

Related resources

  • American Diabetes Association. Diabetes risk calculator. www.diabetes.org/risk-test.jsp.
  • Dagogo-Jack S. The role of antipsychotic agents in the development of diabetes mellitus. Nat Clin Pract Endocrinol Metab. 2009;5(1):22-23. Quick, up-to-date review of the association between atypical antipsychotics and diabetes mellitus.

Disclosure

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

References

1. American Diabetes Association, American Psychiatric Association, American Association of Clinical Endocrinologists, et al. Consensus development conference on antipsychotic drugs and obesity and diabetes. Diabetes Care. 2004;27(2):596-601.

2. U.S. Preventive Services Task Force. Screening for type 2 diabetes mellitus in adults: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2008;148(11):846-854.

3. American Diabetes Association. Standards of medical care in diabetes—2009. Diabetes Care. 2009;32(suppl 1):S13-61.

4. Rodbard HW, Blonde L, Braithwaite SS, et al. American Association of Clinical Endocrinologists medical guidelines for clinical practice for the management of diabetes mellitus. Endocr Pract. 2007;13(suppl 1):1-68.

5. Rydén L, Standl E, Bartnik M, et al. Guidelines on diabetes, pre-diabetes and cardiovascular diseases: executive summary. The Task Force on Diabetes and Cardio-vascular Diseases of the European Society of Cardiology (ESC) and of the European Association for the Study of Diabetes (EASD). Eur Heart J. 2007;28(1):88-136.

6. Knowler WC, Barrett-Connor E, Fowler SE, et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med. 2002;346(6):393-403.

References

1. American Diabetes Association, American Psychiatric Association, American Association of Clinical Endocrinologists, et al. Consensus development conference on antipsychotic drugs and obesity and diabetes. Diabetes Care. 2004;27(2):596-601.

2. U.S. Preventive Services Task Force. Screening for type 2 diabetes mellitus in adults: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2008;148(11):846-854.

3. American Diabetes Association. Standards of medical care in diabetes—2009. Diabetes Care. 2009;32(suppl 1):S13-61.

4. Rodbard HW, Blonde L, Braithwaite SS, et al. American Association of Clinical Endocrinologists medical guidelines for clinical practice for the management of diabetes mellitus. Endocr Pract. 2007;13(suppl 1):1-68.

5. Rydén L, Standl E, Bartnik M, et al. Guidelines on diabetes, pre-diabetes and cardiovascular diseases: executive summary. The Task Force on Diabetes and Cardio-vascular Diseases of the European Society of Cardiology (ESC) and of the European Association for the Study of Diabetes (EASD). Eur Heart J. 2007;28(1):88-136.

6. Knowler WC, Barrett-Connor E, Fowler SE, et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med. 2002;346(6):393-403.

Issue
Current Psychiatry - 08(03)
Issue
Current Psychiatry - 08(03)
Page Number
19-23
Page Number
19-23
Publications
Publications
Article Type
Display Headline
Diabetes screening: Which patients, what tests, and how often?
Display Headline
Diabetes screening: Which patients, what tests, and how often?
Legacy Keywords
diabetes screening;diabetes risk factors;diabetes guidelines;antipsychotics;Craig Keenan;Robert McCarron
Legacy Keywords
diabetes screening;diabetes risk factors;diabetes guidelines;antipsychotics;Craig Keenan;Robert McCarron
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

‘Night owls’: Reset the physiologic clock in delayed sleep phase disorder

Article Type
Changed
Tue, 12/11/2018 - 15:06
Display Headline
‘Night owls’: Reset the physiologic clock in delayed sleep phase disorder

Comment on this article

Jason, age 16, has had difficulty with sleep initiation for 2 years. He describes going to bed at 10:30 PM on school nights but falling asleep no sooner than midnight and typically after 1:30 AM. He denies contributions from an “active mind” or environmental disturbances, and his bedroom contains no TV, computer, or other media devices. He does not sleep better with a change in environment. He denies pervasive low mood symptoms and believes his mood hinges predominantly on his ability to achieve sufficient sleep.

Once asleep, Jason generally enjoys good sleep consolidation until he needs to arise at 6:30 AM. His mother awakens him with difficulty, as he often sleeps through his alarm. He sleeps approximately 5 hours nightly during the school week, endorses impaired concentration, and often dozes during his first several classes. When he returns home from school, he finds it very difficult to resist napping.

On weekends he retires at 1 AM or later and typically falls asleep within 30 minutes. He usually awakens at noon but can sleep as late as 4:30 PM. He feels slightly more refreshed on weekends and describes his mood then as improved. During a recent spring break, he felt much better when allowed to sleep as much as he wanted.

Delayed sleep phase disorder (DSPD)—characterized by a pathological “night owl” circadian preference—is seen most commonly in adolescents and is associated with psychiatric morbidity, psychosocial impairment, and poor academic performance. Proper identification of the condition can be enhanced with a variety of assessment tools, and successful treatment requires an awareness of potential endogenous and exogenous contributors.

This article describes what is known about DSPD and uses the case example to illustrate diagnostic assessment and treatment choices. Intriguing data support various pathophysiologic explanations for DSPD (Box 1).1-6 Facilitating the adjustment of patients’ physiologic clocks is the overall goal in managing DSPD.

Box 1

What causes delayed sleep phase disorder?

In individuals normally entrained to the light/dark cycle, circadian rhythms are:

  • delayed by evening exposure to bright light (≥ 2,500 lux) prior to the core body temperature minimum (Tmin)
  • advanced by morning light exposure after the Tmin.1

These opposing effects attune most people to the light/dark cycle, with sleep and wakefulness occurring on a conventional schedule. Persons with delayed sleep phase disorder (DSPD) live at a delayed phase that resists advancement and is incompatible with their personal and social obligations.

Theories have been proposed, but DSPD’s etiology has not been fully explained. Affected adolescents may exhibit an extreme in circadian preference. Case reports also describe DSPD emerging after traumatic brain injury.2

Intriguing evidence supports various pathophysiologic explanations for DSPD. An abnormally long intrinsic circadian period (>25 hours) was recently demonstrated during temporal isolation in 1 individual with DSPD.3 Both this case report and controlled studies describe deviations from expected relationships between the sleep/wake cycle and physiologic circadian markers. Most consistently described are longer intervals from Tmin4 to sleep offset (final rise time) in DSPD patients compared with controls.

Other research suggests:

  • hypersensitivity to nocturnal photic stimulation in select DSPD patients5
  • impaired homeostatic sleep processes, as DSPD patients show a diminished ability to initiate sleep after sleep restriction, compared with controls.6

Extreme ‘eveningness’

Because of their extreme seemingly innate preference to retire and arise at relatively late clock hours (an “eveningness” trait), school-aged patients with DSPD represent a high-risk population for problematic sleepiness. In a survey of 612 high school students, the 63% who felt they needed more sleep on school nights showed a strong eveningness preference (as assessed by questionnaire), compared with students who described getting sufficient sleep.7 Other studies have revealed psychiatric morbidity (including affective and personality disorders), psychosocial impairment, and poor academic performance associated with the condition.8-10

DSPD may affect 7% to 16% of patients presenting with insomnia complaints in sleep medicine clinics.11 The condition appears most common among young cohorts and has been reported to affect up to 7% of adolescents in the United States.12 Its high frequency in this age group may be a pathologic exaggeration of the normal tendency toward delayed timing of sleep and wakefulness linked with pubertal development.13

Sleep and wakefulness regulation

Conceptually, 2 processes govern sleep and wakefulness:

  • The homeostatic drive to sleep (process S) is proportional to the duration of sleep restriction and becomes maximal at about 40 hours.
  • Circadian regulation (process C) creates a drive for wakefulness that variably opposes process S and depends upon intrinsic rhythms.14

Neurons of the suprachiasmatic nucleus in the hypothalamus exert master coordination of this sleep/wake rhythm, along with other behavioral and physiologic variables.15 Because the typical intrinsic period is slightly longer than 24 hours, synchronization to the 24-hour day (entrainment) is accomplished by environmental inputs (zeitgebers, or “time givers”), the most important of which is exposure to light.16

 

 

Misalignment between endogenous circadian rhythms and the light/dark cycle can result in circadian rhythm sleep disorders, such as:

  • delayed sleep timing (DSPD)
  • advanced sleep timing (advanced sleep phase disorder)
  • erratic sleep timing (irregular sleep/wake rhythm)
  • complete dissociation from the light/dark cycle (circadian rhythm sleep disorder, free-running type).

These 4 conditions are thought to involve predominantly intrinsic mechanisms, but circadian dysrhythmias also can be induced by exogenous factors. Extreme work schedules or rapid travel across time zones can challenge the circadian system’s ability to acclimate and the individual’s ability to achieve a desired sleep schedule.17

Differential diagnosis

Because DSPD relates primarily to an aberration in timing of sleep, it is characterized as a disorder only if the individual’s preferred schedule interferes substantially with social or occupational functioning. The International Classification of Sleep Disorders (ICSD) provides detailed diagnostic criteria (Table).17

Table

Diagnostic criteria for delayed sleep phase disorder

A. Delay exists in the phase of the major sleep period in relation to desired sleep time and wake-up time, as evidenced by:
  • a chronic or recurrent complaint of inability to fall asleep at a desired conventional clock time AND
  • inability to awaken at a desired and socially acceptable time.
B. When allowed to choose a preferred schedule, patients exhibit normal sleep quality and duration for age and maintain a delayed but stable phase of entrainment to the 24-hour sleep/wake pattern.
C. Monitoring with a sleep log or actigraphy (including sleep diary) for at least 7 days demonstrates a stable delay in the timing of the habitual sleep period.
D. The sleep disturbance is not better explained by another sleep disorder, medical or neurologic disorder, mental disorder, medication use, or substance use disorder.
Source: Adapted and reprinted with permission from International classification of sleep disorders. Diagnostic and coding manual. 2nd ed17

Depression and anxiety often manifest with sleep difficulties, as do inadequate sleep hygiene and other conditions associated with prolonged sleep initiation. According to ICSD criteria, primary insomnia can be differentiated from DSPD if the patient readily initiates and maintains sleep when allowed to sleep on his/her desired sleep/wake schedule. Accumulated evidence has largely debunked this notion, however, as polysomnographic studies have demonstrated both prolonged sleep latency and impaired sleep efficiency in DSPD patients versus matched controls.3

Assessment tools can complement the clinical history in diagnosing DSPD. Either a sleep log or actigraphy is required to demonstrate a stable phase delay, but actigraphy typically generates more reliable data.18 Actigraphs are compact “motion detectors” whose output while being worn by patients allows longitudinal assessment of sleep/wake parameters.

Eveningness tendencies of presumptive DSPD patients can be further verified with the Morningness-Eveningness Questionnaire (MEQ) (Box 2).19 Low scores are associated with evening types—felt to correspond to the endogenous circadian period—and can help narrow the differential diagnosis of sleep-initiation complaints.20

Box 2

A morning or evening person? A self-assessment questionnaire

The Morningness-Eveningness Questionnaire (MEQ) developed by Horne and Ostberg19 can be used to verify eveningness tendencies of patients with presumptive delayed sleep phase disorder. The MEQ is a 19-item self-assessment tool with responses that are assigned values totaling up to 86 points. Examples of the questions include:

  • Considering only your own ‘feeling best’ rhythm, at what time would you get up if you were entirely free to plan your day?
  • Considering only your own ‘feeling best’ rhythm, at what time would you go to bed if you were entirely free to plan your day?
  • How easy do you find it to get up each day?
  • When you have no commitments the next day, how much later do you go to bed compared to your usual bedtime?
  • One hears about ‘morning’ and ‘evening’ types of people. Which ONE of these types do you consider yourself to be?

Lower scores are associated with evening types—felt to correspond to the endogenous circadian period—and can help in narrowing the differential diagnosis of sleep-initiation complaints.20 Scores on the MEQ are interpreted as:

  • 70 to 86: definite morning type
  • 59 to 69: moderately morning type
  • 42 to 58: neither type
  • 31 to 41: moderately evening type
  • 16 to 30: definite evening type

CASE CONTINUED: ‘Definite evening type’

Jason scores 28 on the MEQ, consistent with a “definite evening type.” Actigraphic monitoring is scheduled during a school holiday, when he is instructed to sleep according to his preferred schedule with the least possible restriction.

A clearly delayed sleep phase is evident, with the habitual sleep period occurring between 5 AM and 1 PM. Even on days when he was quite sleep-restricted because of an enforced wake time, sleep onset on the ensuing evening was substantially delayed, suggesting an obligate nature for the delayed sleep/wake schedule. Overall, Jason had few complaints with respect to impaired alertness while on this unrestricted schedule and experienced a much more stable mood.

 

 

Interventions

Without physiologic assessments, understanding the patient’s “natural” sleep schedule can allow for rational recommendations about using phototherapy and oral melatonin (Figure21). However, referral to a sleep specialist is required unless the general psychiatrist has experience in treating circadian rhythm sleep disorders.

Morning phototherapy. Properly timed morning bright light therapy (≥2,500 lux) has been shown to help DSPD patients achieve physiologically measured sleep phase advances, objective improvements in daytime alertness, and earlier reported bedtimes compared with controls.22 Unfortunately, the described 2-hour treatment duration make this research protocol clinically impractical, and most clinicians commence with a 30-minute duration of therapy, as described in the seasonal affective disorder literature.

Relatively new and widely available blue light boxes have been reported to exhibit at least equivalent efficacy to bright light devices (as reported in the literature pertaining to seasonal affective disorder), but with markedly decreased light intensity and fewer associated adverse effects.23 As the research addressing their use in the treatment of circadian rhythm sleep disorders is still emerging, their future role remains uncertain.

Precautions. Most psychiatrists would not perform a physiologic determination of a patient’s circadian phase, and further undesired phase delays can occur if phototherapy is administered before the core body temperature minimum (Tmin).24 Also, use caution if prescribing phototherapy to patients taking photosensitizing drugs and/or those with ocular or retinal pathology.20

Evening light avoidance. Whether or not you prescribe morning phototherapy, recommending that DSPD patients avoid evening light is essential to avoid further induction or exacerbation of phase delays. Protective eyewear is warranted in instances where these advisory precautions are insufficient (see Related Resources). Such an intervention has been shown effective in decreasing light exposure and undesired phase advances in studies involving subjects exposed to simulated shift work.25

Oral melatonin. Abundant evidence supports melatonin use in achieving phase advances in individuals with DSPD.26,27 A synergistic effect can be obtained when melatonin is combined with phototherapy.28

Proper timing of melatonin to achieve a maximal phase advance can be estimated based on the individual’s dim light melatonin onset (DLMO), which occurs approximately 14 hours after the habitual (unrestricted) wake time.29 Maximal phase advances appear to occur when melatonin is given approximately 6 hours before the DLMO.26 Thus, a rational practice is to recommend that patients take melatonin 8 hours after their natural wake time. Doses of ≤0.5 mg appear to achieve the maximal chronobiotic effect while avoiding an undesired hypnotic effect.30

Precautions. Verifying the purity of over-the-counter melatonin is difficult. A review by the National Academy of Sciences states that short-term use of melatonin, ≤10 mg/d, appears to be safe in healthy adults but recommends caution in children/adolescents and women of reproductive age. Doses recommended for circadian-based interventions are typically physiologic in nature (i.e., ≤0.5 mg), which may serve to mitigate these concerns.

Adverse effects such as headaches, somnolence, hypotension, hypertension, gastrointestinal upset, and exacerbation of alopecia areata have been reported at higher melatonin doses in healthy adults and at lower doses in persons with preexisting central nervous system, cardiovascular, gastrointestinal, or dermatologic conditions.31


Figure Light and melatonin phase response curves: Normal vs. delayed

This schematic compares ‘normal sleep’ phase response curves (PRCs) to light and exogenous melatonin with postulated PRCs for an individual with delayed sleep phase disorder (DSPD), presumed to be 5 hours ‘out of phase.’ Y-axis shows the direction and relative magnitude of phase shifts produced by light or melatonin at times shown on the x-axis. X-axis covers >24 hours to better illustrate the PRCs.
Relationships between ‘normal sleepers’ and DSPD patients are depicted by:

  • rectangles (sleep period)
  • triangles (core body temperature minimum [Tmin])
  • arrows (dim light melatonin onsets [DLMOs]).

‘Normal’ sleep is shown to occur from midnight to 8 AM, and the DSPD patient’s sleep from 5 AM to 1 PM; DLMO and Tmin are similarly delayed by 5 hours in the DSPD patient. This schematic assumes that phase relationships are maintained in DSPD patients, which is not a certainty.
Source: Adapted from reference 21

CASE CONTINUED: Under the bright lights

Jason starts phototherapy treatment during his winter break, administering bright light daily upon natural awakening using a 10,000 lux light box for at least 30 minutes. As instructed, he gradually advances the time of administration by approximately 30 minutes every other day, striving for a nocturnal sleep period of 11 PM to 7 AM. He also wears protective eyewear to reduce light exposure during evening hours to avoid further delays in sleep phase. To further promote a phase advance, he takes oral melatonin, 0.5 mg/d at approximately 8 PM, as determined by his self-report and results of actigraphic recording.

 

 

Other options

Hypnotics. Little evidence supports the use of hypnotics in DSPD,32 and patients may show resistance to these drugs.33 Nevertheless, hypnotics can heighten confidence in the ability to initiate sleep in individuals with a concomitant conditioned insomnia.

With chronotherapy, patients are prescribed a sleep schedule that is delayed several hours incrementally until sleep is aligned to a target bedtime. The individual then is advised to rigorously maintain a regular sleep/wake schedule, repeating the process as necessary.

Although case reports have shown positive results with chronotherapy for DSPD,34 no controlled trials have demonstrated its efficacy or safety. One study reported high relapse rates,31 and 1 patient with DSPD developed free-running circadian rhythms.35 Clinical experience suggests chronotherapy is impractical for patients who must adhere to a fixed schedule.

Behavioral approaches

For an adolescent with DSPD, consider asking the school district to allow him or her a later school start-time. This alone often can substantially increase total sleep time and mitigate associated impairments.36 In all instances pursue and address external contributors to DSPD, such as poor sleep hygiene (including excessive caffeine use) and substance misuse.

Emphasize regular wake times, as arising later on weekends can cause phase delays.37 DSPD patients may have a concomitant conditioned insomnia that responds to evidence-based behavioral treatments.38

Whatever intervention you choose, schedule a follow-up appointment in approximately 2 months to evaluate patients’ progress and compliance. Encourage them to contact you with questions or concerns in the interim. Review sleep logs or actigraphy during this visit, and adjust the timing and/or nature of interventions as needed. Adolescents can be particularly noncompliant with clinical interventions, and therapeutic goals cannot be reached without their full investment.

Because no guidelines exist on how long to treat DSPD, stop on a “trial-and-error” basis when symptoms are controlled, and resume if they recur. Another approach is to maintain a desired sleep/wake schedule with bedtime melatonin and encourage continued adherence to other measures.

CASE CONTINUED: Maintenance therapy

Jason returns to the clinic approximately 10 weeks later. After an obviously concerted effort to adhere to treatment, his progress is quite remarkable. He rarely falls asleep later than 11 PM, so he is obtaining 2.5 hours more sleep each night before arising for school at 6:30 AM. Sleepiness at school is rarely problematic, and his mood is more stable.

He nevertheless describes a persistent tendency to retire and arise later and asks to continue melatonin and phototherapy. Because no guidelines exist for long-term therapy of DSPD, he is advised to switch melatonin to bedtime dosing (with a presumed phase-neutral “maintenance” effect), and to continue phototherapy as prescribed.

Related resources

  • Wyatt JK. Delayed sleep phase syndrome: pathophysiology and treatment options. Sleep. 2004;27(6):1195-1203.
  • Crowley SJ, Acebo C, Carskadon MA. Sleep, circadian rhythms, and delayed sleep phase in adolescence. Sleep Med. 2007;8(6):602-612.
  • National Sleep Foundation. Adolescent sleep needs and patterns: research report and resource guide. Washington, DC; 2000:1-30.
  • Products designed to assist in the avoidance of light at improper times. www.lowbluelights.com.

Disclosure

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

References

1. Khalsa SB, Jewett ME, Cajochen C, et al. A phase response curve to single bright light pulses in human subjects. J Physiol. 2003;549(Pt 3):945-952.

2. Quinto C, Gellido C, Chokroverty S, et al. Posttraumatic delayed sleep phase syndrome. Neurology. 2000;54(1):250-252.

3. Campbell SS, Murphy PJ. Delayed sleep phase disorder in temporal isolation. Sleep. 2007;30(9):1225-1228.

4. Uchiyama M, Okawa M, Shibui K, et al. Altered phase relation between sleep timing and core body temperature rhythm in delayed sleep phase syndrome and non-24-hour sleep-wake syndrome in humans. Neurosci Lett. 2000;294(2):101-104.

5. Aoki H, Ozeki Y, Yamada N. Hypersensitivity of melatonin suppression in response to light in patients with delayed sleep phase syndrome. Chronobiol Int. 2001;18(2):263-271.

6. Uchiyama M, Okawa M, Shibui K, et al. Poor compensatory function for sleep loss as a pathogenic factor in patients with delayed sleep phase syndrome. Sleep. 2000;23(4):553-558.

7. Mercer PW, Merritt SL, Cowell JM. Differences in reported sleep need among adolescents. J Adolesc Health. 1998;23(5):259-263.

8. Krahn LE, Pankratz VS, Harris AM, et al. Long-term outcome of adolescents with delayed sleep phase disorder [abstract]. Sleep. 2003;26:A115.-

9. Dagan Y, Stein D, Steinbock M, et al. Frequency of delayed sleep phase syndrome among hospitalized adolescent psychiatric patients. J Psychosom Res. 1998;45(1):15-20.

10. Thorpy MJ, Korman E, Spielman AJ, et al. Delayed sleep phase syndrome in adolescents. J Adolesc Health Care. 1998;9(1):22-27.

11. Regestein QR, Monk TH. Delayed sleep phase syndrome: a review of the clinical aspects. Am J Psychiatry. 1995;152(4):602-608.

12. Pelayo RP, Thorpy MJ, Glovinsky P. Prevalence of delayed sleep phase syndrome among adolescents [abstract]. Sleep Res. 1988;17:391.-

13. Gau SF, Soong WT. The transition of sleep-wake patterns in early adolescence. Sleep. 2003;26(4):449-454.

14. Beersma DG, Gordijn MC. Circadian control of the sleep-wake cycle. Physiol Behav. 2007;90(2-3):190-195.

15. Ralph MR, Foster RG, Davis FC, et al. Transplanted suprachiasmatic nucleus determines circadian period. Science. 1990;247(4945):975-978.

16. Waterhouse J, DeCoursey PJ. Chronobiology: biological timekeeping. Sunderland, MA: Sinauer Associates, Inc. Publishers; 2004:291-323.

17. American Academy of Sleep Medicine. International classification of sleep disorders. Diagnostic and coding manual. 2nd ed. Westchester, IL: American Academy of Sleep Medicine; 2005.

18. Bradshaw DA, Yanagi MA, Pak ES, et al. Nightly sleep duration in the 2-week period preceding multiple sleep latency testing. J Clin Sleep Med. 2007;3(6):613-619.

19. Horne JA, Ostberg O. A self-assessment questionnaire to determine morningness-eveningness in human circadian rhythms. Int J Chronobiol. 1976;4(2):97-110.

20. Sack RL, Auckley D, Auger RR, et al. Circadian rhythm sleep disorders: Part I. basic principles, shift work and jet lag disorders. Sleep. 2007;30(11):1460-1483.

21. Burgess HJ, Sharkey KM, Eastman CI. Bright light, dark and melatonin can promote circadian adaptation in night shift workers. Sleep Med Rev. 2002;6(5):407-420.

22. Rosenthal NE, Joseph-Vanderpool JR, Levendosky AA, et al. Phase-shifting effects of bright morning light as treatment for delayed sleep phase syndrome. Sleep. 1990;13(4):354-361.

23. Glickman G, Byrne B, Pineda C, et al. Light therapy for seasonal affective disorder with blue narrow-band light-emitting diodes (LEDs). Biol Psychiatry. 2006;59:502-507.

24. Czeisler C, Wright K, Jr. Influence of light on circadian rhythmicity in humans. New York, NY: Marcel Dekker; 1999:149-180.

25. Crowley SJ, Lee C, Tseng CY, et al. Combinations of bright light, scheduled dark, sunglasses, and melatonin to facilitate circadian entrainment to night shift work. J Biol Rhythms. 2003;18(6):513-523.

26. Mundey K, Benloucif S, Harsanyi K, et al. Phase-dependent treatment of delayed sleep phase syndrome with melatonin. Sleep. 2005;28(10):1271-1278.

27. Sack RL, Auckley D, Auger RR, et al. Circadian rhythm sleep disorders: Part II, advanced sleep phase disorder, delayed sleep phase disorder, free-running disorder, and irregular sleep-wake rhythm. Sleep. 2007;30(11):1484-1501.

28. Revell VL, Burgess HJ, Gazda CJ, et al. Advancing human circadian rhythms with afternoon melatonin and morning intermittent bright light. J Clin Endocrinol Metab. 2006;91(1):54-59.

29. Burgess HJ, Eastman CI. The dim light melatonin onset following fixed and free sleep schedules. J Sleep Res. 2005;14(3):229-237.

30. Lewy AJ. Clinical applications of melatonin in circadian disorders. Dialog Clin Neurosci. 2003;5:399-413.

31. Committee on the Framework for Evaluating the Safety of Dietary Supplements FaNB, Board on Life Sciences, Institute of Medicine and National Research Council of the National Academies. Dietary supplements: a framework for evaluating safety. Washington, DC: The National Academies Press; 2005.

32. Ito A, Ando K, Hayakawa T, et al. Long-term course of adult patients with delayed sleep phase syndrome. Jpn J Psychiatry Neurol. 1993;47(3):563-567.

33. Auger RR. Circadian rhythm sleep disorder, delayed sleep phase type (pediatric case). In: Winkelman JW (chair), Henderson JH, Kotagal S, et al, eds. Case book of sleep medicine. Westchester, IL: American Academy of Sleep Medicine; 2008:195-199.

34. Czeisler C, Weitzman E, Moore, et al. Chronotherapy: resetting the circadian clocks of patients with delayed sleep phase insomnia. Sleep. 1981;4:1-21.

35. Oren DA, Wehr TA. Hypernyctohemeral syndrome after chronotherapy for delayed sleep phase syndrome. N Engl J Med. 1992;327(24):1762.-

36. Wahlstrom K. Changing times: findings from the first longitudinal study of later high school start times. NASSP Bulletin. 2002;86(633):3-21.

37. Burgess HJ, Eastman CI. A late wake time phase delays the human dim light melatonin rhythm. Neurosci Lett. 2006;395(3):191-195.

38. Morgenthaler T, Kramer M, Alessi C, et al. Practice parameters for the psychological and behavioral treatment of insomnia: an update. An American Academy of Sleep Medicine report. Sleep. 2006;29(11):1415-1419.

Article PDF
Author and Disclosure Information

Robert R. Auger, MD
Assistant professor of psychiatry and medicine, Mayo Clinic College of Medicine Consultant, Mayo Center for Sleep Medicine, Rochester, MN

Issue
Current Psychiatry - 08(03)
Publications
Topics
Page Number
55-65
Legacy Keywords
delayed sleep phase disorder;delayed circadian cycle;insomnia;circadian rhythm;circadian preference;phototherapy,light avoidance;actigraphy;R. Robert Auger
Sections
Author and Disclosure Information

Robert R. Auger, MD
Assistant professor of psychiatry and medicine, Mayo Clinic College of Medicine Consultant, Mayo Center for Sleep Medicine, Rochester, MN

Author and Disclosure Information

Robert R. Auger, MD
Assistant professor of psychiatry and medicine, Mayo Clinic College of Medicine Consultant, Mayo Center for Sleep Medicine, Rochester, MN

Article PDF
Article PDF

Comment on this article

Jason, age 16, has had difficulty with sleep initiation for 2 years. He describes going to bed at 10:30 PM on school nights but falling asleep no sooner than midnight and typically after 1:30 AM. He denies contributions from an “active mind” or environmental disturbances, and his bedroom contains no TV, computer, or other media devices. He does not sleep better with a change in environment. He denies pervasive low mood symptoms and believes his mood hinges predominantly on his ability to achieve sufficient sleep.

Once asleep, Jason generally enjoys good sleep consolidation until he needs to arise at 6:30 AM. His mother awakens him with difficulty, as he often sleeps through his alarm. He sleeps approximately 5 hours nightly during the school week, endorses impaired concentration, and often dozes during his first several classes. When he returns home from school, he finds it very difficult to resist napping.

On weekends he retires at 1 AM or later and typically falls asleep within 30 minutes. He usually awakens at noon but can sleep as late as 4:30 PM. He feels slightly more refreshed on weekends and describes his mood then as improved. During a recent spring break, he felt much better when allowed to sleep as much as he wanted.

Delayed sleep phase disorder (DSPD)—characterized by a pathological “night owl” circadian preference—is seen most commonly in adolescents and is associated with psychiatric morbidity, psychosocial impairment, and poor academic performance. Proper identification of the condition can be enhanced with a variety of assessment tools, and successful treatment requires an awareness of potential endogenous and exogenous contributors.

This article describes what is known about DSPD and uses the case example to illustrate diagnostic assessment and treatment choices. Intriguing data support various pathophysiologic explanations for DSPD (Box 1).1-6 Facilitating the adjustment of patients’ physiologic clocks is the overall goal in managing DSPD.

Box 1

What causes delayed sleep phase disorder?

In individuals normally entrained to the light/dark cycle, circadian rhythms are:

  • delayed by evening exposure to bright light (≥ 2,500 lux) prior to the core body temperature minimum (Tmin)
  • advanced by morning light exposure after the Tmin.1

These opposing effects attune most people to the light/dark cycle, with sleep and wakefulness occurring on a conventional schedule. Persons with delayed sleep phase disorder (DSPD) live at a delayed phase that resists advancement and is incompatible with their personal and social obligations.

Theories have been proposed, but DSPD’s etiology has not been fully explained. Affected adolescents may exhibit an extreme in circadian preference. Case reports also describe DSPD emerging after traumatic brain injury.2

Intriguing evidence supports various pathophysiologic explanations for DSPD. An abnormally long intrinsic circadian period (>25 hours) was recently demonstrated during temporal isolation in 1 individual with DSPD.3 Both this case report and controlled studies describe deviations from expected relationships between the sleep/wake cycle and physiologic circadian markers. Most consistently described are longer intervals from Tmin4 to sleep offset (final rise time) in DSPD patients compared with controls.

Other research suggests:

  • hypersensitivity to nocturnal photic stimulation in select DSPD patients5
  • impaired homeostatic sleep processes, as DSPD patients show a diminished ability to initiate sleep after sleep restriction, compared with controls.6

Extreme ‘eveningness’

Because of their extreme seemingly innate preference to retire and arise at relatively late clock hours (an “eveningness” trait), school-aged patients with DSPD represent a high-risk population for problematic sleepiness. In a survey of 612 high school students, the 63% who felt they needed more sleep on school nights showed a strong eveningness preference (as assessed by questionnaire), compared with students who described getting sufficient sleep.7 Other studies have revealed psychiatric morbidity (including affective and personality disorders), psychosocial impairment, and poor academic performance associated with the condition.8-10

DSPD may affect 7% to 16% of patients presenting with insomnia complaints in sleep medicine clinics.11 The condition appears most common among young cohorts and has been reported to affect up to 7% of adolescents in the United States.12 Its high frequency in this age group may be a pathologic exaggeration of the normal tendency toward delayed timing of sleep and wakefulness linked with pubertal development.13

Sleep and wakefulness regulation

Conceptually, 2 processes govern sleep and wakefulness:

  • The homeostatic drive to sleep (process S) is proportional to the duration of sleep restriction and becomes maximal at about 40 hours.
  • Circadian regulation (process C) creates a drive for wakefulness that variably opposes process S and depends upon intrinsic rhythms.14

Neurons of the suprachiasmatic nucleus in the hypothalamus exert master coordination of this sleep/wake rhythm, along with other behavioral and physiologic variables.15 Because the typical intrinsic period is slightly longer than 24 hours, synchronization to the 24-hour day (entrainment) is accomplished by environmental inputs (zeitgebers, or “time givers”), the most important of which is exposure to light.16

 

 

Misalignment between endogenous circadian rhythms and the light/dark cycle can result in circadian rhythm sleep disorders, such as:

  • delayed sleep timing (DSPD)
  • advanced sleep timing (advanced sleep phase disorder)
  • erratic sleep timing (irregular sleep/wake rhythm)
  • complete dissociation from the light/dark cycle (circadian rhythm sleep disorder, free-running type).

These 4 conditions are thought to involve predominantly intrinsic mechanisms, but circadian dysrhythmias also can be induced by exogenous factors. Extreme work schedules or rapid travel across time zones can challenge the circadian system’s ability to acclimate and the individual’s ability to achieve a desired sleep schedule.17

Differential diagnosis

Because DSPD relates primarily to an aberration in timing of sleep, it is characterized as a disorder only if the individual’s preferred schedule interferes substantially with social or occupational functioning. The International Classification of Sleep Disorders (ICSD) provides detailed diagnostic criteria (Table).17

Table

Diagnostic criteria for delayed sleep phase disorder

A. Delay exists in the phase of the major sleep period in relation to desired sleep time and wake-up time, as evidenced by:
  • a chronic or recurrent complaint of inability to fall asleep at a desired conventional clock time AND
  • inability to awaken at a desired and socially acceptable time.
B. When allowed to choose a preferred schedule, patients exhibit normal sleep quality and duration for age and maintain a delayed but stable phase of entrainment to the 24-hour sleep/wake pattern.
C. Monitoring with a sleep log or actigraphy (including sleep diary) for at least 7 days demonstrates a stable delay in the timing of the habitual sleep period.
D. The sleep disturbance is not better explained by another sleep disorder, medical or neurologic disorder, mental disorder, medication use, or substance use disorder.
Source: Adapted and reprinted with permission from International classification of sleep disorders. Diagnostic and coding manual. 2nd ed17

Depression and anxiety often manifest with sleep difficulties, as do inadequate sleep hygiene and other conditions associated with prolonged sleep initiation. According to ICSD criteria, primary insomnia can be differentiated from DSPD if the patient readily initiates and maintains sleep when allowed to sleep on his/her desired sleep/wake schedule. Accumulated evidence has largely debunked this notion, however, as polysomnographic studies have demonstrated both prolonged sleep latency and impaired sleep efficiency in DSPD patients versus matched controls.3

Assessment tools can complement the clinical history in diagnosing DSPD. Either a sleep log or actigraphy is required to demonstrate a stable phase delay, but actigraphy typically generates more reliable data.18 Actigraphs are compact “motion detectors” whose output while being worn by patients allows longitudinal assessment of sleep/wake parameters.

Eveningness tendencies of presumptive DSPD patients can be further verified with the Morningness-Eveningness Questionnaire (MEQ) (Box 2).19 Low scores are associated with evening types—felt to correspond to the endogenous circadian period—and can help narrow the differential diagnosis of sleep-initiation complaints.20

Box 2

A morning or evening person? A self-assessment questionnaire

The Morningness-Eveningness Questionnaire (MEQ) developed by Horne and Ostberg19 can be used to verify eveningness tendencies of patients with presumptive delayed sleep phase disorder. The MEQ is a 19-item self-assessment tool with responses that are assigned values totaling up to 86 points. Examples of the questions include:

  • Considering only your own ‘feeling best’ rhythm, at what time would you get up if you were entirely free to plan your day?
  • Considering only your own ‘feeling best’ rhythm, at what time would you go to bed if you were entirely free to plan your day?
  • How easy do you find it to get up each day?
  • When you have no commitments the next day, how much later do you go to bed compared to your usual bedtime?
  • One hears about ‘morning’ and ‘evening’ types of people. Which ONE of these types do you consider yourself to be?

Lower scores are associated with evening types—felt to correspond to the endogenous circadian period—and can help in narrowing the differential diagnosis of sleep-initiation complaints.20 Scores on the MEQ are interpreted as:

  • 70 to 86: definite morning type
  • 59 to 69: moderately morning type
  • 42 to 58: neither type
  • 31 to 41: moderately evening type
  • 16 to 30: definite evening type

CASE CONTINUED: ‘Definite evening type’

Jason scores 28 on the MEQ, consistent with a “definite evening type.” Actigraphic monitoring is scheduled during a school holiday, when he is instructed to sleep according to his preferred schedule with the least possible restriction.

A clearly delayed sleep phase is evident, with the habitual sleep period occurring between 5 AM and 1 PM. Even on days when he was quite sleep-restricted because of an enforced wake time, sleep onset on the ensuing evening was substantially delayed, suggesting an obligate nature for the delayed sleep/wake schedule. Overall, Jason had few complaints with respect to impaired alertness while on this unrestricted schedule and experienced a much more stable mood.

 

 

Interventions

Without physiologic assessments, understanding the patient’s “natural” sleep schedule can allow for rational recommendations about using phototherapy and oral melatonin (Figure21). However, referral to a sleep specialist is required unless the general psychiatrist has experience in treating circadian rhythm sleep disorders.

Morning phototherapy. Properly timed morning bright light therapy (≥2,500 lux) has been shown to help DSPD patients achieve physiologically measured sleep phase advances, objective improvements in daytime alertness, and earlier reported bedtimes compared with controls.22 Unfortunately, the described 2-hour treatment duration make this research protocol clinically impractical, and most clinicians commence with a 30-minute duration of therapy, as described in the seasonal affective disorder literature.

Relatively new and widely available blue light boxes have been reported to exhibit at least equivalent efficacy to bright light devices (as reported in the literature pertaining to seasonal affective disorder), but with markedly decreased light intensity and fewer associated adverse effects.23 As the research addressing their use in the treatment of circadian rhythm sleep disorders is still emerging, their future role remains uncertain.

Precautions. Most psychiatrists would not perform a physiologic determination of a patient’s circadian phase, and further undesired phase delays can occur if phototherapy is administered before the core body temperature minimum (Tmin).24 Also, use caution if prescribing phototherapy to patients taking photosensitizing drugs and/or those with ocular or retinal pathology.20

Evening light avoidance. Whether or not you prescribe morning phototherapy, recommending that DSPD patients avoid evening light is essential to avoid further induction or exacerbation of phase delays. Protective eyewear is warranted in instances where these advisory precautions are insufficient (see Related Resources). Such an intervention has been shown effective in decreasing light exposure and undesired phase advances in studies involving subjects exposed to simulated shift work.25

Oral melatonin. Abundant evidence supports melatonin use in achieving phase advances in individuals with DSPD.26,27 A synergistic effect can be obtained when melatonin is combined with phototherapy.28

Proper timing of melatonin to achieve a maximal phase advance can be estimated based on the individual’s dim light melatonin onset (DLMO), which occurs approximately 14 hours after the habitual (unrestricted) wake time.29 Maximal phase advances appear to occur when melatonin is given approximately 6 hours before the DLMO.26 Thus, a rational practice is to recommend that patients take melatonin 8 hours after their natural wake time. Doses of ≤0.5 mg appear to achieve the maximal chronobiotic effect while avoiding an undesired hypnotic effect.30

Precautions. Verifying the purity of over-the-counter melatonin is difficult. A review by the National Academy of Sciences states that short-term use of melatonin, ≤10 mg/d, appears to be safe in healthy adults but recommends caution in children/adolescents and women of reproductive age. Doses recommended for circadian-based interventions are typically physiologic in nature (i.e., ≤0.5 mg), which may serve to mitigate these concerns.

Adverse effects such as headaches, somnolence, hypotension, hypertension, gastrointestinal upset, and exacerbation of alopecia areata have been reported at higher melatonin doses in healthy adults and at lower doses in persons with preexisting central nervous system, cardiovascular, gastrointestinal, or dermatologic conditions.31


Figure Light and melatonin phase response curves: Normal vs. delayed

This schematic compares ‘normal sleep’ phase response curves (PRCs) to light and exogenous melatonin with postulated PRCs for an individual with delayed sleep phase disorder (DSPD), presumed to be 5 hours ‘out of phase.’ Y-axis shows the direction and relative magnitude of phase shifts produced by light or melatonin at times shown on the x-axis. X-axis covers >24 hours to better illustrate the PRCs.
Relationships between ‘normal sleepers’ and DSPD patients are depicted by:

  • rectangles (sleep period)
  • triangles (core body temperature minimum [Tmin])
  • arrows (dim light melatonin onsets [DLMOs]).

‘Normal’ sleep is shown to occur from midnight to 8 AM, and the DSPD patient’s sleep from 5 AM to 1 PM; DLMO and Tmin are similarly delayed by 5 hours in the DSPD patient. This schematic assumes that phase relationships are maintained in DSPD patients, which is not a certainty.
Source: Adapted from reference 21

CASE CONTINUED: Under the bright lights

Jason starts phototherapy treatment during his winter break, administering bright light daily upon natural awakening using a 10,000 lux light box for at least 30 minutes. As instructed, he gradually advances the time of administration by approximately 30 minutes every other day, striving for a nocturnal sleep period of 11 PM to 7 AM. He also wears protective eyewear to reduce light exposure during evening hours to avoid further delays in sleep phase. To further promote a phase advance, he takes oral melatonin, 0.5 mg/d at approximately 8 PM, as determined by his self-report and results of actigraphic recording.

 

 

Other options

Hypnotics. Little evidence supports the use of hypnotics in DSPD,32 and patients may show resistance to these drugs.33 Nevertheless, hypnotics can heighten confidence in the ability to initiate sleep in individuals with a concomitant conditioned insomnia.

With chronotherapy, patients are prescribed a sleep schedule that is delayed several hours incrementally until sleep is aligned to a target bedtime. The individual then is advised to rigorously maintain a regular sleep/wake schedule, repeating the process as necessary.

Although case reports have shown positive results with chronotherapy for DSPD,34 no controlled trials have demonstrated its efficacy or safety. One study reported high relapse rates,31 and 1 patient with DSPD developed free-running circadian rhythms.35 Clinical experience suggests chronotherapy is impractical for patients who must adhere to a fixed schedule.

Behavioral approaches

For an adolescent with DSPD, consider asking the school district to allow him or her a later school start-time. This alone often can substantially increase total sleep time and mitigate associated impairments.36 In all instances pursue and address external contributors to DSPD, such as poor sleep hygiene (including excessive caffeine use) and substance misuse.

Emphasize regular wake times, as arising later on weekends can cause phase delays.37 DSPD patients may have a concomitant conditioned insomnia that responds to evidence-based behavioral treatments.38

Whatever intervention you choose, schedule a follow-up appointment in approximately 2 months to evaluate patients’ progress and compliance. Encourage them to contact you with questions or concerns in the interim. Review sleep logs or actigraphy during this visit, and adjust the timing and/or nature of interventions as needed. Adolescents can be particularly noncompliant with clinical interventions, and therapeutic goals cannot be reached without their full investment.

Because no guidelines exist on how long to treat DSPD, stop on a “trial-and-error” basis when symptoms are controlled, and resume if they recur. Another approach is to maintain a desired sleep/wake schedule with bedtime melatonin and encourage continued adherence to other measures.

CASE CONTINUED: Maintenance therapy

Jason returns to the clinic approximately 10 weeks later. After an obviously concerted effort to adhere to treatment, his progress is quite remarkable. He rarely falls asleep later than 11 PM, so he is obtaining 2.5 hours more sleep each night before arising for school at 6:30 AM. Sleepiness at school is rarely problematic, and his mood is more stable.

He nevertheless describes a persistent tendency to retire and arise later and asks to continue melatonin and phototherapy. Because no guidelines exist for long-term therapy of DSPD, he is advised to switch melatonin to bedtime dosing (with a presumed phase-neutral “maintenance” effect), and to continue phototherapy as prescribed.

Related resources

  • Wyatt JK. Delayed sleep phase syndrome: pathophysiology and treatment options. Sleep. 2004;27(6):1195-1203.
  • Crowley SJ, Acebo C, Carskadon MA. Sleep, circadian rhythms, and delayed sleep phase in adolescence. Sleep Med. 2007;8(6):602-612.
  • National Sleep Foundation. Adolescent sleep needs and patterns: research report and resource guide. Washington, DC; 2000:1-30.
  • Products designed to assist in the avoidance of light at improper times. www.lowbluelights.com.

Disclosure

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

Comment on this article

Jason, age 16, has had difficulty with sleep initiation for 2 years. He describes going to bed at 10:30 PM on school nights but falling asleep no sooner than midnight and typically after 1:30 AM. He denies contributions from an “active mind” or environmental disturbances, and his bedroom contains no TV, computer, or other media devices. He does not sleep better with a change in environment. He denies pervasive low mood symptoms and believes his mood hinges predominantly on his ability to achieve sufficient sleep.

Once asleep, Jason generally enjoys good sleep consolidation until he needs to arise at 6:30 AM. His mother awakens him with difficulty, as he often sleeps through his alarm. He sleeps approximately 5 hours nightly during the school week, endorses impaired concentration, and often dozes during his first several classes. When he returns home from school, he finds it very difficult to resist napping.

On weekends he retires at 1 AM or later and typically falls asleep within 30 minutes. He usually awakens at noon but can sleep as late as 4:30 PM. He feels slightly more refreshed on weekends and describes his mood then as improved. During a recent spring break, he felt much better when allowed to sleep as much as he wanted.

Delayed sleep phase disorder (DSPD)—characterized by a pathological “night owl” circadian preference—is seen most commonly in adolescents and is associated with psychiatric morbidity, psychosocial impairment, and poor academic performance. Proper identification of the condition can be enhanced with a variety of assessment tools, and successful treatment requires an awareness of potential endogenous and exogenous contributors.

This article describes what is known about DSPD and uses the case example to illustrate diagnostic assessment and treatment choices. Intriguing data support various pathophysiologic explanations for DSPD (Box 1).1-6 Facilitating the adjustment of patients’ physiologic clocks is the overall goal in managing DSPD.

Box 1

What causes delayed sleep phase disorder?

In individuals normally entrained to the light/dark cycle, circadian rhythms are:

  • delayed by evening exposure to bright light (≥ 2,500 lux) prior to the core body temperature minimum (Tmin)
  • advanced by morning light exposure after the Tmin.1

These opposing effects attune most people to the light/dark cycle, with sleep and wakefulness occurring on a conventional schedule. Persons with delayed sleep phase disorder (DSPD) live at a delayed phase that resists advancement and is incompatible with their personal and social obligations.

Theories have been proposed, but DSPD’s etiology has not been fully explained. Affected adolescents may exhibit an extreme in circadian preference. Case reports also describe DSPD emerging after traumatic brain injury.2

Intriguing evidence supports various pathophysiologic explanations for DSPD. An abnormally long intrinsic circadian period (>25 hours) was recently demonstrated during temporal isolation in 1 individual with DSPD.3 Both this case report and controlled studies describe deviations from expected relationships between the sleep/wake cycle and physiologic circadian markers. Most consistently described are longer intervals from Tmin4 to sleep offset (final rise time) in DSPD patients compared with controls.

Other research suggests:

  • hypersensitivity to nocturnal photic stimulation in select DSPD patients5
  • impaired homeostatic sleep processes, as DSPD patients show a diminished ability to initiate sleep after sleep restriction, compared with controls.6

Extreme ‘eveningness’

Because of their extreme seemingly innate preference to retire and arise at relatively late clock hours (an “eveningness” trait), school-aged patients with DSPD represent a high-risk population for problematic sleepiness. In a survey of 612 high school students, the 63% who felt they needed more sleep on school nights showed a strong eveningness preference (as assessed by questionnaire), compared with students who described getting sufficient sleep.7 Other studies have revealed psychiatric morbidity (including affective and personality disorders), psychosocial impairment, and poor academic performance associated with the condition.8-10

DSPD may affect 7% to 16% of patients presenting with insomnia complaints in sleep medicine clinics.11 The condition appears most common among young cohorts and has been reported to affect up to 7% of adolescents in the United States.12 Its high frequency in this age group may be a pathologic exaggeration of the normal tendency toward delayed timing of sleep and wakefulness linked with pubertal development.13

Sleep and wakefulness regulation

Conceptually, 2 processes govern sleep and wakefulness:

  • The homeostatic drive to sleep (process S) is proportional to the duration of sleep restriction and becomes maximal at about 40 hours.
  • Circadian regulation (process C) creates a drive for wakefulness that variably opposes process S and depends upon intrinsic rhythms.14

Neurons of the suprachiasmatic nucleus in the hypothalamus exert master coordination of this sleep/wake rhythm, along with other behavioral and physiologic variables.15 Because the typical intrinsic period is slightly longer than 24 hours, synchronization to the 24-hour day (entrainment) is accomplished by environmental inputs (zeitgebers, or “time givers”), the most important of which is exposure to light.16

 

 

Misalignment between endogenous circadian rhythms and the light/dark cycle can result in circadian rhythm sleep disorders, such as:

  • delayed sleep timing (DSPD)
  • advanced sleep timing (advanced sleep phase disorder)
  • erratic sleep timing (irregular sleep/wake rhythm)
  • complete dissociation from the light/dark cycle (circadian rhythm sleep disorder, free-running type).

These 4 conditions are thought to involve predominantly intrinsic mechanisms, but circadian dysrhythmias also can be induced by exogenous factors. Extreme work schedules or rapid travel across time zones can challenge the circadian system’s ability to acclimate and the individual’s ability to achieve a desired sleep schedule.17

Differential diagnosis

Because DSPD relates primarily to an aberration in timing of sleep, it is characterized as a disorder only if the individual’s preferred schedule interferes substantially with social or occupational functioning. The International Classification of Sleep Disorders (ICSD) provides detailed diagnostic criteria (Table).17

Table

Diagnostic criteria for delayed sleep phase disorder

A. Delay exists in the phase of the major sleep period in relation to desired sleep time and wake-up time, as evidenced by:
  • a chronic or recurrent complaint of inability to fall asleep at a desired conventional clock time AND
  • inability to awaken at a desired and socially acceptable time.
B. When allowed to choose a preferred schedule, patients exhibit normal sleep quality and duration for age and maintain a delayed but stable phase of entrainment to the 24-hour sleep/wake pattern.
C. Monitoring with a sleep log or actigraphy (including sleep diary) for at least 7 days demonstrates a stable delay in the timing of the habitual sleep period.
D. The sleep disturbance is not better explained by another sleep disorder, medical or neurologic disorder, mental disorder, medication use, or substance use disorder.
Source: Adapted and reprinted with permission from International classification of sleep disorders. Diagnostic and coding manual. 2nd ed17

Depression and anxiety often manifest with sleep difficulties, as do inadequate sleep hygiene and other conditions associated with prolonged sleep initiation. According to ICSD criteria, primary insomnia can be differentiated from DSPD if the patient readily initiates and maintains sleep when allowed to sleep on his/her desired sleep/wake schedule. Accumulated evidence has largely debunked this notion, however, as polysomnographic studies have demonstrated both prolonged sleep latency and impaired sleep efficiency in DSPD patients versus matched controls.3

Assessment tools can complement the clinical history in diagnosing DSPD. Either a sleep log or actigraphy is required to demonstrate a stable phase delay, but actigraphy typically generates more reliable data.18 Actigraphs are compact “motion detectors” whose output while being worn by patients allows longitudinal assessment of sleep/wake parameters.

Eveningness tendencies of presumptive DSPD patients can be further verified with the Morningness-Eveningness Questionnaire (MEQ) (Box 2).19 Low scores are associated with evening types—felt to correspond to the endogenous circadian period—and can help narrow the differential diagnosis of sleep-initiation complaints.20

Box 2

A morning or evening person? A self-assessment questionnaire

The Morningness-Eveningness Questionnaire (MEQ) developed by Horne and Ostberg19 can be used to verify eveningness tendencies of patients with presumptive delayed sleep phase disorder. The MEQ is a 19-item self-assessment tool with responses that are assigned values totaling up to 86 points. Examples of the questions include:

  • Considering only your own ‘feeling best’ rhythm, at what time would you get up if you were entirely free to plan your day?
  • Considering only your own ‘feeling best’ rhythm, at what time would you go to bed if you were entirely free to plan your day?
  • How easy do you find it to get up each day?
  • When you have no commitments the next day, how much later do you go to bed compared to your usual bedtime?
  • One hears about ‘morning’ and ‘evening’ types of people. Which ONE of these types do you consider yourself to be?

Lower scores are associated with evening types—felt to correspond to the endogenous circadian period—and can help in narrowing the differential diagnosis of sleep-initiation complaints.20 Scores on the MEQ are interpreted as:

  • 70 to 86: definite morning type
  • 59 to 69: moderately morning type
  • 42 to 58: neither type
  • 31 to 41: moderately evening type
  • 16 to 30: definite evening type

CASE CONTINUED: ‘Definite evening type’

Jason scores 28 on the MEQ, consistent with a “definite evening type.” Actigraphic monitoring is scheduled during a school holiday, when he is instructed to sleep according to his preferred schedule with the least possible restriction.

A clearly delayed sleep phase is evident, with the habitual sleep period occurring between 5 AM and 1 PM. Even on days when he was quite sleep-restricted because of an enforced wake time, sleep onset on the ensuing evening was substantially delayed, suggesting an obligate nature for the delayed sleep/wake schedule. Overall, Jason had few complaints with respect to impaired alertness while on this unrestricted schedule and experienced a much more stable mood.

 

 

Interventions

Without physiologic assessments, understanding the patient’s “natural” sleep schedule can allow for rational recommendations about using phototherapy and oral melatonin (Figure21). However, referral to a sleep specialist is required unless the general psychiatrist has experience in treating circadian rhythm sleep disorders.

Morning phototherapy. Properly timed morning bright light therapy (≥2,500 lux) has been shown to help DSPD patients achieve physiologically measured sleep phase advances, objective improvements in daytime alertness, and earlier reported bedtimes compared with controls.22 Unfortunately, the described 2-hour treatment duration make this research protocol clinically impractical, and most clinicians commence with a 30-minute duration of therapy, as described in the seasonal affective disorder literature.

Relatively new and widely available blue light boxes have been reported to exhibit at least equivalent efficacy to bright light devices (as reported in the literature pertaining to seasonal affective disorder), but with markedly decreased light intensity and fewer associated adverse effects.23 As the research addressing their use in the treatment of circadian rhythm sleep disorders is still emerging, their future role remains uncertain.

Precautions. Most psychiatrists would not perform a physiologic determination of a patient’s circadian phase, and further undesired phase delays can occur if phototherapy is administered before the core body temperature minimum (Tmin).24 Also, use caution if prescribing phototherapy to patients taking photosensitizing drugs and/or those with ocular or retinal pathology.20

Evening light avoidance. Whether or not you prescribe morning phototherapy, recommending that DSPD patients avoid evening light is essential to avoid further induction or exacerbation of phase delays. Protective eyewear is warranted in instances where these advisory precautions are insufficient (see Related Resources). Such an intervention has been shown effective in decreasing light exposure and undesired phase advances in studies involving subjects exposed to simulated shift work.25

Oral melatonin. Abundant evidence supports melatonin use in achieving phase advances in individuals with DSPD.26,27 A synergistic effect can be obtained when melatonin is combined with phototherapy.28

Proper timing of melatonin to achieve a maximal phase advance can be estimated based on the individual’s dim light melatonin onset (DLMO), which occurs approximately 14 hours after the habitual (unrestricted) wake time.29 Maximal phase advances appear to occur when melatonin is given approximately 6 hours before the DLMO.26 Thus, a rational practice is to recommend that patients take melatonin 8 hours after their natural wake time. Doses of ≤0.5 mg appear to achieve the maximal chronobiotic effect while avoiding an undesired hypnotic effect.30

Precautions. Verifying the purity of over-the-counter melatonin is difficult. A review by the National Academy of Sciences states that short-term use of melatonin, ≤10 mg/d, appears to be safe in healthy adults but recommends caution in children/adolescents and women of reproductive age. Doses recommended for circadian-based interventions are typically physiologic in nature (i.e., ≤0.5 mg), which may serve to mitigate these concerns.

Adverse effects such as headaches, somnolence, hypotension, hypertension, gastrointestinal upset, and exacerbation of alopecia areata have been reported at higher melatonin doses in healthy adults and at lower doses in persons with preexisting central nervous system, cardiovascular, gastrointestinal, or dermatologic conditions.31


Figure Light and melatonin phase response curves: Normal vs. delayed

This schematic compares ‘normal sleep’ phase response curves (PRCs) to light and exogenous melatonin with postulated PRCs for an individual with delayed sleep phase disorder (DSPD), presumed to be 5 hours ‘out of phase.’ Y-axis shows the direction and relative magnitude of phase shifts produced by light or melatonin at times shown on the x-axis. X-axis covers >24 hours to better illustrate the PRCs.
Relationships between ‘normal sleepers’ and DSPD patients are depicted by:

  • rectangles (sleep period)
  • triangles (core body temperature minimum [Tmin])
  • arrows (dim light melatonin onsets [DLMOs]).

‘Normal’ sleep is shown to occur from midnight to 8 AM, and the DSPD patient’s sleep from 5 AM to 1 PM; DLMO and Tmin are similarly delayed by 5 hours in the DSPD patient. This schematic assumes that phase relationships are maintained in DSPD patients, which is not a certainty.
Source: Adapted from reference 21

CASE CONTINUED: Under the bright lights

Jason starts phototherapy treatment during his winter break, administering bright light daily upon natural awakening using a 10,000 lux light box for at least 30 minutes. As instructed, he gradually advances the time of administration by approximately 30 minutes every other day, striving for a nocturnal sleep period of 11 PM to 7 AM. He also wears protective eyewear to reduce light exposure during evening hours to avoid further delays in sleep phase. To further promote a phase advance, he takes oral melatonin, 0.5 mg/d at approximately 8 PM, as determined by his self-report and results of actigraphic recording.

 

 

Other options

Hypnotics. Little evidence supports the use of hypnotics in DSPD,32 and patients may show resistance to these drugs.33 Nevertheless, hypnotics can heighten confidence in the ability to initiate sleep in individuals with a concomitant conditioned insomnia.

With chronotherapy, patients are prescribed a sleep schedule that is delayed several hours incrementally until sleep is aligned to a target bedtime. The individual then is advised to rigorously maintain a regular sleep/wake schedule, repeating the process as necessary.

Although case reports have shown positive results with chronotherapy for DSPD,34 no controlled trials have demonstrated its efficacy or safety. One study reported high relapse rates,31 and 1 patient with DSPD developed free-running circadian rhythms.35 Clinical experience suggests chronotherapy is impractical for patients who must adhere to a fixed schedule.

Behavioral approaches

For an adolescent with DSPD, consider asking the school district to allow him or her a later school start-time. This alone often can substantially increase total sleep time and mitigate associated impairments.36 In all instances pursue and address external contributors to DSPD, such as poor sleep hygiene (including excessive caffeine use) and substance misuse.

Emphasize regular wake times, as arising later on weekends can cause phase delays.37 DSPD patients may have a concomitant conditioned insomnia that responds to evidence-based behavioral treatments.38

Whatever intervention you choose, schedule a follow-up appointment in approximately 2 months to evaluate patients’ progress and compliance. Encourage them to contact you with questions or concerns in the interim. Review sleep logs or actigraphy during this visit, and adjust the timing and/or nature of interventions as needed. Adolescents can be particularly noncompliant with clinical interventions, and therapeutic goals cannot be reached without their full investment.

Because no guidelines exist on how long to treat DSPD, stop on a “trial-and-error” basis when symptoms are controlled, and resume if they recur. Another approach is to maintain a desired sleep/wake schedule with bedtime melatonin and encourage continued adherence to other measures.

CASE CONTINUED: Maintenance therapy

Jason returns to the clinic approximately 10 weeks later. After an obviously concerted effort to adhere to treatment, his progress is quite remarkable. He rarely falls asleep later than 11 PM, so he is obtaining 2.5 hours more sleep each night before arising for school at 6:30 AM. Sleepiness at school is rarely problematic, and his mood is more stable.

He nevertheless describes a persistent tendency to retire and arise later and asks to continue melatonin and phototherapy. Because no guidelines exist for long-term therapy of DSPD, he is advised to switch melatonin to bedtime dosing (with a presumed phase-neutral “maintenance” effect), and to continue phototherapy as prescribed.

Related resources

  • Wyatt JK. Delayed sleep phase syndrome: pathophysiology and treatment options. Sleep. 2004;27(6):1195-1203.
  • Crowley SJ, Acebo C, Carskadon MA. Sleep, circadian rhythms, and delayed sleep phase in adolescence. Sleep Med. 2007;8(6):602-612.
  • National Sleep Foundation. Adolescent sleep needs and patterns: research report and resource guide. Washington, DC; 2000:1-30.
  • Products designed to assist in the avoidance of light at improper times. www.lowbluelights.com.

Disclosure

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

References

1. Khalsa SB, Jewett ME, Cajochen C, et al. A phase response curve to single bright light pulses in human subjects. J Physiol. 2003;549(Pt 3):945-952.

2. Quinto C, Gellido C, Chokroverty S, et al. Posttraumatic delayed sleep phase syndrome. Neurology. 2000;54(1):250-252.

3. Campbell SS, Murphy PJ. Delayed sleep phase disorder in temporal isolation. Sleep. 2007;30(9):1225-1228.

4. Uchiyama M, Okawa M, Shibui K, et al. Altered phase relation between sleep timing and core body temperature rhythm in delayed sleep phase syndrome and non-24-hour sleep-wake syndrome in humans. Neurosci Lett. 2000;294(2):101-104.

5. Aoki H, Ozeki Y, Yamada N. Hypersensitivity of melatonin suppression in response to light in patients with delayed sleep phase syndrome. Chronobiol Int. 2001;18(2):263-271.

6. Uchiyama M, Okawa M, Shibui K, et al. Poor compensatory function for sleep loss as a pathogenic factor in patients with delayed sleep phase syndrome. Sleep. 2000;23(4):553-558.

7. Mercer PW, Merritt SL, Cowell JM. Differences in reported sleep need among adolescents. J Adolesc Health. 1998;23(5):259-263.

8. Krahn LE, Pankratz VS, Harris AM, et al. Long-term outcome of adolescents with delayed sleep phase disorder [abstract]. Sleep. 2003;26:A115.-

9. Dagan Y, Stein D, Steinbock M, et al. Frequency of delayed sleep phase syndrome among hospitalized adolescent psychiatric patients. J Psychosom Res. 1998;45(1):15-20.

10. Thorpy MJ, Korman E, Spielman AJ, et al. Delayed sleep phase syndrome in adolescents. J Adolesc Health Care. 1998;9(1):22-27.

11. Regestein QR, Monk TH. Delayed sleep phase syndrome: a review of the clinical aspects. Am J Psychiatry. 1995;152(4):602-608.

12. Pelayo RP, Thorpy MJ, Glovinsky P. Prevalence of delayed sleep phase syndrome among adolescents [abstract]. Sleep Res. 1988;17:391.-

13. Gau SF, Soong WT. The transition of sleep-wake patterns in early adolescence. Sleep. 2003;26(4):449-454.

14. Beersma DG, Gordijn MC. Circadian control of the sleep-wake cycle. Physiol Behav. 2007;90(2-3):190-195.

15. Ralph MR, Foster RG, Davis FC, et al. Transplanted suprachiasmatic nucleus determines circadian period. Science. 1990;247(4945):975-978.

16. Waterhouse J, DeCoursey PJ. Chronobiology: biological timekeeping. Sunderland, MA: Sinauer Associates, Inc. Publishers; 2004:291-323.

17. American Academy of Sleep Medicine. International classification of sleep disorders. Diagnostic and coding manual. 2nd ed. Westchester, IL: American Academy of Sleep Medicine; 2005.

18. Bradshaw DA, Yanagi MA, Pak ES, et al. Nightly sleep duration in the 2-week period preceding multiple sleep latency testing. J Clin Sleep Med. 2007;3(6):613-619.

19. Horne JA, Ostberg O. A self-assessment questionnaire to determine morningness-eveningness in human circadian rhythms. Int J Chronobiol. 1976;4(2):97-110.

20. Sack RL, Auckley D, Auger RR, et al. Circadian rhythm sleep disorders: Part I. basic principles, shift work and jet lag disorders. Sleep. 2007;30(11):1460-1483.

21. Burgess HJ, Sharkey KM, Eastman CI. Bright light, dark and melatonin can promote circadian adaptation in night shift workers. Sleep Med Rev. 2002;6(5):407-420.

22. Rosenthal NE, Joseph-Vanderpool JR, Levendosky AA, et al. Phase-shifting effects of bright morning light as treatment for delayed sleep phase syndrome. Sleep. 1990;13(4):354-361.

23. Glickman G, Byrne B, Pineda C, et al. Light therapy for seasonal affective disorder with blue narrow-band light-emitting diodes (LEDs). Biol Psychiatry. 2006;59:502-507.

24. Czeisler C, Wright K, Jr. Influence of light on circadian rhythmicity in humans. New York, NY: Marcel Dekker; 1999:149-180.

25. Crowley SJ, Lee C, Tseng CY, et al. Combinations of bright light, scheduled dark, sunglasses, and melatonin to facilitate circadian entrainment to night shift work. J Biol Rhythms. 2003;18(6):513-523.

26. Mundey K, Benloucif S, Harsanyi K, et al. Phase-dependent treatment of delayed sleep phase syndrome with melatonin. Sleep. 2005;28(10):1271-1278.

27. Sack RL, Auckley D, Auger RR, et al. Circadian rhythm sleep disorders: Part II, advanced sleep phase disorder, delayed sleep phase disorder, free-running disorder, and irregular sleep-wake rhythm. Sleep. 2007;30(11):1484-1501.

28. Revell VL, Burgess HJ, Gazda CJ, et al. Advancing human circadian rhythms with afternoon melatonin and morning intermittent bright light. J Clin Endocrinol Metab. 2006;91(1):54-59.

29. Burgess HJ, Eastman CI. The dim light melatonin onset following fixed and free sleep schedules. J Sleep Res. 2005;14(3):229-237.

30. Lewy AJ. Clinical applications of melatonin in circadian disorders. Dialog Clin Neurosci. 2003;5:399-413.

31. Committee on the Framework for Evaluating the Safety of Dietary Supplements FaNB, Board on Life Sciences, Institute of Medicine and National Research Council of the National Academies. Dietary supplements: a framework for evaluating safety. Washington, DC: The National Academies Press; 2005.

32. Ito A, Ando K, Hayakawa T, et al. Long-term course of adult patients with delayed sleep phase syndrome. Jpn J Psychiatry Neurol. 1993;47(3):563-567.

33. Auger RR. Circadian rhythm sleep disorder, delayed sleep phase type (pediatric case). In: Winkelman JW (chair), Henderson JH, Kotagal S, et al, eds. Case book of sleep medicine. Westchester, IL: American Academy of Sleep Medicine; 2008:195-199.

34. Czeisler C, Weitzman E, Moore, et al. Chronotherapy: resetting the circadian clocks of patients with delayed sleep phase insomnia. Sleep. 1981;4:1-21.

35. Oren DA, Wehr TA. Hypernyctohemeral syndrome after chronotherapy for delayed sleep phase syndrome. N Engl J Med. 1992;327(24):1762.-

36. Wahlstrom K. Changing times: findings from the first longitudinal study of later high school start times. NASSP Bulletin. 2002;86(633):3-21.

37. Burgess HJ, Eastman CI. A late wake time phase delays the human dim light melatonin rhythm. Neurosci Lett. 2006;395(3):191-195.

38. Morgenthaler T, Kramer M, Alessi C, et al. Practice parameters for the psychological and behavioral treatment of insomnia: an update. An American Academy of Sleep Medicine report. Sleep. 2006;29(11):1415-1419.

References

1. Khalsa SB, Jewett ME, Cajochen C, et al. A phase response curve to single bright light pulses in human subjects. J Physiol. 2003;549(Pt 3):945-952.

2. Quinto C, Gellido C, Chokroverty S, et al. Posttraumatic delayed sleep phase syndrome. Neurology. 2000;54(1):250-252.

3. Campbell SS, Murphy PJ. Delayed sleep phase disorder in temporal isolation. Sleep. 2007;30(9):1225-1228.

4. Uchiyama M, Okawa M, Shibui K, et al. Altered phase relation between sleep timing and core body temperature rhythm in delayed sleep phase syndrome and non-24-hour sleep-wake syndrome in humans. Neurosci Lett. 2000;294(2):101-104.

5. Aoki H, Ozeki Y, Yamada N. Hypersensitivity of melatonin suppression in response to light in patients with delayed sleep phase syndrome. Chronobiol Int. 2001;18(2):263-271.

6. Uchiyama M, Okawa M, Shibui K, et al. Poor compensatory function for sleep loss as a pathogenic factor in patients with delayed sleep phase syndrome. Sleep. 2000;23(4):553-558.

7. Mercer PW, Merritt SL, Cowell JM. Differences in reported sleep need among adolescents. J Adolesc Health. 1998;23(5):259-263.

8. Krahn LE, Pankratz VS, Harris AM, et al. Long-term outcome of adolescents with delayed sleep phase disorder [abstract]. Sleep. 2003;26:A115.-

9. Dagan Y, Stein D, Steinbock M, et al. Frequency of delayed sleep phase syndrome among hospitalized adolescent psychiatric patients. J Psychosom Res. 1998;45(1):15-20.

10. Thorpy MJ, Korman E, Spielman AJ, et al. Delayed sleep phase syndrome in adolescents. J Adolesc Health Care. 1998;9(1):22-27.

11. Regestein QR, Monk TH. Delayed sleep phase syndrome: a review of the clinical aspects. Am J Psychiatry. 1995;152(4):602-608.

12. Pelayo RP, Thorpy MJ, Glovinsky P. Prevalence of delayed sleep phase syndrome among adolescents [abstract]. Sleep Res. 1988;17:391.-

13. Gau SF, Soong WT. The transition of sleep-wake patterns in early adolescence. Sleep. 2003;26(4):449-454.

14. Beersma DG, Gordijn MC. Circadian control of the sleep-wake cycle. Physiol Behav. 2007;90(2-3):190-195.

15. Ralph MR, Foster RG, Davis FC, et al. Transplanted suprachiasmatic nucleus determines circadian period. Science. 1990;247(4945):975-978.

16. Waterhouse J, DeCoursey PJ. Chronobiology: biological timekeeping. Sunderland, MA: Sinauer Associates, Inc. Publishers; 2004:291-323.

17. American Academy of Sleep Medicine. International classification of sleep disorders. Diagnostic and coding manual. 2nd ed. Westchester, IL: American Academy of Sleep Medicine; 2005.

18. Bradshaw DA, Yanagi MA, Pak ES, et al. Nightly sleep duration in the 2-week period preceding multiple sleep latency testing. J Clin Sleep Med. 2007;3(6):613-619.

19. Horne JA, Ostberg O. A self-assessment questionnaire to determine morningness-eveningness in human circadian rhythms. Int J Chronobiol. 1976;4(2):97-110.

20. Sack RL, Auckley D, Auger RR, et al. Circadian rhythm sleep disorders: Part I. basic principles, shift work and jet lag disorders. Sleep. 2007;30(11):1460-1483.

21. Burgess HJ, Sharkey KM, Eastman CI. Bright light, dark and melatonin can promote circadian adaptation in night shift workers. Sleep Med Rev. 2002;6(5):407-420.

22. Rosenthal NE, Joseph-Vanderpool JR, Levendosky AA, et al. Phase-shifting effects of bright morning light as treatment for delayed sleep phase syndrome. Sleep. 1990;13(4):354-361.

23. Glickman G, Byrne B, Pineda C, et al. Light therapy for seasonal affective disorder with blue narrow-band light-emitting diodes (LEDs). Biol Psychiatry. 2006;59:502-507.

24. Czeisler C, Wright K, Jr. Influence of light on circadian rhythmicity in humans. New York, NY: Marcel Dekker; 1999:149-180.

25. Crowley SJ, Lee C, Tseng CY, et al. Combinations of bright light, scheduled dark, sunglasses, and melatonin to facilitate circadian entrainment to night shift work. J Biol Rhythms. 2003;18(6):513-523.

26. Mundey K, Benloucif S, Harsanyi K, et al. Phase-dependent treatment of delayed sleep phase syndrome with melatonin. Sleep. 2005;28(10):1271-1278.

27. Sack RL, Auckley D, Auger RR, et al. Circadian rhythm sleep disorders: Part II, advanced sleep phase disorder, delayed sleep phase disorder, free-running disorder, and irregular sleep-wake rhythm. Sleep. 2007;30(11):1484-1501.

28. Revell VL, Burgess HJ, Gazda CJ, et al. Advancing human circadian rhythms with afternoon melatonin and morning intermittent bright light. J Clin Endocrinol Metab. 2006;91(1):54-59.

29. Burgess HJ, Eastman CI. The dim light melatonin onset following fixed and free sleep schedules. J Sleep Res. 2005;14(3):229-237.

30. Lewy AJ. Clinical applications of melatonin in circadian disorders. Dialog Clin Neurosci. 2003;5:399-413.

31. Committee on the Framework for Evaluating the Safety of Dietary Supplements FaNB, Board on Life Sciences, Institute of Medicine and National Research Council of the National Academies. Dietary supplements: a framework for evaluating safety. Washington, DC: The National Academies Press; 2005.

32. Ito A, Ando K, Hayakawa T, et al. Long-term course of adult patients with delayed sleep phase syndrome. Jpn J Psychiatry Neurol. 1993;47(3):563-567.

33. Auger RR. Circadian rhythm sleep disorder, delayed sleep phase type (pediatric case). In: Winkelman JW (chair), Henderson JH, Kotagal S, et al, eds. Case book of sleep medicine. Westchester, IL: American Academy of Sleep Medicine; 2008:195-199.

34. Czeisler C, Weitzman E, Moore, et al. Chronotherapy: resetting the circadian clocks of patients with delayed sleep phase insomnia. Sleep. 1981;4:1-21.

35. Oren DA, Wehr TA. Hypernyctohemeral syndrome after chronotherapy for delayed sleep phase syndrome. N Engl J Med. 1992;327(24):1762.-

36. Wahlstrom K. Changing times: findings from the first longitudinal study of later high school start times. NASSP Bulletin. 2002;86(633):3-21.

37. Burgess HJ, Eastman CI. A late wake time phase delays the human dim light melatonin rhythm. Neurosci Lett. 2006;395(3):191-195.

38. Morgenthaler T, Kramer M, Alessi C, et al. Practice parameters for the psychological and behavioral treatment of insomnia: an update. An American Academy of Sleep Medicine report. Sleep. 2006;29(11):1415-1419.

Issue
Current Psychiatry - 08(03)
Issue
Current Psychiatry - 08(03)
Page Number
55-65
Page Number
55-65
Publications
Publications
Topics
Article Type
Display Headline
‘Night owls’: Reset the physiologic clock in delayed sleep phase disorder
Display Headline
‘Night owls’: Reset the physiologic clock in delayed sleep phase disorder
Legacy Keywords
delayed sleep phase disorder;delayed circadian cycle;insomnia;circadian rhythm;circadian preference;phototherapy,light avoidance;actigraphy;R. Robert Auger
Legacy Keywords
delayed sleep phase disorder;delayed circadian cycle;insomnia;circadian rhythm;circadian preference;phototherapy,light avoidance;actigraphy;R. Robert Auger
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Fibromyalgia: Psychiatric drugs target CNS-linked symptoms

Article Type
Changed
Tue, 12/11/2018 - 15:06
Display Headline
Fibromyalgia: Psychiatric drugs target CNS-linked symptoms

WEB AUDIO
Listen to Dr. Stanford discuss,
"Is fibromyalgia a somatoform disorder?

Comment on this article

Patients with fibromyalgia are a heterogeneous group, yet many describe a common experience: seeing multiple physicians who seem unable or unwilling to provide a diagnosis or treat their symptoms. This situation may be changing with the recent FDA approval of an anticonvulsant and 2 antidepressants for managing fibromyalgia symptoms.

These medications—pregabalin, duloxetine, and milnacipran—reflect a revised understanding of fibromyalgia as a CNS condition, rather than an inflammatory process in the muscles or connective tissue. As a result, psychiatrists—because of our experience with CNS phenomena and managing antidepressant and anticonvulsant medications—are likely to play a larger role in treating fibromyalgia.

CASE REPORT: ‘Just too tired’

Ms. D, age 50, has a history of migraine headaches and is referred by her primary physician for evaluation of depression and anxiety. She reports deteriorating mood over 6 months, beginning when a minor car accident left her “very sore the next day.”

“Nothing helps” the persistent pain in her back, shoulders, and thighs, which she rates as 7 to 8 on a 0-to-10 pain scale. She describes an intense ache, “like having the flu,” that worsens with activity and in stressful situations. She also experiences nausea and intermittent diarrhea, debilitating fatigue, and sleep disturbance.

Ms. D reports she is depressed because she feels “just too tired” after work to keep up with social activities or housework. Her physician’s referral notes a normal physical exam except for tenderness over her upper back and hips. Laboratory testing is negative.

Making the diagnosis

American College of Rheumatology (ACR) criteria for fibromyalgia require widespread pain for at least 3 months. “Widespread” is defined as pain in the axial skeleton, left and right sides of the body, and above and below the waist. Pain must be found in at least 11 of 18 tender point sites on digital palpation using a force of approximately 4 kg/cm2.1 For many fibromyalgia patients, however, musculoskeletal pain is not their most problematic symptom (Table 1). They may suffer:

  • migraine and tension headaches (10% to 80% of patients)
  • irritable bowel syndrome (32% to 80%)2
  • mood disorders (major depressive disorder [62%], bipolar disorder [11%])
  • anxiety disorders (panic disorder [29%], posttraumatic stress disorder [21%], social phobia [19%]).3

ACR criteria are useful in research but lack many common symptoms and comorbidities. A structured interview that follows the DSM-IV-TR format incorporates other symptoms into the diagnosis (Table 2).4

Because patients with fibromyalgia often meet criteria for somatization or somatoform disorders, how to classify them—as medically or psychiatrically ill—is controversial. Some patients believe their mood or anxiety problem stems from the difficulty they experience dealing with their physical symptoms, and if they could feel better physically they would not be depressed or anxious. Others believe their psychiatric symptoms impede their ability to help themselves feel better.

Consider fibromyalgia in any patient with widespread pain of unknown cause. Before making the diagnosis, rule out other illnesses that present with similar symptoms (Table 3). Because many patients newly diagnosed with fibromyalgia worry that something “more serious” may be going on, confirm the diagnosis with appropriate testing and physical examination, usually by a rheumatologist or primary care physician.

Table 1

Medical and cognitive symptoms related to fibromyalgia

Neurologic
Tension/migraine headache
Psychiatric
Memory and cognitive difficulties
Mood disturbance
Anxiety disorders
Ear, nose, throat
Sicca symptoms
Vasomotor rhinitis
Vestibular complaints
Cardiovascular
Neurally mediated hypotension
Mitral valve prolapse
Noncardiac chest pain
Gastrointestinal
Esophageal dysmotility
Irritable bowel syndrome
Urological
Interstitial cystitis
Gynecological
Vulvodynia
Chronic pelvic pain
Oral/dental
Temporomandibular joint syndrome
Other (general)
Chronic fatigue syndrome
Sleep disturbances
Idiopathic low back pain
Multiple chemical sensitivity
Table 2

Fibromyalgia: Structured interview for diagnosis

A. Generalized pain affecting the axial, plus upper and lower segments, plus left and rights sides of the body
Either B or C:
B. At least 11 of 18 reproducible tender points
C. At least 4 of the following symptoms:
  • Generalized fatigue
  • Headaches
  • Sleep disturbance
  • Neuropsychiatric complaints
  • Numbness, tingling sensations
  • Irritable bowel symptoms
D. It cannot be established that disturbance was due to another systematic condition
Source: Reference 4
Table 3

Differentiating fibromyalgia from illnesses with similar symptoms

IllnessTests to differentiate from primary fibromyalgia
Rheumatic diseases
Osteoarthritis
Spondyloarthropathies, rheumatoid arthritis
Systemic lupus erythematosus, polymyalgia rheumatica
Osteomalacia
Myopathy

Radiographs
Rheumatic markers (antinuclear antibody, rheumatoid factor, antibodies)
Inflammatory markers (ESR, C-reactive protein)
Vitamin D level
CPK
Neurologic
Multiple sclerosis, Chiari’s malformation, spinal stenosis, radiculopathy
Neuropathy

MRI
EMG
Endocrine
Hypothyroidism
Diabetes

TSH
Basic chemistry panel with fasting glucose
Other
Infectious
  Lyme disease
  Hepatitis
Anemia
Cancers

CBC
Lyme titer
Hepatitis antibody panel, liver function tests
Hemoglobin/hematocrit
Routine cancer screening tests, bone scan, blood chemistries specific for suspected primary cancer
ESR: erythrocyte sedimentation rate; CPK: creatine phosphokinase; EMG: electromyography; TSH: thyroid-stimulating hormone; CBC: complete blood count
 

 

CASE CONTINUED: Central pain sensitization

As you elicit more details about Ms. D’s mood, she continues to focus on her physical symptoms. She states that some days she wishes to die because her pain gets so bad, but she denies any plan or intent to harm herself. She worries that her symptoms will worsen and that she will become completely disabled.

Her primary physician attempted to relieve Ms. D’s pain with multiple trials of nonsteroidal anti-inflammatory drugs (NSAIDs) and cyclobenzaprine. She says she gained no benefit from the NSAIDs and discontinued the muscle relaxant because it made her too sleepy. Fibromyalgia affects 3.5% of women and 0.5% of men.5 It runs in families with histories of fibromyalgia and major mood and anxiety disorders, suggesting genetic links.6 Defects in genes controlling serotonin and norepinephrine have been implicated.7-9

Fibromyalgia patients show lower levels of serotonin, norepinephrine, and dopamine metabolites in cerebrospinal fluid (CSF), compared with controls.10 These neurotransmitters may inhibit descending pain pathways in the CNS, and low levels in the brain and spinal cord may inhibit CNS regulation of pain impulses from the periphery.11

Although many patients describe muscle pain, evidence suggests central pain augmentation rather than an abnormality of muscle or connective tissue.12 Some studies have found evidence of “windup,” in which second-order neurons in the spinal cord become sensitized by repeated signals from first-order neurons in the periphery, resulting in amplified and prolonged pain signals traveling to the brain.13

Levels of substance P—a primary transmitter of pain impulses—are significantly higher in CSF of fibromyalgia patients compared with controls.14 This finding, in addition to low levels of serotonin and norepinephrine, indicates that pain signals are ascending unchecked to be processed by the brain.

Neuroimaging studies confirm this observation. In a study using functional magnetic resonance imaging (fMRI), researchers applied pressure to the thumbnails of fibromyalgia patients and controls until each subject reported pain:

  • Twice as much pressure was required before controls rated their pain at a level similar to that of fibromyalgia patients.
  • When controls and fibromyalgia patients reported similar pain, a very high degree of overlap was seen in brain areas responsible for pain processing. This indicates that fibromyalgia patients and controls were experiencing the pain they reported in the same way.15

Treating the whole patient

As a clinician who specializes in fibromyalgia, I counteract my patients’ and my own frustration with this condition by structuring office visits, determining realistic treatment goals, and treating all symptoms as part of a common syndrome rather than individual illnesses.

Structure office visits. Before every visit, have patients rate each symptom domain and write their top 2 or 3 concerns for that day (Click here for a sample form). Focusing on the patient’s most troublesome symptoms can help both of you feel greater satisfaction with treatment.

Educate patients. Ask them to discuss their beliefs about fibromyalgia; many know others with this condition or have researched diagnosis and treatment. Before developing a treatment plan, explain that their symptoms are chronic and all part of the same syndrome. Describe their pain as a complex phenomenon with possible peripheral and CNS components. Guide them to reputable Web sites and resources (see Related Resources).

Set realistic expectations. Many patients expect to resume an energetic and pain-free life, which usually is not the case with fibromyalgia (Box). Most medications are considered successful if they reduce pain by 30% to 50%, and side effects can be problematic. Discuss side effects before treatment begins to reduce patients’ anxiety and improve compliance in the first weeks.

Cognitive-behavioral therapy (CBT) for fibromyalgia incorporates relaxation techniques, helping patients view symptoms as manageable, reinforcing adaptive coping skills, and teaching them how to monitor thoughts, feelings, and behavior to change the view that they are helpless victims. A modest course of 6 weekly group CBT sessions significantly improved physical functioning in 25% of fibromyalgia patients (n=76) compared with 12% in a standard-care group (n=69), even though patients’ pain severity did not improve.16

Recommend exercise, lifestyle changes. Aerobic exercise can significantly improve well-being and physical functioning in fibromyalgia patients.17 Low-impact aerobics, such as done in warm water, usually are well tolerated, although any low-impact exercise can help. Because fibromyalgia symptoms often increase with physical activity, counsel patients to begin with a few minutes daily and increase very slowly each week.
 

 


Lifestyle changes are as important as medications in controlling fibromyalgia symptoms. In addition to exercise, recommend that patients:

  • follow a daily routine
  • pace activity to avoid exacerbating symptoms
  • reduce stress.
Sometimes, I use the analogy of diabetes: treating fibromyalgia with medication but without changing lifestyle is like prescribing medication for a diabetic patient without changing diet. Follow up on this “homework” at each visit to reinforce that patients helping themselves is an important part of treatment.

Box

Managing unrealistic expectations of fibromyalgia patients

BELIEF: ‘A magic pill exists that will resolve all my symptoms and have no side effects’

Clinical evidence: Most medications that have been studied were effective in 30% to 50% of patients and reduced pain scores by 30% to 50%.

Patient education: Explain to the patient with a pain rating of 7 at the first visit that achieving a pain level of 3 to 4 may be possible with treatment. Even with successful treatment, symptoms may flare intermittently. As with any treatment, adverse effects may occur. Discuss these, so the patient is not surprised.

BELIEF: ‘I can’t exercise’

Clinical evidence: Most patients experience more fatigue and pain with physical activity, but exercise is important to maintain physical function.

Patient education: When discussing an exercise program, focus on what the patient can do. Most patients attempt too much, too soon; advise them to start at a tolerable level (such as 2 to 3 minutes of aerobic activity daily for the first week) and gradually increase as tolerated.

BELIEF: ‘You (the psychiatrist) can make me feel better’

Clinical evidence: Psychiatrists can help by prescribing appropriate medications, but much of the burden falls on the patient to maintain a healthy, active lifestyle and to manage stressors in an adaptive manner.

Patient education: A fibromyalgia patient may find relief with a medication, but symptoms may flare if they ‘overdo’ and take on too many physical or emotional stressors. A consistent, healthy routine is ideal.

BELIEF: ‘I will eventually become disabled by fibromyalgia’

Clinical evidence: Despite little long-term research on fibromyalgia patients, most evidence points to a chronic, fluctuating syndrome that does not worsen with age. Factors that may worsen symptoms include uncontrolled comorbid conditions, chronic opiate use, inactivity, and deconditioning.

Patient education: Discourage long-term physical disability; exercise and maintaining an active daily routine helps patients avoid focusing in a nonadaptive manner on their dysfunction and symptoms.

Source: Sharon B. (Shay) Stanford, MD

New direction with medications

Pregabalin is an anticonvulsant that binds to the alpha-2-delta subunits of neurons’ voltage-gated calcium channels. This activity reduces calcium influx at nerve terminals and inhibits release of excitatory neurotransmitters, such as substance P and glutamate.18 In June 2007, pregabalin was the first medication FDA-approved for fibromyalgia.

Two placebo-controlled trials19,20 showed that pregabalin at 150 mg bid, 225 mg bid, or 300 mg bid significantly reduced weekly mean pain scores in fibromyalgia patients. Click here for details of these trials. The most common side effects—dizziness, somnolence, peripheral edema, blurred vision, and weight gain—were regarded as mild to moderate in 87% of patients.21

Although a dosage of 300 mg bid also was studied, the FDA approved pregabalin at dosages of 150 mg bid and 225 mg bid for fibromyalgia.22

Duloxetine is a serotonin/norepinephrine reuptake inhibitor (SNRI) thought to inhibit dorsal horn neurons’ response to peripheral pain signals by increasing serotonin and norepinephrine in the brain and spinal cord. This SNRI was first FDA-approved for diabetic peripheral neuropathic pain and major depressive disorder. Approval for fibromyalgia at 60 mg/d in June 2008 was based on 2 placebo-controlled, double-blind, 12-week trials comprising 874 patients.23,24Click here for detailed findings of these studies and a 6-month fixed-dose trial.25

In clinical trials, duloxetine dosages of 60 mg/d and 120 mg/d were significantly more effective than placebo. The most common side effects were nausea, constipation, excessive sweating, and somnolence.23-25

Milnacipran is an SNRI that was approved for treating fibromyalgia in January 2009 at dosages of 50 mg bid and 100 mg bid. Like other SNRIs, milnacipran is thought to work by inhibiting pain signals through increasing serotonin and norepinephrine in the brain and spinal cord. Milnacipran has a higher selectivity for norepinephrine reuptake compared with duloxetine, which may mean these medications will have different effects in different patients. Although milnacipran is approved as an antidepressant in other countries, the FDA has not approved it for treating depression in the United States.

Click here for details of a 15-week, double-blind, placebo-controlled trial of milnacipran in patients with fibromyalgia. Side effects in clinical trials were similar to those of duloxetine, with nausea, constipation, and increased sweating being most prominent.26

 

 

Other medications, such as the first-line agent amitriptyline, have shown beneficial effects in fibromyalgia but are not FDA-approved for this indication (Table 4).27-32

Choosing medications. When prescribing one of the FDA-approved medications to treat fibromyalgia, consider their benefits and side effects.

Pregabalin may be a beneficial first choice for patients who report pain and sleep as major issues. Although the medication’s label recommends starting with twice-daily dosing, patients might better tolerate an initial dose of 50 to 75 mg in the evening, with the morning dose added later. Pregabalin can be useful in patients taking multiple medications because of its renal clearance, resulting in low risk of interactions with drugs metabolized by liver enzymes. It also can be useful in patients who have not tolerated antidepressants in the past or in whom antidepressants are contraindicated.

If a patient has a history of depression or discontinuing medications because of sedating side effects, an antidepressant such as duloxetine or milnacipran may be more successful than starting with pregabalin. In general, if a patient does not respond to one of these SNRIs, moving on to the other might help. Milnacipran’s more selective effect on norepinephrine could be beneficial for some patients, especially those with excessive fatigue. Others, especially those with a high level of anxiety, might respond better to a more balanced SNRI such as duloxetine.

Table 4

Off-label medications shown to benefit patients with fibromyalgia

DrugComment
Amitriptyline27,28Considered first-line because of studies supporting its use, low cost, and wide availability; may be associated with more side effects than newer medications
Gabapentin29Possible alternative to pregabalin but may not be as well tolerated
Tramadol30May help with breakthrough pain; use with extreme caution in patients taking antidepressants because of serotonin syndrome risk
Fluoxetine31Dosages of 40 to 60 mg/d may help patients who do not tolerate SNRIs
Venlafaxine32Dosages of 150 to 225 mg/d may be an alternative to other SNRIs
SNRIs: serotonin/norepinephrine reuptake inhibitors

CASE CONTINUED: Not as hopeless

Ms. D’s primary care physician confirms your presumptive diagnosis of fibromyalgia. He prescribes a trial of amitriptyline, which she does not tolerate well because of sedation and weight gain. At her next psychiatric visit, she tells you she remains very frustrated about her physical symptoms and reports that her doctor “has given up on me.”

You discuss what a fibromyalgia diagnosis means to her and educate her about the syndrome. You refer her to a colleague who does CBT with chronic pain patients and start her on a low dose of duloxetine (30 mg once daily) to minimize side effects. You discuss possible side effects and that she may need a higher dose to notice improvement in her pain. She seems receptive to starting a graded exercise program, and you encourage her to reduce physical and emotional stress in her life.

When she returns, she reports her pain is somewhat improved and medication side effects have subsided. She is not as hopeless and tells you she is up to 10 minutes of walking daily. You increase duloxetine to 60 mg/d and reinforce her ability to exercise and manage her stress.

Related Resources

Drug Brand Names

  • Amitriptyline • Elavil, Endep
  • Cyclobenzaprine • Flexeril
  • Duloxetine • Cymbalta
  • Fluoxetine • Prozac
  • Gabapentin • Neurontin
  • Milnacipran • Savella
  • Pregabalin • Lyrica
  • Tramadol • Ultram, Ultracet
  • Venlafaxine • Effexor, Effexor XR
Disclosure

Dr. Stanford receives grant/research support from Eli Lilly and Company, Pfizer, Cypress Bioscience, and Allergan.

References

1. Wolfe F, Smythe HA, Yunus MB, et al. The American College of Rheumatology 1990 criteria for the classification of fibromyalgia. Report of the Multicenter Criteria Committee. Arthritis Rheum. 1990;33(2):160-172.

2. Aaron LA, Buchwald D. Chronic diffuse musculoskeletal pain, fibromyalgia and co-morbid unexplained clinical conditions. Best Prac Res Clin Rheumatol. 2003;17(4):563-574.

3. Arnold LM, Hudson JI, Keck PE, et al. Comorbidity of fibromyalgia and psychiatric disorders. J Clin Psychiatry. 2006;67(8):1219-1225.

4. Pope HG, Jr, Hudson JI. A supplemental interview for forms of “affective spectrum disorder.” Int J Psychiatry Med. 1991;21(3):205-232.

5. Wolfe F, Ross K, Anderson J, et al. The prevalence and characteristics of fibromyalgia in the general population. Arthritis Rheum. 1995;38(1):19-28.

6. Arnold LM, Hudson JI, Hess EV, et al. Family study of fibromyalgia. Arthritis Rheum. 2004;50(3):944-952.

7. Bondy B, Spaeth M, Offenbaecher M, et al. The T102C polymorphism of the 5-HT2A-receptor gene in fibromyalgia. Neurobiol Dis. 1999;6(5):433-439.

8. Offenbaecher M, Bondy B, de Jonge S, et al. Possible association of fibromyalgia with a polymorphism in the serotonin transporter gene regulatory region. Arthritis Rheum. 1999;42(11):2482-2488.

9. Gürsoy S, Erdal E, Herken H, et al. Significance of catechol-O-methyltransferase gene polymorphism in fibromyalgia syndrome. Rheumatol Int. 2003;23(3):104-107.

10. Russell IJ, Vaeroy H, Javors M, et al. Cerebrospinal fluid biogenic amine metabolites in fibromyalgia/fibrositis syndrome and rheumatoid arthritis. Arthritis Rheum. 1992;35(5):550-556.

11. Fields HL, Basbaum AI. In: Wall PD, Melzack R, eds. Textbook of pain. 4th ed. New York, NY: Churchill Livingstone; 1999: 309-329.

12. Clauw DJ, Crofford LJ. Chronic widespread pain and fibromyalgia: what we know, and what we need to know. Best Pract Res Clin Rheumatol. 2003;17(4):685-701.

13. Staud R, Cannon RC, Mauderli AP, et al. Temporal summation of pain from mechanical stimulation of muscle tissue in normal controls and subjects with fibromyalgia syndrome. Pain. 2003;102(1-2):87-95.

14. Russell IJ, Orr MD, Littman B, et al. Elevated cerebrospinal fluid levels of substance P in patients with the fibromyalgia syndrome. Arthritis Rheum. 1994;37(11):1593-1601.

15. Gracely RH, Petzke F, Wolf JM, et al. Functional magnetic resonance imaging evidence of augmented pain processing in fibromyalgia. Arthritis Rheum. 2002;46(5):1333-1343.

16. Williams DA, Cary MA, Groner KH. Improving physical functional status in patients with fibromyalgia: a brief cognitive behavioral intervention. J Rheumatol. 2002;29:1280-1286.

17. Busch AJ, Schachter CL, Overend TJ, et al. Exercise for fibromyalgia: a systematic review. J Rheumatol. 2008;35(6):1130-1144.

18. Field MJ, Cox PJ, Stott E. Identification of the alpha2-delta-1 subunit of voltage-dependent calcium channels as a molecular target for pain mediating the analgesic actions of pregabalin. Proc Natl Acad Sci USA. 2006;103(46):17537-17542.

19. Arnold LM, Russel IJ, Diri EW, et al. A 14-week, randomized, double-blind, placebo-controlled, monotherapy trial of pregabalin in patients with fibromyalgia. J Pain. 2008;9(9):792-805.

20. Mease PJ, Russel IJ, Arnold LM, et al. A randomized, double-blind, placebo-controlled, phase III trial of pregabalin in the treatment of patients with fibromyalgia. J Rheumatol. 2008;35(3):502-514.

21. Crofford LJ, Mease J, Simpson SL, et al. Fibromyalgia relapse evaluation and efficacy for durability of meaningful relief (FREEDOM): a 6-month, double-blind, placebo-controlled trial with pregabalin. Pain. 2008;136(3):419-431.

22. Pregabalin [package insert]. New York, NY: Pfizer, Inc.; 2004.

23. Arnold LM, Pritchett YL, D’Souza DN, et al. Duloxetine for the treatment of fibromyalgia in women: pooled results from two randomized, placebo-controlled clinical trials. J Womens Health (Larchmt). 2007;16(8):1145-1156.

24. Arnold LM, Lu Y, Crofford LJ, et al. A double-blind, multicenter trial comparing duloxetine with placebo in the treatment of fibromyalgia patients with or without major depressive disorder. Arthritis Rheum. 2004;50(9):2974-2984.

25. Russell IJ, Mease PJ, Smith TR, et al. Efficacy and safety of duloxetine for treatment of fibromyalgia in patients with or without major depressive disorder: results from a 6-month, randomized, double-blind, placebo-controlled, fixed-dose trial. Pain. 2008;136(3):432-444.

26. Clauw DJ, Mease P, Palmer RH, et al. Milnacipran for the treatment of fibromyalgia in adults: a 15-week, multicenter, randomized, double-blind, placebo-controlled, multiple-dose clinical trial. Clin Ther. 2008;30(11):1988-2004.

27. Goldenberg DL, Felson DT, Dinerman H. A randomized, controlled trial of amitriptyline and naproxen in the treatment of patients with fibromyalgia. Arthritis Rheum. 1986;29(11):1371-1377.

28. Hannonen P, Malminiemi K, Yli-Kerttula U, et al. A randomized, double-blind, placebo-controlled study of moclobemide and amitriptyline in the treatment of fibromyalgia in females without psychiatric disorder. Br J Rheumatol. 1998;37:1279-1286.

29. Arnold LM, Goldenberg DL, Stanford SB, et al. Gabapentin in the treatment of fibromyalgia: a randomized, double-blind, placebo-controlled, multicenter trial. Arthritis Rheum. 2007;56:1336-1344.

30. Bennett RM, Schein J, Kosinski MR, et al. Impact of fibromyalgia pain on health-related quality of life before and after treatment with tramadol/ acetaminophen. Arthritis Rheum. 2005;53:519-527.

31. Arnold LM, Hess EV, Hudson JI, et al. Randomized, placebo-controlled, double-blind, flexible-dose study of fluoxetine in the treatment of women with fibromyalgia. Am J Med. 2002;112:191-197.

32. Dwight MM, Arnold LM, O’Brien H, et al. An open clinical trial of venlafaxine treatment of fibromyalgia. Psychosomatics. 1998;39:14-17.

Article PDF
Author and Disclosure Information

Sharon B. (Shay) Stanford, MD
Assistant professor of psychiatry and family medicine, Assistant director, Women’s Health Research Program, University of Cincinnati College of Medicine, Cincinnati, OH

Issue
Current Psychiatry - 08(03)
Publications
Page Number
37-50
Legacy Keywords
fibromyalgia;pregabalin;duloxetine;milnacipran; somataform disorder;somatization disorder;Sharon Stanford;Sharon Shay
Sections
Author and Disclosure Information

Sharon B. (Shay) Stanford, MD
Assistant professor of psychiatry and family medicine, Assistant director, Women’s Health Research Program, University of Cincinnati College of Medicine, Cincinnati, OH

Author and Disclosure Information

Sharon B. (Shay) Stanford, MD
Assistant professor of psychiatry and family medicine, Assistant director, Women’s Health Research Program, University of Cincinnati College of Medicine, Cincinnati, OH

Article PDF
Article PDF

WEB AUDIO
Listen to Dr. Stanford discuss,
"Is fibromyalgia a somatoform disorder?

Comment on this article

Patients with fibromyalgia are a heterogeneous group, yet many describe a common experience: seeing multiple physicians who seem unable or unwilling to provide a diagnosis or treat their symptoms. This situation may be changing with the recent FDA approval of an anticonvulsant and 2 antidepressants for managing fibromyalgia symptoms.

These medications—pregabalin, duloxetine, and milnacipran—reflect a revised understanding of fibromyalgia as a CNS condition, rather than an inflammatory process in the muscles or connective tissue. As a result, psychiatrists—because of our experience with CNS phenomena and managing antidepressant and anticonvulsant medications—are likely to play a larger role in treating fibromyalgia.

CASE REPORT: ‘Just too tired’

Ms. D, age 50, has a history of migraine headaches and is referred by her primary physician for evaluation of depression and anxiety. She reports deteriorating mood over 6 months, beginning when a minor car accident left her “very sore the next day.”

“Nothing helps” the persistent pain in her back, shoulders, and thighs, which she rates as 7 to 8 on a 0-to-10 pain scale. She describes an intense ache, “like having the flu,” that worsens with activity and in stressful situations. She also experiences nausea and intermittent diarrhea, debilitating fatigue, and sleep disturbance.

Ms. D reports she is depressed because she feels “just too tired” after work to keep up with social activities or housework. Her physician’s referral notes a normal physical exam except for tenderness over her upper back and hips. Laboratory testing is negative.

Making the diagnosis

American College of Rheumatology (ACR) criteria for fibromyalgia require widespread pain for at least 3 months. “Widespread” is defined as pain in the axial skeleton, left and right sides of the body, and above and below the waist. Pain must be found in at least 11 of 18 tender point sites on digital palpation using a force of approximately 4 kg/cm2.1 For many fibromyalgia patients, however, musculoskeletal pain is not their most problematic symptom (Table 1). They may suffer:

  • migraine and tension headaches (10% to 80% of patients)
  • irritable bowel syndrome (32% to 80%)2
  • mood disorders (major depressive disorder [62%], bipolar disorder [11%])
  • anxiety disorders (panic disorder [29%], posttraumatic stress disorder [21%], social phobia [19%]).3

ACR criteria are useful in research but lack many common symptoms and comorbidities. A structured interview that follows the DSM-IV-TR format incorporates other symptoms into the diagnosis (Table 2).4

Because patients with fibromyalgia often meet criteria for somatization or somatoform disorders, how to classify them—as medically or psychiatrically ill—is controversial. Some patients believe their mood or anxiety problem stems from the difficulty they experience dealing with their physical symptoms, and if they could feel better physically they would not be depressed or anxious. Others believe their psychiatric symptoms impede their ability to help themselves feel better.

Consider fibromyalgia in any patient with widespread pain of unknown cause. Before making the diagnosis, rule out other illnesses that present with similar symptoms (Table 3). Because many patients newly diagnosed with fibromyalgia worry that something “more serious” may be going on, confirm the diagnosis with appropriate testing and physical examination, usually by a rheumatologist or primary care physician.

Table 1

Medical and cognitive symptoms related to fibromyalgia

Neurologic
Tension/migraine headache
Psychiatric
Memory and cognitive difficulties
Mood disturbance
Anxiety disorders
Ear, nose, throat
Sicca symptoms
Vasomotor rhinitis
Vestibular complaints
Cardiovascular
Neurally mediated hypotension
Mitral valve prolapse
Noncardiac chest pain
Gastrointestinal
Esophageal dysmotility
Irritable bowel syndrome
Urological
Interstitial cystitis
Gynecological
Vulvodynia
Chronic pelvic pain
Oral/dental
Temporomandibular joint syndrome
Other (general)
Chronic fatigue syndrome
Sleep disturbances
Idiopathic low back pain
Multiple chemical sensitivity
Table 2

Fibromyalgia: Structured interview for diagnosis

A. Generalized pain affecting the axial, plus upper and lower segments, plus left and rights sides of the body
Either B or C:
B. At least 11 of 18 reproducible tender points
C. At least 4 of the following symptoms:
  • Generalized fatigue
  • Headaches
  • Sleep disturbance
  • Neuropsychiatric complaints
  • Numbness, tingling sensations
  • Irritable bowel symptoms
D. It cannot be established that disturbance was due to another systematic condition
Source: Reference 4
Table 3

Differentiating fibromyalgia from illnesses with similar symptoms

IllnessTests to differentiate from primary fibromyalgia
Rheumatic diseases
Osteoarthritis
Spondyloarthropathies, rheumatoid arthritis
Systemic lupus erythematosus, polymyalgia rheumatica
Osteomalacia
Myopathy

Radiographs
Rheumatic markers (antinuclear antibody, rheumatoid factor, antibodies)
Inflammatory markers (ESR, C-reactive protein)
Vitamin D level
CPK
Neurologic
Multiple sclerosis, Chiari’s malformation, spinal stenosis, radiculopathy
Neuropathy

MRI
EMG
Endocrine
Hypothyroidism
Diabetes

TSH
Basic chemistry panel with fasting glucose
Other
Infectious
  Lyme disease
  Hepatitis
Anemia
Cancers

CBC
Lyme titer
Hepatitis antibody panel, liver function tests
Hemoglobin/hematocrit
Routine cancer screening tests, bone scan, blood chemistries specific for suspected primary cancer
ESR: erythrocyte sedimentation rate; CPK: creatine phosphokinase; EMG: electromyography; TSH: thyroid-stimulating hormone; CBC: complete blood count
 

 

CASE CONTINUED: Central pain sensitization

As you elicit more details about Ms. D’s mood, she continues to focus on her physical symptoms. She states that some days she wishes to die because her pain gets so bad, but she denies any plan or intent to harm herself. She worries that her symptoms will worsen and that she will become completely disabled.

Her primary physician attempted to relieve Ms. D’s pain with multiple trials of nonsteroidal anti-inflammatory drugs (NSAIDs) and cyclobenzaprine. She says she gained no benefit from the NSAIDs and discontinued the muscle relaxant because it made her too sleepy. Fibromyalgia affects 3.5% of women and 0.5% of men.5 It runs in families with histories of fibromyalgia and major mood and anxiety disorders, suggesting genetic links.6 Defects in genes controlling serotonin and norepinephrine have been implicated.7-9

Fibromyalgia patients show lower levels of serotonin, norepinephrine, and dopamine metabolites in cerebrospinal fluid (CSF), compared with controls.10 These neurotransmitters may inhibit descending pain pathways in the CNS, and low levels in the brain and spinal cord may inhibit CNS regulation of pain impulses from the periphery.11

Although many patients describe muscle pain, evidence suggests central pain augmentation rather than an abnormality of muscle or connective tissue.12 Some studies have found evidence of “windup,” in which second-order neurons in the spinal cord become sensitized by repeated signals from first-order neurons in the periphery, resulting in amplified and prolonged pain signals traveling to the brain.13

Levels of substance P—a primary transmitter of pain impulses—are significantly higher in CSF of fibromyalgia patients compared with controls.14 This finding, in addition to low levels of serotonin and norepinephrine, indicates that pain signals are ascending unchecked to be processed by the brain.

Neuroimaging studies confirm this observation. In a study using functional magnetic resonance imaging (fMRI), researchers applied pressure to the thumbnails of fibromyalgia patients and controls until each subject reported pain:

  • Twice as much pressure was required before controls rated their pain at a level similar to that of fibromyalgia patients.
  • When controls and fibromyalgia patients reported similar pain, a very high degree of overlap was seen in brain areas responsible for pain processing. This indicates that fibromyalgia patients and controls were experiencing the pain they reported in the same way.15

Treating the whole patient

As a clinician who specializes in fibromyalgia, I counteract my patients’ and my own frustration with this condition by structuring office visits, determining realistic treatment goals, and treating all symptoms as part of a common syndrome rather than individual illnesses.

Structure office visits. Before every visit, have patients rate each symptom domain and write their top 2 or 3 concerns for that day (Click here for a sample form). Focusing on the patient’s most troublesome symptoms can help both of you feel greater satisfaction with treatment.

Educate patients. Ask them to discuss their beliefs about fibromyalgia; many know others with this condition or have researched diagnosis and treatment. Before developing a treatment plan, explain that their symptoms are chronic and all part of the same syndrome. Describe their pain as a complex phenomenon with possible peripheral and CNS components. Guide them to reputable Web sites and resources (see Related Resources).

Set realistic expectations. Many patients expect to resume an energetic and pain-free life, which usually is not the case with fibromyalgia (Box). Most medications are considered successful if they reduce pain by 30% to 50%, and side effects can be problematic. Discuss side effects before treatment begins to reduce patients’ anxiety and improve compliance in the first weeks.

Cognitive-behavioral therapy (CBT) for fibromyalgia incorporates relaxation techniques, helping patients view symptoms as manageable, reinforcing adaptive coping skills, and teaching them how to monitor thoughts, feelings, and behavior to change the view that they are helpless victims. A modest course of 6 weekly group CBT sessions significantly improved physical functioning in 25% of fibromyalgia patients (n=76) compared with 12% in a standard-care group (n=69), even though patients’ pain severity did not improve.16

Recommend exercise, lifestyle changes. Aerobic exercise can significantly improve well-being and physical functioning in fibromyalgia patients.17 Low-impact aerobics, such as done in warm water, usually are well tolerated, although any low-impact exercise can help. Because fibromyalgia symptoms often increase with physical activity, counsel patients to begin with a few minutes daily and increase very slowly each week.
 

 


Lifestyle changes are as important as medications in controlling fibromyalgia symptoms. In addition to exercise, recommend that patients:

  • follow a daily routine
  • pace activity to avoid exacerbating symptoms
  • reduce stress.
Sometimes, I use the analogy of diabetes: treating fibromyalgia with medication but without changing lifestyle is like prescribing medication for a diabetic patient without changing diet. Follow up on this “homework” at each visit to reinforce that patients helping themselves is an important part of treatment.

Box

Managing unrealistic expectations of fibromyalgia patients

BELIEF: ‘A magic pill exists that will resolve all my symptoms and have no side effects’

Clinical evidence: Most medications that have been studied were effective in 30% to 50% of patients and reduced pain scores by 30% to 50%.

Patient education: Explain to the patient with a pain rating of 7 at the first visit that achieving a pain level of 3 to 4 may be possible with treatment. Even with successful treatment, symptoms may flare intermittently. As with any treatment, adverse effects may occur. Discuss these, so the patient is not surprised.

BELIEF: ‘I can’t exercise’

Clinical evidence: Most patients experience more fatigue and pain with physical activity, but exercise is important to maintain physical function.

Patient education: When discussing an exercise program, focus on what the patient can do. Most patients attempt too much, too soon; advise them to start at a tolerable level (such as 2 to 3 minutes of aerobic activity daily for the first week) and gradually increase as tolerated.

BELIEF: ‘You (the psychiatrist) can make me feel better’

Clinical evidence: Psychiatrists can help by prescribing appropriate medications, but much of the burden falls on the patient to maintain a healthy, active lifestyle and to manage stressors in an adaptive manner.

Patient education: A fibromyalgia patient may find relief with a medication, but symptoms may flare if they ‘overdo’ and take on too many physical or emotional stressors. A consistent, healthy routine is ideal.

BELIEF: ‘I will eventually become disabled by fibromyalgia’

Clinical evidence: Despite little long-term research on fibromyalgia patients, most evidence points to a chronic, fluctuating syndrome that does not worsen with age. Factors that may worsen symptoms include uncontrolled comorbid conditions, chronic opiate use, inactivity, and deconditioning.

Patient education: Discourage long-term physical disability; exercise and maintaining an active daily routine helps patients avoid focusing in a nonadaptive manner on their dysfunction and symptoms.

Source: Sharon B. (Shay) Stanford, MD

New direction with medications

Pregabalin is an anticonvulsant that binds to the alpha-2-delta subunits of neurons’ voltage-gated calcium channels. This activity reduces calcium influx at nerve terminals and inhibits release of excitatory neurotransmitters, such as substance P and glutamate.18 In June 2007, pregabalin was the first medication FDA-approved for fibromyalgia.

Two placebo-controlled trials19,20 showed that pregabalin at 150 mg bid, 225 mg bid, or 300 mg bid significantly reduced weekly mean pain scores in fibromyalgia patients. Click here for details of these trials. The most common side effects—dizziness, somnolence, peripheral edema, blurred vision, and weight gain—were regarded as mild to moderate in 87% of patients.21

Although a dosage of 300 mg bid also was studied, the FDA approved pregabalin at dosages of 150 mg bid and 225 mg bid for fibromyalgia.22

Duloxetine is a serotonin/norepinephrine reuptake inhibitor (SNRI) thought to inhibit dorsal horn neurons’ response to peripheral pain signals by increasing serotonin and norepinephrine in the brain and spinal cord. This SNRI was first FDA-approved for diabetic peripheral neuropathic pain and major depressive disorder. Approval for fibromyalgia at 60 mg/d in June 2008 was based on 2 placebo-controlled, double-blind, 12-week trials comprising 874 patients.23,24Click here for detailed findings of these studies and a 6-month fixed-dose trial.25

In clinical trials, duloxetine dosages of 60 mg/d and 120 mg/d were significantly more effective than placebo. The most common side effects were nausea, constipation, excessive sweating, and somnolence.23-25

Milnacipran is an SNRI that was approved for treating fibromyalgia in January 2009 at dosages of 50 mg bid and 100 mg bid. Like other SNRIs, milnacipran is thought to work by inhibiting pain signals through increasing serotonin and norepinephrine in the brain and spinal cord. Milnacipran has a higher selectivity for norepinephrine reuptake compared with duloxetine, which may mean these medications will have different effects in different patients. Although milnacipran is approved as an antidepressant in other countries, the FDA has not approved it for treating depression in the United States.

Click here for details of a 15-week, double-blind, placebo-controlled trial of milnacipran in patients with fibromyalgia. Side effects in clinical trials were similar to those of duloxetine, with nausea, constipation, and increased sweating being most prominent.26

 

 

Other medications, such as the first-line agent amitriptyline, have shown beneficial effects in fibromyalgia but are not FDA-approved for this indication (Table 4).27-32

Choosing medications. When prescribing one of the FDA-approved medications to treat fibromyalgia, consider their benefits and side effects.

Pregabalin may be a beneficial first choice for patients who report pain and sleep as major issues. Although the medication’s label recommends starting with twice-daily dosing, patients might better tolerate an initial dose of 50 to 75 mg in the evening, with the morning dose added later. Pregabalin can be useful in patients taking multiple medications because of its renal clearance, resulting in low risk of interactions with drugs metabolized by liver enzymes. It also can be useful in patients who have not tolerated antidepressants in the past or in whom antidepressants are contraindicated.

If a patient has a history of depression or discontinuing medications because of sedating side effects, an antidepressant such as duloxetine or milnacipran may be more successful than starting with pregabalin. In general, if a patient does not respond to one of these SNRIs, moving on to the other might help. Milnacipran’s more selective effect on norepinephrine could be beneficial for some patients, especially those with excessive fatigue. Others, especially those with a high level of anxiety, might respond better to a more balanced SNRI such as duloxetine.

Table 4

Off-label medications shown to benefit patients with fibromyalgia

DrugComment
Amitriptyline27,28Considered first-line because of studies supporting its use, low cost, and wide availability; may be associated with more side effects than newer medications
Gabapentin29Possible alternative to pregabalin but may not be as well tolerated
Tramadol30May help with breakthrough pain; use with extreme caution in patients taking antidepressants because of serotonin syndrome risk
Fluoxetine31Dosages of 40 to 60 mg/d may help patients who do not tolerate SNRIs
Venlafaxine32Dosages of 150 to 225 mg/d may be an alternative to other SNRIs
SNRIs: serotonin/norepinephrine reuptake inhibitors

CASE CONTINUED: Not as hopeless

Ms. D’s primary care physician confirms your presumptive diagnosis of fibromyalgia. He prescribes a trial of amitriptyline, which she does not tolerate well because of sedation and weight gain. At her next psychiatric visit, she tells you she remains very frustrated about her physical symptoms and reports that her doctor “has given up on me.”

You discuss what a fibromyalgia diagnosis means to her and educate her about the syndrome. You refer her to a colleague who does CBT with chronic pain patients and start her on a low dose of duloxetine (30 mg once daily) to minimize side effects. You discuss possible side effects and that she may need a higher dose to notice improvement in her pain. She seems receptive to starting a graded exercise program, and you encourage her to reduce physical and emotional stress in her life.

When she returns, she reports her pain is somewhat improved and medication side effects have subsided. She is not as hopeless and tells you she is up to 10 minutes of walking daily. You increase duloxetine to 60 mg/d and reinforce her ability to exercise and manage her stress.

Related Resources

Drug Brand Names

  • Amitriptyline • Elavil, Endep
  • Cyclobenzaprine • Flexeril
  • Duloxetine • Cymbalta
  • Fluoxetine • Prozac
  • Gabapentin • Neurontin
  • Milnacipran • Savella
  • Pregabalin • Lyrica
  • Tramadol • Ultram, Ultracet
  • Venlafaxine • Effexor, Effexor XR
Disclosure

Dr. Stanford receives grant/research support from Eli Lilly and Company, Pfizer, Cypress Bioscience, and Allergan.

WEB AUDIO
Listen to Dr. Stanford discuss,
"Is fibromyalgia a somatoform disorder?

Comment on this article

Patients with fibromyalgia are a heterogeneous group, yet many describe a common experience: seeing multiple physicians who seem unable or unwilling to provide a diagnosis or treat their symptoms. This situation may be changing with the recent FDA approval of an anticonvulsant and 2 antidepressants for managing fibromyalgia symptoms.

These medications—pregabalin, duloxetine, and milnacipran—reflect a revised understanding of fibromyalgia as a CNS condition, rather than an inflammatory process in the muscles or connective tissue. As a result, psychiatrists—because of our experience with CNS phenomena and managing antidepressant and anticonvulsant medications—are likely to play a larger role in treating fibromyalgia.

CASE REPORT: ‘Just too tired’

Ms. D, age 50, has a history of migraine headaches and is referred by her primary physician for evaluation of depression and anxiety. She reports deteriorating mood over 6 months, beginning when a minor car accident left her “very sore the next day.”

“Nothing helps” the persistent pain in her back, shoulders, and thighs, which she rates as 7 to 8 on a 0-to-10 pain scale. She describes an intense ache, “like having the flu,” that worsens with activity and in stressful situations. She also experiences nausea and intermittent diarrhea, debilitating fatigue, and sleep disturbance.

Ms. D reports she is depressed because she feels “just too tired” after work to keep up with social activities or housework. Her physician’s referral notes a normal physical exam except for tenderness over her upper back and hips. Laboratory testing is negative.

Making the diagnosis

American College of Rheumatology (ACR) criteria for fibromyalgia require widespread pain for at least 3 months. “Widespread” is defined as pain in the axial skeleton, left and right sides of the body, and above and below the waist. Pain must be found in at least 11 of 18 tender point sites on digital palpation using a force of approximately 4 kg/cm2.1 For many fibromyalgia patients, however, musculoskeletal pain is not their most problematic symptom (Table 1). They may suffer:

  • migraine and tension headaches (10% to 80% of patients)
  • irritable bowel syndrome (32% to 80%)2
  • mood disorders (major depressive disorder [62%], bipolar disorder [11%])
  • anxiety disorders (panic disorder [29%], posttraumatic stress disorder [21%], social phobia [19%]).3

ACR criteria are useful in research but lack many common symptoms and comorbidities. A structured interview that follows the DSM-IV-TR format incorporates other symptoms into the diagnosis (Table 2).4

Because patients with fibromyalgia often meet criteria for somatization or somatoform disorders, how to classify them—as medically or psychiatrically ill—is controversial. Some patients believe their mood or anxiety problem stems from the difficulty they experience dealing with their physical symptoms, and if they could feel better physically they would not be depressed or anxious. Others believe their psychiatric symptoms impede their ability to help themselves feel better.

Consider fibromyalgia in any patient with widespread pain of unknown cause. Before making the diagnosis, rule out other illnesses that present with similar symptoms (Table 3). Because many patients newly diagnosed with fibromyalgia worry that something “more serious” may be going on, confirm the diagnosis with appropriate testing and physical examination, usually by a rheumatologist or primary care physician.

Table 1

Medical and cognitive symptoms related to fibromyalgia

Neurologic
Tension/migraine headache
Psychiatric
Memory and cognitive difficulties
Mood disturbance
Anxiety disorders
Ear, nose, throat
Sicca symptoms
Vasomotor rhinitis
Vestibular complaints
Cardiovascular
Neurally mediated hypotension
Mitral valve prolapse
Noncardiac chest pain
Gastrointestinal
Esophageal dysmotility
Irritable bowel syndrome
Urological
Interstitial cystitis
Gynecological
Vulvodynia
Chronic pelvic pain
Oral/dental
Temporomandibular joint syndrome
Other (general)
Chronic fatigue syndrome
Sleep disturbances
Idiopathic low back pain
Multiple chemical sensitivity
Table 2

Fibromyalgia: Structured interview for diagnosis

A. Generalized pain affecting the axial, plus upper and lower segments, plus left and rights sides of the body
Either B or C:
B. At least 11 of 18 reproducible tender points
C. At least 4 of the following symptoms:
  • Generalized fatigue
  • Headaches
  • Sleep disturbance
  • Neuropsychiatric complaints
  • Numbness, tingling sensations
  • Irritable bowel symptoms
D. It cannot be established that disturbance was due to another systematic condition
Source: Reference 4
Table 3

Differentiating fibromyalgia from illnesses with similar symptoms

IllnessTests to differentiate from primary fibromyalgia
Rheumatic diseases
Osteoarthritis
Spondyloarthropathies, rheumatoid arthritis
Systemic lupus erythematosus, polymyalgia rheumatica
Osteomalacia
Myopathy

Radiographs
Rheumatic markers (antinuclear antibody, rheumatoid factor, antibodies)
Inflammatory markers (ESR, C-reactive protein)
Vitamin D level
CPK
Neurologic
Multiple sclerosis, Chiari’s malformation, spinal stenosis, radiculopathy
Neuropathy

MRI
EMG
Endocrine
Hypothyroidism
Diabetes

TSH
Basic chemistry panel with fasting glucose
Other
Infectious
  Lyme disease
  Hepatitis
Anemia
Cancers

CBC
Lyme titer
Hepatitis antibody panel, liver function tests
Hemoglobin/hematocrit
Routine cancer screening tests, bone scan, blood chemistries specific for suspected primary cancer
ESR: erythrocyte sedimentation rate; CPK: creatine phosphokinase; EMG: electromyography; TSH: thyroid-stimulating hormone; CBC: complete blood count
 

 

CASE CONTINUED: Central pain sensitization

As you elicit more details about Ms. D’s mood, she continues to focus on her physical symptoms. She states that some days she wishes to die because her pain gets so bad, but she denies any plan or intent to harm herself. She worries that her symptoms will worsen and that she will become completely disabled.

Her primary physician attempted to relieve Ms. D’s pain with multiple trials of nonsteroidal anti-inflammatory drugs (NSAIDs) and cyclobenzaprine. She says she gained no benefit from the NSAIDs and discontinued the muscle relaxant because it made her too sleepy. Fibromyalgia affects 3.5% of women and 0.5% of men.5 It runs in families with histories of fibromyalgia and major mood and anxiety disorders, suggesting genetic links.6 Defects in genes controlling serotonin and norepinephrine have been implicated.7-9

Fibromyalgia patients show lower levels of serotonin, norepinephrine, and dopamine metabolites in cerebrospinal fluid (CSF), compared with controls.10 These neurotransmitters may inhibit descending pain pathways in the CNS, and low levels in the brain and spinal cord may inhibit CNS regulation of pain impulses from the periphery.11

Although many patients describe muscle pain, evidence suggests central pain augmentation rather than an abnormality of muscle or connective tissue.12 Some studies have found evidence of “windup,” in which second-order neurons in the spinal cord become sensitized by repeated signals from first-order neurons in the periphery, resulting in amplified and prolonged pain signals traveling to the brain.13

Levels of substance P—a primary transmitter of pain impulses—are significantly higher in CSF of fibromyalgia patients compared with controls.14 This finding, in addition to low levels of serotonin and norepinephrine, indicates that pain signals are ascending unchecked to be processed by the brain.

Neuroimaging studies confirm this observation. In a study using functional magnetic resonance imaging (fMRI), researchers applied pressure to the thumbnails of fibromyalgia patients and controls until each subject reported pain:

  • Twice as much pressure was required before controls rated their pain at a level similar to that of fibromyalgia patients.
  • When controls and fibromyalgia patients reported similar pain, a very high degree of overlap was seen in brain areas responsible for pain processing. This indicates that fibromyalgia patients and controls were experiencing the pain they reported in the same way.15

Treating the whole patient

As a clinician who specializes in fibromyalgia, I counteract my patients’ and my own frustration with this condition by structuring office visits, determining realistic treatment goals, and treating all symptoms as part of a common syndrome rather than individual illnesses.

Structure office visits. Before every visit, have patients rate each symptom domain and write their top 2 or 3 concerns for that day (Click here for a sample form). Focusing on the patient’s most troublesome symptoms can help both of you feel greater satisfaction with treatment.

Educate patients. Ask them to discuss their beliefs about fibromyalgia; many know others with this condition or have researched diagnosis and treatment. Before developing a treatment plan, explain that their symptoms are chronic and all part of the same syndrome. Describe their pain as a complex phenomenon with possible peripheral and CNS components. Guide them to reputable Web sites and resources (see Related Resources).

Set realistic expectations. Many patients expect to resume an energetic and pain-free life, which usually is not the case with fibromyalgia (Box). Most medications are considered successful if they reduce pain by 30% to 50%, and side effects can be problematic. Discuss side effects before treatment begins to reduce patients’ anxiety and improve compliance in the first weeks.

Cognitive-behavioral therapy (CBT) for fibromyalgia incorporates relaxation techniques, helping patients view symptoms as manageable, reinforcing adaptive coping skills, and teaching them how to monitor thoughts, feelings, and behavior to change the view that they are helpless victims. A modest course of 6 weekly group CBT sessions significantly improved physical functioning in 25% of fibromyalgia patients (n=76) compared with 12% in a standard-care group (n=69), even though patients’ pain severity did not improve.16

Recommend exercise, lifestyle changes. Aerobic exercise can significantly improve well-being and physical functioning in fibromyalgia patients.17 Low-impact aerobics, such as done in warm water, usually are well tolerated, although any low-impact exercise can help. Because fibromyalgia symptoms often increase with physical activity, counsel patients to begin with a few minutes daily and increase very slowly each week.
 

 


Lifestyle changes are as important as medications in controlling fibromyalgia symptoms. In addition to exercise, recommend that patients:

  • follow a daily routine
  • pace activity to avoid exacerbating symptoms
  • reduce stress.
Sometimes, I use the analogy of diabetes: treating fibromyalgia with medication but without changing lifestyle is like prescribing medication for a diabetic patient without changing diet. Follow up on this “homework” at each visit to reinforce that patients helping themselves is an important part of treatment.

Box

Managing unrealistic expectations of fibromyalgia patients

BELIEF: ‘A magic pill exists that will resolve all my symptoms and have no side effects’

Clinical evidence: Most medications that have been studied were effective in 30% to 50% of patients and reduced pain scores by 30% to 50%.

Patient education: Explain to the patient with a pain rating of 7 at the first visit that achieving a pain level of 3 to 4 may be possible with treatment. Even with successful treatment, symptoms may flare intermittently. As with any treatment, adverse effects may occur. Discuss these, so the patient is not surprised.

BELIEF: ‘I can’t exercise’

Clinical evidence: Most patients experience more fatigue and pain with physical activity, but exercise is important to maintain physical function.

Patient education: When discussing an exercise program, focus on what the patient can do. Most patients attempt too much, too soon; advise them to start at a tolerable level (such as 2 to 3 minutes of aerobic activity daily for the first week) and gradually increase as tolerated.

BELIEF: ‘You (the psychiatrist) can make me feel better’

Clinical evidence: Psychiatrists can help by prescribing appropriate medications, but much of the burden falls on the patient to maintain a healthy, active lifestyle and to manage stressors in an adaptive manner.

Patient education: A fibromyalgia patient may find relief with a medication, but symptoms may flare if they ‘overdo’ and take on too many physical or emotional stressors. A consistent, healthy routine is ideal.

BELIEF: ‘I will eventually become disabled by fibromyalgia’

Clinical evidence: Despite little long-term research on fibromyalgia patients, most evidence points to a chronic, fluctuating syndrome that does not worsen with age. Factors that may worsen symptoms include uncontrolled comorbid conditions, chronic opiate use, inactivity, and deconditioning.

Patient education: Discourage long-term physical disability; exercise and maintaining an active daily routine helps patients avoid focusing in a nonadaptive manner on their dysfunction and symptoms.

Source: Sharon B. (Shay) Stanford, MD

New direction with medications

Pregabalin is an anticonvulsant that binds to the alpha-2-delta subunits of neurons’ voltage-gated calcium channels. This activity reduces calcium influx at nerve terminals and inhibits release of excitatory neurotransmitters, such as substance P and glutamate.18 In June 2007, pregabalin was the first medication FDA-approved for fibromyalgia.

Two placebo-controlled trials19,20 showed that pregabalin at 150 mg bid, 225 mg bid, or 300 mg bid significantly reduced weekly mean pain scores in fibromyalgia patients. Click here for details of these trials. The most common side effects—dizziness, somnolence, peripheral edema, blurred vision, and weight gain—were regarded as mild to moderate in 87% of patients.21

Although a dosage of 300 mg bid also was studied, the FDA approved pregabalin at dosages of 150 mg bid and 225 mg bid for fibromyalgia.22

Duloxetine is a serotonin/norepinephrine reuptake inhibitor (SNRI) thought to inhibit dorsal horn neurons’ response to peripheral pain signals by increasing serotonin and norepinephrine in the brain and spinal cord. This SNRI was first FDA-approved for diabetic peripheral neuropathic pain and major depressive disorder. Approval for fibromyalgia at 60 mg/d in June 2008 was based on 2 placebo-controlled, double-blind, 12-week trials comprising 874 patients.23,24Click here for detailed findings of these studies and a 6-month fixed-dose trial.25

In clinical trials, duloxetine dosages of 60 mg/d and 120 mg/d were significantly more effective than placebo. The most common side effects were nausea, constipation, excessive sweating, and somnolence.23-25

Milnacipran is an SNRI that was approved for treating fibromyalgia in January 2009 at dosages of 50 mg bid and 100 mg bid. Like other SNRIs, milnacipran is thought to work by inhibiting pain signals through increasing serotonin and norepinephrine in the brain and spinal cord. Milnacipran has a higher selectivity for norepinephrine reuptake compared with duloxetine, which may mean these medications will have different effects in different patients. Although milnacipran is approved as an antidepressant in other countries, the FDA has not approved it for treating depression in the United States.

Click here for details of a 15-week, double-blind, placebo-controlled trial of milnacipran in patients with fibromyalgia. Side effects in clinical trials were similar to those of duloxetine, with nausea, constipation, and increased sweating being most prominent.26

 

 

Other medications, such as the first-line agent amitriptyline, have shown beneficial effects in fibromyalgia but are not FDA-approved for this indication (Table 4).27-32

Choosing medications. When prescribing one of the FDA-approved medications to treat fibromyalgia, consider their benefits and side effects.

Pregabalin may be a beneficial first choice for patients who report pain and sleep as major issues. Although the medication’s label recommends starting with twice-daily dosing, patients might better tolerate an initial dose of 50 to 75 mg in the evening, with the morning dose added later. Pregabalin can be useful in patients taking multiple medications because of its renal clearance, resulting in low risk of interactions with drugs metabolized by liver enzymes. It also can be useful in patients who have not tolerated antidepressants in the past or in whom antidepressants are contraindicated.

If a patient has a history of depression or discontinuing medications because of sedating side effects, an antidepressant such as duloxetine or milnacipran may be more successful than starting with pregabalin. In general, if a patient does not respond to one of these SNRIs, moving on to the other might help. Milnacipran’s more selective effect on norepinephrine could be beneficial for some patients, especially those with excessive fatigue. Others, especially those with a high level of anxiety, might respond better to a more balanced SNRI such as duloxetine.

Table 4

Off-label medications shown to benefit patients with fibromyalgia

DrugComment
Amitriptyline27,28Considered first-line because of studies supporting its use, low cost, and wide availability; may be associated with more side effects than newer medications
Gabapentin29Possible alternative to pregabalin but may not be as well tolerated
Tramadol30May help with breakthrough pain; use with extreme caution in patients taking antidepressants because of serotonin syndrome risk
Fluoxetine31Dosages of 40 to 60 mg/d may help patients who do not tolerate SNRIs
Venlafaxine32Dosages of 150 to 225 mg/d may be an alternative to other SNRIs
SNRIs: serotonin/norepinephrine reuptake inhibitors

CASE CONTINUED: Not as hopeless

Ms. D’s primary care physician confirms your presumptive diagnosis of fibromyalgia. He prescribes a trial of amitriptyline, which she does not tolerate well because of sedation and weight gain. At her next psychiatric visit, she tells you she remains very frustrated about her physical symptoms and reports that her doctor “has given up on me.”

You discuss what a fibromyalgia diagnosis means to her and educate her about the syndrome. You refer her to a colleague who does CBT with chronic pain patients and start her on a low dose of duloxetine (30 mg once daily) to minimize side effects. You discuss possible side effects and that she may need a higher dose to notice improvement in her pain. She seems receptive to starting a graded exercise program, and you encourage her to reduce physical and emotional stress in her life.

When she returns, she reports her pain is somewhat improved and medication side effects have subsided. She is not as hopeless and tells you she is up to 10 minutes of walking daily. You increase duloxetine to 60 mg/d and reinforce her ability to exercise and manage her stress.

Related Resources

Drug Brand Names

  • Amitriptyline • Elavil, Endep
  • Cyclobenzaprine • Flexeril
  • Duloxetine • Cymbalta
  • Fluoxetine • Prozac
  • Gabapentin • Neurontin
  • Milnacipran • Savella
  • Pregabalin • Lyrica
  • Tramadol • Ultram, Ultracet
  • Venlafaxine • Effexor, Effexor XR
Disclosure

Dr. Stanford receives grant/research support from Eli Lilly and Company, Pfizer, Cypress Bioscience, and Allergan.

References

1. Wolfe F, Smythe HA, Yunus MB, et al. The American College of Rheumatology 1990 criteria for the classification of fibromyalgia. Report of the Multicenter Criteria Committee. Arthritis Rheum. 1990;33(2):160-172.

2. Aaron LA, Buchwald D. Chronic diffuse musculoskeletal pain, fibromyalgia and co-morbid unexplained clinical conditions. Best Prac Res Clin Rheumatol. 2003;17(4):563-574.

3. Arnold LM, Hudson JI, Keck PE, et al. Comorbidity of fibromyalgia and psychiatric disorders. J Clin Psychiatry. 2006;67(8):1219-1225.

4. Pope HG, Jr, Hudson JI. A supplemental interview for forms of “affective spectrum disorder.” Int J Psychiatry Med. 1991;21(3):205-232.

5. Wolfe F, Ross K, Anderson J, et al. The prevalence and characteristics of fibromyalgia in the general population. Arthritis Rheum. 1995;38(1):19-28.

6. Arnold LM, Hudson JI, Hess EV, et al. Family study of fibromyalgia. Arthritis Rheum. 2004;50(3):944-952.

7. Bondy B, Spaeth M, Offenbaecher M, et al. The T102C polymorphism of the 5-HT2A-receptor gene in fibromyalgia. Neurobiol Dis. 1999;6(5):433-439.

8. Offenbaecher M, Bondy B, de Jonge S, et al. Possible association of fibromyalgia with a polymorphism in the serotonin transporter gene regulatory region. Arthritis Rheum. 1999;42(11):2482-2488.

9. Gürsoy S, Erdal E, Herken H, et al. Significance of catechol-O-methyltransferase gene polymorphism in fibromyalgia syndrome. Rheumatol Int. 2003;23(3):104-107.

10. Russell IJ, Vaeroy H, Javors M, et al. Cerebrospinal fluid biogenic amine metabolites in fibromyalgia/fibrositis syndrome and rheumatoid arthritis. Arthritis Rheum. 1992;35(5):550-556.

11. Fields HL, Basbaum AI. In: Wall PD, Melzack R, eds. Textbook of pain. 4th ed. New York, NY: Churchill Livingstone; 1999: 309-329.

12. Clauw DJ, Crofford LJ. Chronic widespread pain and fibromyalgia: what we know, and what we need to know. Best Pract Res Clin Rheumatol. 2003;17(4):685-701.

13. Staud R, Cannon RC, Mauderli AP, et al. Temporal summation of pain from mechanical stimulation of muscle tissue in normal controls and subjects with fibromyalgia syndrome. Pain. 2003;102(1-2):87-95.

14. Russell IJ, Orr MD, Littman B, et al. Elevated cerebrospinal fluid levels of substance P in patients with the fibromyalgia syndrome. Arthritis Rheum. 1994;37(11):1593-1601.

15. Gracely RH, Petzke F, Wolf JM, et al. Functional magnetic resonance imaging evidence of augmented pain processing in fibromyalgia. Arthritis Rheum. 2002;46(5):1333-1343.

16. Williams DA, Cary MA, Groner KH. Improving physical functional status in patients with fibromyalgia: a brief cognitive behavioral intervention. J Rheumatol. 2002;29:1280-1286.

17. Busch AJ, Schachter CL, Overend TJ, et al. Exercise for fibromyalgia: a systematic review. J Rheumatol. 2008;35(6):1130-1144.

18. Field MJ, Cox PJ, Stott E. Identification of the alpha2-delta-1 subunit of voltage-dependent calcium channels as a molecular target for pain mediating the analgesic actions of pregabalin. Proc Natl Acad Sci USA. 2006;103(46):17537-17542.

19. Arnold LM, Russel IJ, Diri EW, et al. A 14-week, randomized, double-blind, placebo-controlled, monotherapy trial of pregabalin in patients with fibromyalgia. J Pain. 2008;9(9):792-805.

20. Mease PJ, Russel IJ, Arnold LM, et al. A randomized, double-blind, placebo-controlled, phase III trial of pregabalin in the treatment of patients with fibromyalgia. J Rheumatol. 2008;35(3):502-514.

21. Crofford LJ, Mease J, Simpson SL, et al. Fibromyalgia relapse evaluation and efficacy for durability of meaningful relief (FREEDOM): a 6-month, double-blind, placebo-controlled trial with pregabalin. Pain. 2008;136(3):419-431.

22. Pregabalin [package insert]. New York, NY: Pfizer, Inc.; 2004.

23. Arnold LM, Pritchett YL, D’Souza DN, et al. Duloxetine for the treatment of fibromyalgia in women: pooled results from two randomized, placebo-controlled clinical trials. J Womens Health (Larchmt). 2007;16(8):1145-1156.

24. Arnold LM, Lu Y, Crofford LJ, et al. A double-blind, multicenter trial comparing duloxetine with placebo in the treatment of fibromyalgia patients with or without major depressive disorder. Arthritis Rheum. 2004;50(9):2974-2984.

25. Russell IJ, Mease PJ, Smith TR, et al. Efficacy and safety of duloxetine for treatment of fibromyalgia in patients with or without major depressive disorder: results from a 6-month, randomized, double-blind, placebo-controlled, fixed-dose trial. Pain. 2008;136(3):432-444.

26. Clauw DJ, Mease P, Palmer RH, et al. Milnacipran for the treatment of fibromyalgia in adults: a 15-week, multicenter, randomized, double-blind, placebo-controlled, multiple-dose clinical trial. Clin Ther. 2008;30(11):1988-2004.

27. Goldenberg DL, Felson DT, Dinerman H. A randomized, controlled trial of amitriptyline and naproxen in the treatment of patients with fibromyalgia. Arthritis Rheum. 1986;29(11):1371-1377.

28. Hannonen P, Malminiemi K, Yli-Kerttula U, et al. A randomized, double-blind, placebo-controlled study of moclobemide and amitriptyline in the treatment of fibromyalgia in females without psychiatric disorder. Br J Rheumatol. 1998;37:1279-1286.

29. Arnold LM, Goldenberg DL, Stanford SB, et al. Gabapentin in the treatment of fibromyalgia: a randomized, double-blind, placebo-controlled, multicenter trial. Arthritis Rheum. 2007;56:1336-1344.

30. Bennett RM, Schein J, Kosinski MR, et al. Impact of fibromyalgia pain on health-related quality of life before and after treatment with tramadol/ acetaminophen. Arthritis Rheum. 2005;53:519-527.

31. Arnold LM, Hess EV, Hudson JI, et al. Randomized, placebo-controlled, double-blind, flexible-dose study of fluoxetine in the treatment of women with fibromyalgia. Am J Med. 2002;112:191-197.

32. Dwight MM, Arnold LM, O’Brien H, et al. An open clinical trial of venlafaxine treatment of fibromyalgia. Psychosomatics. 1998;39:14-17.

References

1. Wolfe F, Smythe HA, Yunus MB, et al. The American College of Rheumatology 1990 criteria for the classification of fibromyalgia. Report of the Multicenter Criteria Committee. Arthritis Rheum. 1990;33(2):160-172.

2. Aaron LA, Buchwald D. Chronic diffuse musculoskeletal pain, fibromyalgia and co-morbid unexplained clinical conditions. Best Prac Res Clin Rheumatol. 2003;17(4):563-574.

3. Arnold LM, Hudson JI, Keck PE, et al. Comorbidity of fibromyalgia and psychiatric disorders. J Clin Psychiatry. 2006;67(8):1219-1225.

4. Pope HG, Jr, Hudson JI. A supplemental interview for forms of “affective spectrum disorder.” Int J Psychiatry Med. 1991;21(3):205-232.

5. Wolfe F, Ross K, Anderson J, et al. The prevalence and characteristics of fibromyalgia in the general population. Arthritis Rheum. 1995;38(1):19-28.

6. Arnold LM, Hudson JI, Hess EV, et al. Family study of fibromyalgia. Arthritis Rheum. 2004;50(3):944-952.

7. Bondy B, Spaeth M, Offenbaecher M, et al. The T102C polymorphism of the 5-HT2A-receptor gene in fibromyalgia. Neurobiol Dis. 1999;6(5):433-439.

8. Offenbaecher M, Bondy B, de Jonge S, et al. Possible association of fibromyalgia with a polymorphism in the serotonin transporter gene regulatory region. Arthritis Rheum. 1999;42(11):2482-2488.

9. Gürsoy S, Erdal E, Herken H, et al. Significance of catechol-O-methyltransferase gene polymorphism in fibromyalgia syndrome. Rheumatol Int. 2003;23(3):104-107.

10. Russell IJ, Vaeroy H, Javors M, et al. Cerebrospinal fluid biogenic amine metabolites in fibromyalgia/fibrositis syndrome and rheumatoid arthritis. Arthritis Rheum. 1992;35(5):550-556.

11. Fields HL, Basbaum AI. In: Wall PD, Melzack R, eds. Textbook of pain. 4th ed. New York, NY: Churchill Livingstone; 1999: 309-329.

12. Clauw DJ, Crofford LJ. Chronic widespread pain and fibromyalgia: what we know, and what we need to know. Best Pract Res Clin Rheumatol. 2003;17(4):685-701.

13. Staud R, Cannon RC, Mauderli AP, et al. Temporal summation of pain from mechanical stimulation of muscle tissue in normal controls and subjects with fibromyalgia syndrome. Pain. 2003;102(1-2):87-95.

14. Russell IJ, Orr MD, Littman B, et al. Elevated cerebrospinal fluid levels of substance P in patients with the fibromyalgia syndrome. Arthritis Rheum. 1994;37(11):1593-1601.

15. Gracely RH, Petzke F, Wolf JM, et al. Functional magnetic resonance imaging evidence of augmented pain processing in fibromyalgia. Arthritis Rheum. 2002;46(5):1333-1343.

16. Williams DA, Cary MA, Groner KH. Improving physical functional status in patients with fibromyalgia: a brief cognitive behavioral intervention. J Rheumatol. 2002;29:1280-1286.

17. Busch AJ, Schachter CL, Overend TJ, et al. Exercise for fibromyalgia: a systematic review. J Rheumatol. 2008;35(6):1130-1144.

18. Field MJ, Cox PJ, Stott E. Identification of the alpha2-delta-1 subunit of voltage-dependent calcium channels as a molecular target for pain mediating the analgesic actions of pregabalin. Proc Natl Acad Sci USA. 2006;103(46):17537-17542.

19. Arnold LM, Russel IJ, Diri EW, et al. A 14-week, randomized, double-blind, placebo-controlled, monotherapy trial of pregabalin in patients with fibromyalgia. J Pain. 2008;9(9):792-805.

20. Mease PJ, Russel IJ, Arnold LM, et al. A randomized, double-blind, placebo-controlled, phase III trial of pregabalin in the treatment of patients with fibromyalgia. J Rheumatol. 2008;35(3):502-514.

21. Crofford LJ, Mease J, Simpson SL, et al. Fibromyalgia relapse evaluation and efficacy for durability of meaningful relief (FREEDOM): a 6-month, double-blind, placebo-controlled trial with pregabalin. Pain. 2008;136(3):419-431.

22. Pregabalin [package insert]. New York, NY: Pfizer, Inc.; 2004.

23. Arnold LM, Pritchett YL, D’Souza DN, et al. Duloxetine for the treatment of fibromyalgia in women: pooled results from two randomized, placebo-controlled clinical trials. J Womens Health (Larchmt). 2007;16(8):1145-1156.

24. Arnold LM, Lu Y, Crofford LJ, et al. A double-blind, multicenter trial comparing duloxetine with placebo in the treatment of fibromyalgia patients with or without major depressive disorder. Arthritis Rheum. 2004;50(9):2974-2984.

25. Russell IJ, Mease PJ, Smith TR, et al. Efficacy and safety of duloxetine for treatment of fibromyalgia in patients with or without major depressive disorder: results from a 6-month, randomized, double-blind, placebo-controlled, fixed-dose trial. Pain. 2008;136(3):432-444.

26. Clauw DJ, Mease P, Palmer RH, et al. Milnacipran for the treatment of fibromyalgia in adults: a 15-week, multicenter, randomized, double-blind, placebo-controlled, multiple-dose clinical trial. Clin Ther. 2008;30(11):1988-2004.

27. Goldenberg DL, Felson DT, Dinerman H. A randomized, controlled trial of amitriptyline and naproxen in the treatment of patients with fibromyalgia. Arthritis Rheum. 1986;29(11):1371-1377.

28. Hannonen P, Malminiemi K, Yli-Kerttula U, et al. A randomized, double-blind, placebo-controlled study of moclobemide and amitriptyline in the treatment of fibromyalgia in females without psychiatric disorder. Br J Rheumatol. 1998;37:1279-1286.

29. Arnold LM, Goldenberg DL, Stanford SB, et al. Gabapentin in the treatment of fibromyalgia: a randomized, double-blind, placebo-controlled, multicenter trial. Arthritis Rheum. 2007;56:1336-1344.

30. Bennett RM, Schein J, Kosinski MR, et al. Impact of fibromyalgia pain on health-related quality of life before and after treatment with tramadol/ acetaminophen. Arthritis Rheum. 2005;53:519-527.

31. Arnold LM, Hess EV, Hudson JI, et al. Randomized, placebo-controlled, double-blind, flexible-dose study of fluoxetine in the treatment of women with fibromyalgia. Am J Med. 2002;112:191-197.

32. Dwight MM, Arnold LM, O’Brien H, et al. An open clinical trial of venlafaxine treatment of fibromyalgia. Psychosomatics. 1998;39:14-17.

Issue
Current Psychiatry - 08(03)
Issue
Current Psychiatry - 08(03)
Page Number
37-50
Page Number
37-50
Publications
Publications
Article Type
Display Headline
Fibromyalgia: Psychiatric drugs target CNS-linked symptoms
Display Headline
Fibromyalgia: Psychiatric drugs target CNS-linked symptoms
Legacy Keywords
fibromyalgia;pregabalin;duloxetine;milnacipran; somataform disorder;somatization disorder;Sharon Stanford;Sharon Shay
Legacy Keywords
fibromyalgia;pregabalin;duloxetine;milnacipran; somataform disorder;somatization disorder;Sharon Stanford;Sharon Shay
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Compulsive bruxism: How to protect your patients’ teeth

Article Type
Changed
Tue, 12/11/2018 - 15:06
Display Headline
Compulsive bruxism: How to protect your patients’ teeth

Comment on this article

Oral habits such as bruxism—compulsive grinding or clenching of the teeth—can be a manifestation of obsessive-compulsive disorder (OCD) and other anxiety disorders.1 Bruxism also may be a side effect of selective serotonin reuptake inhibitors (SSRIs)2,3 used to treat OCD4 and depression. Other oral conditions can complicate treatment of these disorders (Table 1).

Potentially serious sequelae of bruxism and similar behaviors include:

  • wearing down of teeth (more common)
  • necrosis of the pulpal tissues that results in non-vital teeth (less common).

The following case underlines the need for early referral to a dentist and close follow-up for patients who have tooth-related behaviors or are taking medications associated with a risk for such behaviors.

Table 1

Oral conditions associated with anxiety disorders and depression

Bruxism
Canker sores
Dry mouth
Temporomandibular joint disorders
Lichen planus (redness or mouth ulcers)
Non-vital teeth
Tooth wear, fracture

CASE REPORT: A compulsive oral habit

Mr. G, age 26, presents to our school of dental medicine with the chief complaint that he needs a crown. On clinical intraoral examination, we find he has multiple restorations, some areas of recurrent tooth decay, and a fractured cusp on a maxillary molar. His mandibular incisors show greater wear on their incisal surfaces than would be expected for a patient his age. This is especially true of his mandibular right lateral incisor (tooth #26) and canine (tooth #27) (Photo 1).

Clinical examination also reveals a restoration on tooth #26 that was consistent with an access cavity drilled for endodontic (root canal) therapy (Photo 2). This finding is consistent with his dental history. Soft tissue examination is within normal limits. He reports that he saw a dentist 7 months earlier but could not afford the fees.

During his medical history, Mr. G states he has mild Tourette’s syndrome that was diagnosed when he was 10. In the early 1990s he tried several medications, including haloperidol and pimozide, as a subject in research studies of Tourette’s. He could not recall the dosages of the medications or for how long he took them. Because these medications did not improve his symptoms, he stopped taking them after the studies ended.


Photo 1 Unexpected tooth wear: A clue to an anxiety-related oral habit

Teeth of a 26-year-old man show greater than expected wear, particularly on the mandibular right lateral incisor (tooth #26) and canine (tooth #27).

Photo 2 Radiographic evidence of tooth non-vitality

Radiolucencies (dark areas) in the bone at the apices of the tooth roots are a radiographic sign of non-vitality. Tooth #26 has undergone root canal.Mr. G reports that when he was in second grade, a psychiatrist diagnosed him with OCD, and has received treatment since then. We observe that while Mr. G is seated, he continually raises his right arm above his head and rubs his fingers together. He reports and demonstrates numerous other compulsive rituals, including head movements and rubbing his elbows against his side. His right elbow has a large scab.

He has been taking sertraline, 150 mg/d, for the past month. He says his psychiatrist prescribed this medication to help him break out of what he describes as episodes where he “gets into a mental loop.” Sertraline has improved Mr. G’s symptoms but they have not resolved.

He further reports that he has begun to “grind” his anterior teeth. Technically, he does not engage in grinding or bruxing; he has a habit of pushing his mandible forward so that his mandibular (lower) teeth are anterior to the maxillary (top) teeth, then forcefully pulling his mandible back so that the lingual (back) surfaces of the mandibular incisors push up against the buccal (outside) surfaces of the maxillary incisors (Photo 3). He states that he engaged in this habit frequently from approximately age 19 to 22. When Mr. G was 22, his dentist reduced the height of tooth #23, which Mr. G says he used “to set things in motion.” The dentist’s maneuver cut down but did not eliminate Mr. G’s habit.

Mr. G had not complained of nor had any clinician asked him about his bruxism-like behavior. He noted that the oral habit began prior to sertraline treatment, thus suggesting no relationship between the medication and the behavior. Interestingly, although some studies have reported bruxism as a side effect of SSRIs,2,3 at least 1 case report found that SSRIs reduced nocturnal bruxism.12


Photo 3 Obsessive-compulsive disorder manifested in Mr. G’s oral habit

 

 

Starting with his mandible pushed forward so that his mandibular (lower) teeth were anterior to the maxillary (upper) teeth, the patient would forcefully pull his mandible back so that the lingual (back) surfaces of the mandibular incisors pushed against the buccal (outside) surfaces of the maxillary incisors.As part of Mr. G’s dental examination, we take a full series of intraoral radiographs. These reveal radiolucencies at the apices of teeth #23 through #26 (Photo 2). The films also show root canal therapy on tooth #26.

Differential diagnosis for lesions in the periapical region of the mandibular incisors includes periapical cemental dysplasia (PCD), which typically is found in middle-aged African-American females,5 and lesions resulting from non-vitality of the teeth. Histopathologically, lesions resulting from the latter include an apical abscess, cyst, or granuloma.

As is customary when periapical lesions are noted, we test the vitality of the affected teeth. None of the affected teeth responded to cold or electric pulp testing, which indicated they were non-vital. Tooth vitality is not affected in PCD, which allowed us to exclude this condition.

Non-vital teeth indicate that the pulpal tissue is necrotic. Most commonly, non-vitality occurs when decay has penetrated the pulp chamber or as a complication of physical trauma. No decay was present on Mr. G’s mandibular anterior teeth and he denied a history of trauma such as a blow to the teeth. This left his oral habit as the likely cause of non-vitality.

Treatment for a non-vital tooth is a root canal, which had been done on tooth #26. We successfully performed root canal on Mr. G’s other non-vital teeth. We informed the patient of reason for his non-vital teeth, and made a protective occlusal guard to try to prevent additional trauma to the affected teeth.

Recognizing oral habits

Restoration of worn teeth, particularly those of the mandibular anterior, is technically difficult and—depending on the nature of the restoration—quite expensive. Endodontic therapy is more successful in teeth without periapical disease.6 Thus, preventing tooth-related problems in patients who grind their teeth or engage in other destructive dental behaviors is important.

As this case illustrates, teeth can become non-vital without clinical evidence of tooth wear; clinical evidence may be subtle or nonexistent (note teeth #23, #24, and #25 in Photo 2). Absence of tooth wear is not a reliable sign of tooth vitality. Mild to moderate tooth wear usually goes unnoticed by patients and clinicians.7

Patients with bruxism may complain of masticatory muscle soreness or increased wear of the teeth.7 In extreme cases, they may self-extract teeth as a result of bruxism.8

Screen patients who have anxiety disorders or depression for signs of bruxism or related behaviors (Table 2). If you detect signs of bruxism or related behavior, refer the patient to a dentist. Ask the dentist to look for signs of wear and perform vitality testing of teeth on a regular basis (twice a year is reasonable). Any signs of changes in pulp vitality should be followed up with intraoral periapical radiographs, which these patients might need more frequently than FDA guidelines recommend.9

Table 2

Screening for bruxism: 3 questions for patients

1. Do you have pain or discomfort in the jaw or facial muscles, headaches or earaches, or increased tooth sensitivity?
2. Have you noticed changes in the way your teeth fit together or wearing down of your teeth?
3. Has your sleeping partner noticed any noise at night that might be the result of teeth grinding?

An occlusal guard may provide the most definitive tooth protection for patients who engage in bruxism or similar behaviors. Occlusal guards are made of material that is softer than enamel, so the patient will wear away the guard rather than tooth structure. When the guard is worn away, the patient needs a new one.

Pharmacologic strategies for bruxism or related oral habits involving the teeth are not well developed. One short-term, placebo-controlled trial for acute treatment in 10 drug-free patients with sleep bruxism consisted of a predrug night, a placebo night, and a clonazepam night. Clonazepam, 1 mg 30 minutes before bedtime, significantly improved bruxism and sleep quality as determined by objective and subjective measures.10

Kast11 reported 4 cases in which tiagabine suppressed nocturnal bruxism, trismus, and consequent morning pain in the teeth, masticatory musculature, jaw, and temporomandibular joint areas. This gammaaminobutyric acid reuptake inhibitor anticonvulsant approved for treating partial seizures was dosed at 4 to 8 mg at bedtime. These dosages are lower than those used to treat seizures.

 

 

Related resources

Drug brand names

  • Clonazepam • Klonopin
  • Haloperidol • Haldol
  • Pimozide • Orap
  • Sertraline • Zoloft
  • Tiagabine • Gabitril

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. Herren C, Lindroth J. Obsessive compulsive disorder: a case report. J Contemp Dent Pract. 2001;2:41-49.

2. Ellison JM, Stanziani P. SSRI-associated nocturnal bruxism in four patients. J Clin Psychiatry. 1993;54:432-434.

3. Gerber PE, Lynd LD. Selective serotonin-reuptake inhibitor-induced movement disorders. Ann Pharmacother. 1998;32:692-698.

4. Kent JM, Coplan JD, Gorman JM. Clinical utility of the selective serotonin reuptake inhibitors in the spectrum of anxiety. Biol Psychiatry. 1998;44:812-824.

5. White SC, Pharoah MJ. Oral radiology: principles and interpretation. St. Louis, MO: Mosby; 2004:492.

6. Salehrabi R, Rotstein I. Endodontic treatment outcome in a large patient population in the USA: an epidemiological study. J Endod. 2004;30:846-850.

7. Lobbezoo F, van Denderen RJ, Verheij JG, et al. Reports of SSRI-associated bruxism in the family physician’s office. J Orofac Pain. 2001;15:340-346.

8. Eisenhauer GL, Woody RC. Self-mutilation and Tourette’s disorder. J Child Neurol. 1987;2:265-267.

9. American Dental Association. Guidelines for prescribing dental radiographs. Available at: http://www.ada.org/prof/resources/topics/topics_radiography_chart.pdf. Accessed December 17, 2008.

10. Saletu A, Parapatics S, Saletu B, et al. On the pharmacotherapy of sleep bruxism: placebo-controlled polysomnographic and psychometric studies with clonazepam. Neuropsychobiology. 2005;51:214-225.

11. Kast RE. Tiagabine may reduce bruxism and associated temporomandibular joint pain. Anesth Prog. 2005;52:102-104.

12. Stein DJ, Van Greunen G, Niehaus D. Can bruxism respond to serotonin reuptake inhibitors? [letter]. J Clin Psychiatry. 1998;59:133.-

Article PDF
Author and Disclosure Information

Bernard Friedland, BChD, MSc, JD
Assistant professor of oral medicine, infection, and immunity, Harvard School of Dental Medicine, Boston, MA

Theo C. Manschreck, MD, MPH
Professor of psychiatry, Harvard Medical School, Boston, MA

Issue
Current Psychiatry - 08(03)
Publications
Page Number
24-32
Legacy Keywords
bruxism;teeth grinding;anxiety;oral habits;selective serotonin reuptake inhibitors;SSRIs;Bernard Friedland;Theo Manschreck
Sections
Author and Disclosure Information

Bernard Friedland, BChD, MSc, JD
Assistant professor of oral medicine, infection, and immunity, Harvard School of Dental Medicine, Boston, MA

Theo C. Manschreck, MD, MPH
Professor of psychiatry, Harvard Medical School, Boston, MA

Author and Disclosure Information

Bernard Friedland, BChD, MSc, JD
Assistant professor of oral medicine, infection, and immunity, Harvard School of Dental Medicine, Boston, MA

Theo C. Manschreck, MD, MPH
Professor of psychiatry, Harvard Medical School, Boston, MA

Article PDF
Article PDF

Comment on this article

Oral habits such as bruxism—compulsive grinding or clenching of the teeth—can be a manifestation of obsessive-compulsive disorder (OCD) and other anxiety disorders.1 Bruxism also may be a side effect of selective serotonin reuptake inhibitors (SSRIs)2,3 used to treat OCD4 and depression. Other oral conditions can complicate treatment of these disorders (Table 1).

Potentially serious sequelae of bruxism and similar behaviors include:

  • wearing down of teeth (more common)
  • necrosis of the pulpal tissues that results in non-vital teeth (less common).

The following case underlines the need for early referral to a dentist and close follow-up for patients who have tooth-related behaviors or are taking medications associated with a risk for such behaviors.

Table 1

Oral conditions associated with anxiety disorders and depression

Bruxism
Canker sores
Dry mouth
Temporomandibular joint disorders
Lichen planus (redness or mouth ulcers)
Non-vital teeth
Tooth wear, fracture

CASE REPORT: A compulsive oral habit

Mr. G, age 26, presents to our school of dental medicine with the chief complaint that he needs a crown. On clinical intraoral examination, we find he has multiple restorations, some areas of recurrent tooth decay, and a fractured cusp on a maxillary molar. His mandibular incisors show greater wear on their incisal surfaces than would be expected for a patient his age. This is especially true of his mandibular right lateral incisor (tooth #26) and canine (tooth #27) (Photo 1).

Clinical examination also reveals a restoration on tooth #26 that was consistent with an access cavity drilled for endodontic (root canal) therapy (Photo 2). This finding is consistent with his dental history. Soft tissue examination is within normal limits. He reports that he saw a dentist 7 months earlier but could not afford the fees.

During his medical history, Mr. G states he has mild Tourette’s syndrome that was diagnosed when he was 10. In the early 1990s he tried several medications, including haloperidol and pimozide, as a subject in research studies of Tourette’s. He could not recall the dosages of the medications or for how long he took them. Because these medications did not improve his symptoms, he stopped taking them after the studies ended.


Photo 1 Unexpected tooth wear: A clue to an anxiety-related oral habit

Teeth of a 26-year-old man show greater than expected wear, particularly on the mandibular right lateral incisor (tooth #26) and canine (tooth #27).

Photo 2 Radiographic evidence of tooth non-vitality

Radiolucencies (dark areas) in the bone at the apices of the tooth roots are a radiographic sign of non-vitality. Tooth #26 has undergone root canal.Mr. G reports that when he was in second grade, a psychiatrist diagnosed him with OCD, and has received treatment since then. We observe that while Mr. G is seated, he continually raises his right arm above his head and rubs his fingers together. He reports and demonstrates numerous other compulsive rituals, including head movements and rubbing his elbows against his side. His right elbow has a large scab.

He has been taking sertraline, 150 mg/d, for the past month. He says his psychiatrist prescribed this medication to help him break out of what he describes as episodes where he “gets into a mental loop.” Sertraline has improved Mr. G’s symptoms but they have not resolved.

He further reports that he has begun to “grind” his anterior teeth. Technically, he does not engage in grinding or bruxing; he has a habit of pushing his mandible forward so that his mandibular (lower) teeth are anterior to the maxillary (top) teeth, then forcefully pulling his mandible back so that the lingual (back) surfaces of the mandibular incisors push up against the buccal (outside) surfaces of the maxillary incisors (Photo 3). He states that he engaged in this habit frequently from approximately age 19 to 22. When Mr. G was 22, his dentist reduced the height of tooth #23, which Mr. G says he used “to set things in motion.” The dentist’s maneuver cut down but did not eliminate Mr. G’s habit.

Mr. G had not complained of nor had any clinician asked him about his bruxism-like behavior. He noted that the oral habit began prior to sertraline treatment, thus suggesting no relationship between the medication and the behavior. Interestingly, although some studies have reported bruxism as a side effect of SSRIs,2,3 at least 1 case report found that SSRIs reduced nocturnal bruxism.12


Photo 3 Obsessive-compulsive disorder manifested in Mr. G’s oral habit

 

 

Starting with his mandible pushed forward so that his mandibular (lower) teeth were anterior to the maxillary (upper) teeth, the patient would forcefully pull his mandible back so that the lingual (back) surfaces of the mandibular incisors pushed against the buccal (outside) surfaces of the maxillary incisors.As part of Mr. G’s dental examination, we take a full series of intraoral radiographs. These reveal radiolucencies at the apices of teeth #23 through #26 (Photo 2). The films also show root canal therapy on tooth #26.

Differential diagnosis for lesions in the periapical region of the mandibular incisors includes periapical cemental dysplasia (PCD), which typically is found in middle-aged African-American females,5 and lesions resulting from non-vitality of the teeth. Histopathologically, lesions resulting from the latter include an apical abscess, cyst, or granuloma.

As is customary when periapical lesions are noted, we test the vitality of the affected teeth. None of the affected teeth responded to cold or electric pulp testing, which indicated they were non-vital. Tooth vitality is not affected in PCD, which allowed us to exclude this condition.

Non-vital teeth indicate that the pulpal tissue is necrotic. Most commonly, non-vitality occurs when decay has penetrated the pulp chamber or as a complication of physical trauma. No decay was present on Mr. G’s mandibular anterior teeth and he denied a history of trauma such as a blow to the teeth. This left his oral habit as the likely cause of non-vitality.

Treatment for a non-vital tooth is a root canal, which had been done on tooth #26. We successfully performed root canal on Mr. G’s other non-vital teeth. We informed the patient of reason for his non-vital teeth, and made a protective occlusal guard to try to prevent additional trauma to the affected teeth.

Recognizing oral habits

Restoration of worn teeth, particularly those of the mandibular anterior, is technically difficult and—depending on the nature of the restoration—quite expensive. Endodontic therapy is more successful in teeth without periapical disease.6 Thus, preventing tooth-related problems in patients who grind their teeth or engage in other destructive dental behaviors is important.

As this case illustrates, teeth can become non-vital without clinical evidence of tooth wear; clinical evidence may be subtle or nonexistent (note teeth #23, #24, and #25 in Photo 2). Absence of tooth wear is not a reliable sign of tooth vitality. Mild to moderate tooth wear usually goes unnoticed by patients and clinicians.7

Patients with bruxism may complain of masticatory muscle soreness or increased wear of the teeth.7 In extreme cases, they may self-extract teeth as a result of bruxism.8

Screen patients who have anxiety disorders or depression for signs of bruxism or related behaviors (Table 2). If you detect signs of bruxism or related behavior, refer the patient to a dentist. Ask the dentist to look for signs of wear and perform vitality testing of teeth on a regular basis (twice a year is reasonable). Any signs of changes in pulp vitality should be followed up with intraoral periapical radiographs, which these patients might need more frequently than FDA guidelines recommend.9

Table 2

Screening for bruxism: 3 questions for patients

1. Do you have pain or discomfort in the jaw or facial muscles, headaches or earaches, or increased tooth sensitivity?
2. Have you noticed changes in the way your teeth fit together or wearing down of your teeth?
3. Has your sleeping partner noticed any noise at night that might be the result of teeth grinding?

An occlusal guard may provide the most definitive tooth protection for patients who engage in bruxism or similar behaviors. Occlusal guards are made of material that is softer than enamel, so the patient will wear away the guard rather than tooth structure. When the guard is worn away, the patient needs a new one.

Pharmacologic strategies for bruxism or related oral habits involving the teeth are not well developed. One short-term, placebo-controlled trial for acute treatment in 10 drug-free patients with sleep bruxism consisted of a predrug night, a placebo night, and a clonazepam night. Clonazepam, 1 mg 30 minutes before bedtime, significantly improved bruxism and sleep quality as determined by objective and subjective measures.10

Kast11 reported 4 cases in which tiagabine suppressed nocturnal bruxism, trismus, and consequent morning pain in the teeth, masticatory musculature, jaw, and temporomandibular joint areas. This gammaaminobutyric acid reuptake inhibitor anticonvulsant approved for treating partial seizures was dosed at 4 to 8 mg at bedtime. These dosages are lower than those used to treat seizures.

 

 

Related resources

Drug brand names

  • Clonazepam • Klonopin
  • Haloperidol • Haldol
  • Pimozide • Orap
  • Sertraline • Zoloft
  • Tiagabine • Gabitril

Disclosure

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

Comment on this article

Oral habits such as bruxism—compulsive grinding or clenching of the teeth—can be a manifestation of obsessive-compulsive disorder (OCD) and other anxiety disorders.1 Bruxism also may be a side effect of selective serotonin reuptake inhibitors (SSRIs)2,3 used to treat OCD4 and depression. Other oral conditions can complicate treatment of these disorders (Table 1).

Potentially serious sequelae of bruxism and similar behaviors include:

  • wearing down of teeth (more common)
  • necrosis of the pulpal tissues that results in non-vital teeth (less common).

The following case underlines the need for early referral to a dentist and close follow-up for patients who have tooth-related behaviors or are taking medications associated with a risk for such behaviors.

Table 1

Oral conditions associated with anxiety disorders and depression

Bruxism
Canker sores
Dry mouth
Temporomandibular joint disorders
Lichen planus (redness or mouth ulcers)
Non-vital teeth
Tooth wear, fracture

CASE REPORT: A compulsive oral habit

Mr. G, age 26, presents to our school of dental medicine with the chief complaint that he needs a crown. On clinical intraoral examination, we find he has multiple restorations, some areas of recurrent tooth decay, and a fractured cusp on a maxillary molar. His mandibular incisors show greater wear on their incisal surfaces than would be expected for a patient his age. This is especially true of his mandibular right lateral incisor (tooth #26) and canine (tooth #27) (Photo 1).

Clinical examination also reveals a restoration on tooth #26 that was consistent with an access cavity drilled for endodontic (root canal) therapy (Photo 2). This finding is consistent with his dental history. Soft tissue examination is within normal limits. He reports that he saw a dentist 7 months earlier but could not afford the fees.

During his medical history, Mr. G states he has mild Tourette’s syndrome that was diagnosed when he was 10. In the early 1990s he tried several medications, including haloperidol and pimozide, as a subject in research studies of Tourette’s. He could not recall the dosages of the medications or for how long he took them. Because these medications did not improve his symptoms, he stopped taking them after the studies ended.


Photo 1 Unexpected tooth wear: A clue to an anxiety-related oral habit

Teeth of a 26-year-old man show greater than expected wear, particularly on the mandibular right lateral incisor (tooth #26) and canine (tooth #27).

Photo 2 Radiographic evidence of tooth non-vitality

Radiolucencies (dark areas) in the bone at the apices of the tooth roots are a radiographic sign of non-vitality. Tooth #26 has undergone root canal.Mr. G reports that when he was in second grade, a psychiatrist diagnosed him with OCD, and has received treatment since then. We observe that while Mr. G is seated, he continually raises his right arm above his head and rubs his fingers together. He reports and demonstrates numerous other compulsive rituals, including head movements and rubbing his elbows against his side. His right elbow has a large scab.

He has been taking sertraline, 150 mg/d, for the past month. He says his psychiatrist prescribed this medication to help him break out of what he describes as episodes where he “gets into a mental loop.” Sertraline has improved Mr. G’s symptoms but they have not resolved.

He further reports that he has begun to “grind” his anterior teeth. Technically, he does not engage in grinding or bruxing; he has a habit of pushing his mandible forward so that his mandibular (lower) teeth are anterior to the maxillary (top) teeth, then forcefully pulling his mandible back so that the lingual (back) surfaces of the mandibular incisors push up against the buccal (outside) surfaces of the maxillary incisors (Photo 3). He states that he engaged in this habit frequently from approximately age 19 to 22. When Mr. G was 22, his dentist reduced the height of tooth #23, which Mr. G says he used “to set things in motion.” The dentist’s maneuver cut down but did not eliminate Mr. G’s habit.

Mr. G had not complained of nor had any clinician asked him about his bruxism-like behavior. He noted that the oral habit began prior to sertraline treatment, thus suggesting no relationship between the medication and the behavior. Interestingly, although some studies have reported bruxism as a side effect of SSRIs,2,3 at least 1 case report found that SSRIs reduced nocturnal bruxism.12


Photo 3 Obsessive-compulsive disorder manifested in Mr. G’s oral habit

 

 

Starting with his mandible pushed forward so that his mandibular (lower) teeth were anterior to the maxillary (upper) teeth, the patient would forcefully pull his mandible back so that the lingual (back) surfaces of the mandibular incisors pushed against the buccal (outside) surfaces of the maxillary incisors.As part of Mr. G’s dental examination, we take a full series of intraoral radiographs. These reveal radiolucencies at the apices of teeth #23 through #26 (Photo 2). The films also show root canal therapy on tooth #26.

Differential diagnosis for lesions in the periapical region of the mandibular incisors includes periapical cemental dysplasia (PCD), which typically is found in middle-aged African-American females,5 and lesions resulting from non-vitality of the teeth. Histopathologically, lesions resulting from the latter include an apical abscess, cyst, or granuloma.

As is customary when periapical lesions are noted, we test the vitality of the affected teeth. None of the affected teeth responded to cold or electric pulp testing, which indicated they were non-vital. Tooth vitality is not affected in PCD, which allowed us to exclude this condition.

Non-vital teeth indicate that the pulpal tissue is necrotic. Most commonly, non-vitality occurs when decay has penetrated the pulp chamber or as a complication of physical trauma. No decay was present on Mr. G’s mandibular anterior teeth and he denied a history of trauma such as a blow to the teeth. This left his oral habit as the likely cause of non-vitality.

Treatment for a non-vital tooth is a root canal, which had been done on tooth #26. We successfully performed root canal on Mr. G’s other non-vital teeth. We informed the patient of reason for his non-vital teeth, and made a protective occlusal guard to try to prevent additional trauma to the affected teeth.

Recognizing oral habits

Restoration of worn teeth, particularly those of the mandibular anterior, is technically difficult and—depending on the nature of the restoration—quite expensive. Endodontic therapy is more successful in teeth without periapical disease.6 Thus, preventing tooth-related problems in patients who grind their teeth or engage in other destructive dental behaviors is important.

As this case illustrates, teeth can become non-vital without clinical evidence of tooth wear; clinical evidence may be subtle or nonexistent (note teeth #23, #24, and #25 in Photo 2). Absence of tooth wear is not a reliable sign of tooth vitality. Mild to moderate tooth wear usually goes unnoticed by patients and clinicians.7

Patients with bruxism may complain of masticatory muscle soreness or increased wear of the teeth.7 In extreme cases, they may self-extract teeth as a result of bruxism.8

Screen patients who have anxiety disorders or depression for signs of bruxism or related behaviors (Table 2). If you detect signs of bruxism or related behavior, refer the patient to a dentist. Ask the dentist to look for signs of wear and perform vitality testing of teeth on a regular basis (twice a year is reasonable). Any signs of changes in pulp vitality should be followed up with intraoral periapical radiographs, which these patients might need more frequently than FDA guidelines recommend.9

Table 2

Screening for bruxism: 3 questions for patients

1. Do you have pain or discomfort in the jaw or facial muscles, headaches or earaches, or increased tooth sensitivity?
2. Have you noticed changes in the way your teeth fit together or wearing down of your teeth?
3. Has your sleeping partner noticed any noise at night that might be the result of teeth grinding?

An occlusal guard may provide the most definitive tooth protection for patients who engage in bruxism or similar behaviors. Occlusal guards are made of material that is softer than enamel, so the patient will wear away the guard rather than tooth structure. When the guard is worn away, the patient needs a new one.

Pharmacologic strategies for bruxism or related oral habits involving the teeth are not well developed. One short-term, placebo-controlled trial for acute treatment in 10 drug-free patients with sleep bruxism consisted of a predrug night, a placebo night, and a clonazepam night. Clonazepam, 1 mg 30 minutes before bedtime, significantly improved bruxism and sleep quality as determined by objective and subjective measures.10

Kast11 reported 4 cases in which tiagabine suppressed nocturnal bruxism, trismus, and consequent morning pain in the teeth, masticatory musculature, jaw, and temporomandibular joint areas. This gammaaminobutyric acid reuptake inhibitor anticonvulsant approved for treating partial seizures was dosed at 4 to 8 mg at bedtime. These dosages are lower than those used to treat seizures.

 

 

Related resources

Drug brand names

  • Clonazepam • Klonopin
  • Haloperidol • Haldol
  • Pimozide • Orap
  • Sertraline • Zoloft
  • Tiagabine • Gabitril

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. Herren C, Lindroth J. Obsessive compulsive disorder: a case report. J Contemp Dent Pract. 2001;2:41-49.

2. Ellison JM, Stanziani P. SSRI-associated nocturnal bruxism in four patients. J Clin Psychiatry. 1993;54:432-434.

3. Gerber PE, Lynd LD. Selective serotonin-reuptake inhibitor-induced movement disorders. Ann Pharmacother. 1998;32:692-698.

4. Kent JM, Coplan JD, Gorman JM. Clinical utility of the selective serotonin reuptake inhibitors in the spectrum of anxiety. Biol Psychiatry. 1998;44:812-824.

5. White SC, Pharoah MJ. Oral radiology: principles and interpretation. St. Louis, MO: Mosby; 2004:492.

6. Salehrabi R, Rotstein I. Endodontic treatment outcome in a large patient population in the USA: an epidemiological study. J Endod. 2004;30:846-850.

7. Lobbezoo F, van Denderen RJ, Verheij JG, et al. Reports of SSRI-associated bruxism in the family physician’s office. J Orofac Pain. 2001;15:340-346.

8. Eisenhauer GL, Woody RC. Self-mutilation and Tourette’s disorder. J Child Neurol. 1987;2:265-267.

9. American Dental Association. Guidelines for prescribing dental radiographs. Available at: http://www.ada.org/prof/resources/topics/topics_radiography_chart.pdf. Accessed December 17, 2008.

10. Saletu A, Parapatics S, Saletu B, et al. On the pharmacotherapy of sleep bruxism: placebo-controlled polysomnographic and psychometric studies with clonazepam. Neuropsychobiology. 2005;51:214-225.

11. Kast RE. Tiagabine may reduce bruxism and associated temporomandibular joint pain. Anesth Prog. 2005;52:102-104.

12. Stein DJ, Van Greunen G, Niehaus D. Can bruxism respond to serotonin reuptake inhibitors? [letter]. J Clin Psychiatry. 1998;59:133.-

References

1. Herren C, Lindroth J. Obsessive compulsive disorder: a case report. J Contemp Dent Pract. 2001;2:41-49.

2. Ellison JM, Stanziani P. SSRI-associated nocturnal bruxism in four patients. J Clin Psychiatry. 1993;54:432-434.

3. Gerber PE, Lynd LD. Selective serotonin-reuptake inhibitor-induced movement disorders. Ann Pharmacother. 1998;32:692-698.

4. Kent JM, Coplan JD, Gorman JM. Clinical utility of the selective serotonin reuptake inhibitors in the spectrum of anxiety. Biol Psychiatry. 1998;44:812-824.

5. White SC, Pharoah MJ. Oral radiology: principles and interpretation. St. Louis, MO: Mosby; 2004:492.

6. Salehrabi R, Rotstein I. Endodontic treatment outcome in a large patient population in the USA: an epidemiological study. J Endod. 2004;30:846-850.

7. Lobbezoo F, van Denderen RJ, Verheij JG, et al. Reports of SSRI-associated bruxism in the family physician’s office. J Orofac Pain. 2001;15:340-346.

8. Eisenhauer GL, Woody RC. Self-mutilation and Tourette’s disorder. J Child Neurol. 1987;2:265-267.

9. American Dental Association. Guidelines for prescribing dental radiographs. Available at: http://www.ada.org/prof/resources/topics/topics_radiography_chart.pdf. Accessed December 17, 2008.

10. Saletu A, Parapatics S, Saletu B, et al. On the pharmacotherapy of sleep bruxism: placebo-controlled polysomnographic and psychometric studies with clonazepam. Neuropsychobiology. 2005;51:214-225.

11. Kast RE. Tiagabine may reduce bruxism and associated temporomandibular joint pain. Anesth Prog. 2005;52:102-104.

12. Stein DJ, Van Greunen G, Niehaus D. Can bruxism respond to serotonin reuptake inhibitors? [letter]. J Clin Psychiatry. 1998;59:133.-

Issue
Current Psychiatry - 08(03)
Issue
Current Psychiatry - 08(03)
Page Number
24-32
Page Number
24-32
Publications
Publications
Article Type
Display Headline
Compulsive bruxism: How to protect your patients’ teeth
Display Headline
Compulsive bruxism: How to protect your patients’ teeth
Legacy Keywords
bruxism;teeth grinding;anxiety;oral habits;selective serotonin reuptake inhibitors;SSRIs;Bernard Friedland;Theo Manschreck
Legacy Keywords
bruxism;teeth grinding;anxiety;oral habits;selective serotonin reuptake inhibitors;SSRIs;Bernard Friedland;Theo Manschreck
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Postpartum psychosis: Strategies to protect infant and mother from harm

Article Type
Changed
Tue, 12/11/2018 - 15:06
Display Headline
Postpartum psychosis: Strategies to protect infant and mother from harm

Comment on this article

In June 2001, Andrea Yates drowned her 5 children ages 6 months to 7 years in the bathtub of their home. She had delusions that her house was bugged and television cameras were monitoring her mothering skills. She came to believe that “the one and only Satan” was within her, and that her children would burn in hell if she did not save their souls while they were still innocent.

Her conviction of capital murder in her first trial was overturned on appeal. She was found not guilty by reason of insanity at her retrial in 2006 and committed to a Texas state mental hospital.1

Postpartum psychosis (PPP) presents dramatically days to weeks after delivery, with wide-ranging symptoms that can include dysphoric mania and delirium. Because untreated PPP has an estimated 4% risk of infanticide (murder of the infant in the first year of life),2 and a 5% risk of suicide,3 psychiatric hospitalization usually is required to protect the mother and her baby.

The diagnosis may be missed, however, because postpartum psychotic symptoms wax and wane and suspiciousness or poor insight cause some women—such as Andrea Yates—to hide their delusional thinking from their families. This article discusses the risk factors, prevention, and treatment of PPP, including a review of:

  • infanticide and suicide risks in the postpartum period
  • increased susceptibility to PPP in women with bipolar disorder and other psychiatric disorders
  • hospitalization for support and safety of the mother and her infant.

Risks of infanticide and suicide

A number of motives exist for infanticide (Table 1).4 Psychiatric literature shows that mothers who kill their children often have experienced psychosis, suicidality, depression, and considerable life stress.5 Common factors include alcohol use, limited social support, and a personal history of abuse. Studies on infanticide found a significant increase in common psychiatric disorders and financial stress among the mothers. Neonaticide (murder of the infant in the first day of life) generally is not related to PPP because PPP usually does not begin until after the day of delivery.6

Among women who develop psychiatric illness, homicidal ideation is more frequent in those with a perinatal onset of psychopathology.7 Infanticidal ideas and behavior are associated with psychotic ideas about the infant.8 Suicide is the cause of up to 20% of postpartum deaths.9

Table 1

Motives for infanticide: Mental illness or something else?

MotivesExamples
Likely related to postpartum psychosis or depression
AltruisticA depressed or psychotic mother may believe she is sending her baby to heaven to prevent suffering on earth
A suicidal mother may kill her infant along with herself rather than leave the child alone
Acutely psychoticA mother kills her baby for no comprehensible reason, such as in response to command hallucinations or the confusion of delirium
Rarely related to postpartum psychosis
Fatal maltreatment‘Battered child’ syndrome is the most common cause of infanticide; death often occurs after chronic abuse or neglect
A minority of perpetrators are psychotic; a mother out of touch with reality may have difficulty providing for her infant’s needs
Not likely related to postpartum psychosis
Unwanted childParent does not want child because of inconvenience or out-of-wedlock birth
Spouse revengeMurder of a child to cause emotional suffering for the other parent is the least frequent motive for infanticide
Source: Reference 4

The bipolar connection

Many factors can elevate the risk of PPP, including sleep deprivation in susceptible women, the hormonal shifts after birth, and psychiatric comorbidity (Table 2). Nearly three-fourths (>72%) of mothers with PPP have bipolar disorder or schizoaffective disorder, whereas 12% have schizophrenia.10 Some authors consider PPP to be bipolar disorder until proven otherwise. Mothers with a history of bipolar disorder or PPP have a 100-fold increase in rates of psychiatric hospitalization in the postpartum period.11

PPP is not categorized as a distinct disorder in DSM-IV-TR, and lack of a consistent terminology has led to differing definitions. Brief psychotic disorder, psychotic disorder not otherwise specified, and affective disorders are sometimes proffered.12 Some DSM disorders permit the specifier “with postpartum onset” if the symptoms occur in mothers within 4 weeks of birth.

Presentation. PPP is relatively rare, occurring at a rate of 1 to 3 cases per 1,000 births. Symptoms often have an abrupt onset, within days to weeks of delivery.10 In at least one-half of cases, symptoms begin by the third postpartum day,13 when many mothers have been discharged home and may be solely responsible for their infants.

Symptoms include confusion, bizarre behaviors, hallucinations (including rarer types such as tactile and olfactory), mood lability (ranging from euphoria to depression), decreased need for sleep or insomnia, restlessness, agitation, disorganized thinking, and bizarre delusions of relatively rapid onset.13 One mother might believe God wants her baby to be sacrificed as the second coming of the Messiah, a second may believe she has special powers, and a third that her baby is defective.

 

 

Table 2

Postpartum psychosis: Risk factors supported by evidence

Sleep deprivation in susceptible women
Hormonal shifts after birth (primarily the rapid drop in estrogen)
Psychosocial stressors such as marital problems, older age, single motherhood, lower socioeconomic status
Bipolar disorder or schizoaffective disorder
Past history of postpartum psychosis
Family history of postpartum psychosis
Previous psychiatric hospitalization, especially during the prenatal period for a bipolar or psychotic condition
Menstruation or cessation of lactation
Obstetric factors that can cause a small increase in relative risk:
  • first pregnancy
  • delivery complications
  • preterm birth
  • acute Caesarean section
  • long duration of labor
Source: For bibliographic citations

Differential diagnosis

When evaluating a postpartum woman with psychotic symptoms, stay in contact with her obstetrician and the child’s pediatrician. Rule out delirium and organic causes of the mother’s symptoms (Box).11

The psychiatric differential diagnosis includes “baby blues”—mild, transient mood swings, sadness, irritability, anxiety, and insomnia that most new mothers experience in the first postpartum week. Schizophrenia’s delusional thinking and hallucinations have a more gradual onset, compared with those of postpartum psychosis.

Postpartum depression (PPD) occurs in approximately 10% to 15% of new mothers.14 Depressive symptoms occur within weeks to months after delivery and often coexist with anxious symptoms. Some women with severe depression may present with psychotic symptoms. A mother may experience insomnia, sometimes not being able to sleep when the baby is sleeping. She may lack interest in caring for her baby and experience difficulty bonding.

At times it can be difficult to distinguish PPD from PPP. When evaluating a mother who is referred for “postpartum depression,” consider PPP in the differential diagnosis. A woman with PPD or PPP may report depressed mood, but in PPP this symptom usually is related to rapid mood changes. Other clinical features that point toward PPP are abnormal hallucinations (such as olfactory or tactile), hypomanic or mixed mood symptoms, and confusion.

Box

Medical workup in differential diagnosis of postpartum psychosis

When evaluating a postpartum woman with psychotic symptoms, stay in contact with her obstetrician and the child’s pediatrician. Rule out delirium and organic causes of the mother’s symptoms, giving special consideration to metabolic, neurologic, cardiovascular, infectious, and substance- or medication-induced origins. The extensive differential diagnosis includes:

  • thyroiditis
  • tumor
  • CNS infection
  • head injury
  • embolism
  • eclampsia
  • substance withdrawal
  • medication-induced (such as corticosteroids)
  • electrolyte anomalies
  • anoxia
  • vitamin B12 deficiency.11
Suicidal thoughts or thoughts of harming the infant may be present in either PPD or PPP. Both elevate the risk of infanticide; one study found that 41 out of 100 depressed mothers acknowledged having thoughts of harming their infants.15

Psychosis vs OCD. Psychotic thinking and behaviors also must be differentiated from obsessive thoughts and compulsions.10,16 Obsessive compulsive disorder (OCD) may be exacerbated or emerge for the first time during the perinatal period.17

In postpartum OCD, women may experience intrusive thoughts of accidental or purposeful harm to their baby. As opposed to women with PPP, mothers with OCD are not out of touch with reality and their thoughts are ego-dystonic.17 When these mothers have thoughts of their infants being harmed, they realize that these thoughts are not plans but fears and they try to avoid the thoughts.

Preventing PPP

Bipolar disorder is one of the most difficult disorders to treat during pregnancy because the serious risks of untreated illness must be balanced against the potential teratogenic risk of medications. Nevertheless, proactively managing bipolar disorder during pregnancy may reduce the risk of PPP.10

Closely monitor women with a history of bipolar disorder or PPP. During pregnancy, counsel them—and their partners—to:

  • anticipate that depressive or psychotic symptoms could develop within days after delivery18
  • seek treatment immediately if this occurs.
Some women will prefer to remain off medication during the first trimester—which is critical in organogenesis—and then restart medication later in pregnancy. This approach is not without risks, however (see Related Resources).

Postpartum medication. Whether or not a woman with bipolar disorder takes medication during pregnancy, consider treatment with mood stabilizers or atypical antipsychotics in the postpartum to prevent PPP (Table 3). Evidence is limited, but a search of PubMed found 1 study in which prophylactic lithium was given late in the third trimester or immediately after delivery to 21 women with a history of bipolar disorder or PPP. Only 2 patients had a psychotic recurrence while on prophylactic lithium; 1 unexplained stillbirth occurred.19

A retrospective study examined the course of women with bipolar disorder, some of whom were given prophylactic mood stabilizers immediately in the postpartum. One of 14 who received antimanic agents relapsed within the first 3 months postpartum, compared with 8 of 13 who were not so treated.18

 

 

Compared with antiepileptics, less information is available about the use of atypical antipsychotics in pregnancy and lactation. Antipsychotics’ potential advantage in women at risk for PPP is that these agents may help prevent or treat both manic and psychotic symptoms.

In a small, naturalistic, prospective study, 11 women at risk for PPP received olanzapine alone or with an antidepressant or mood stabilizer for at least 4 weeks after delivery. Two (18%) experienced a postpartum mood episode, compared with 8 (57%) of 14 other at-risk women who received antidepressants, mood stabilizers, or no medication.20

Breast-feeding. Consider treatment effects on lactation and discuss this with the mother and the baby’s pediatrician, when possible. For useful reviews of risks and benefits of mood stabilizers and antipsychotics during breast-feeding, see Related Resources.

When you discuss breast-feeding, consider possible risks to the neonate as well as potential sleep interruption for the mother. If a mother has a supportive partner, the partner might be put in charge of night-time feedings in a routine combining breast-feeding and bottle-feeding. In some cases you may need to recommend cessation of lactation.21

Table 3

Treating postpartum psychosis: Consider 3 components

ComponentRecommendations
Hospitalization vs home careHospitalize in most cases because of emergent severe symptoms and fluctuating course; base decision on risk evaluation/safety issues for patient and infant
After discharge, visiting nurses are useful to help monitor the mother’s condition at home
PsychoeducationEducate patient, family, and social support network; address risks to mother and infant and risks in future pregnancies
MedicationWhen prescribing mood stabilizers and/or antipsychotics, consider:
  • whether mother is breast-feeding (discuss with patient, family, and pediatrician)
  • maternal side effects, including sedation

Managing PPP

Early symptoms. Because of its severity and rapid evolution, PPP often presents as a psychiatric emergency. Monitor atrisk patients’ sleep patterns and mood for early signs of psychosis.22 Watch especially for hypomanic symptoms such as elevated or mixed mood and decreased judgment, which are common early in PPP.13

A mother with few signs of abnormal mood, good social support, and close follow-up may potentially be safely managed as an outpatient. Initial evaluation and management of PPP usually requires hospitalization, however, because of the risks of suicide, infanticide, and child maltreatment.23

Hospitalization. Mother-infant bonding is important, but safety is paramount if a mother is psychotic—especially if she is experiencing psychotic thoughts about her infant. If possible, the infant should remain with family members during the mother’s hospitalization. Supervised mother-infant visits are often arranged, as appropriate.

Mood-stabilizing medications, including antipsychotics, are mainstays of treatment.24 In some cases, conventional antipsychotics such as haloperidol may be useful because of a lower risk of weight gain or of sedation that could impair a mother’s ability to respond to her infant. Electroconvulsive therapy often yields rapid symptomatic improvement for mothers with postpartum mood or psychotic symptoms.25

During the mother’s hospitalization, encourage the staff to be supportive and convey hopefulness.26 In an interview study, women who had been treated for PPP said they experienced anger and frustration while hospitalized because they believed that they and their families received inadequate information and support.27

Discharge planning. Assuming that the mother adheres to prescribed treatment, discharge may occur within 1 week. Plan discharge arrangements carefully (Table 4).28 A team approach can be very useful within the outpatient clinic. In the model of the Perinatal Psychiatry Clinic of Connections in suburban Cleveland, OH, the mother’s treatment team includes perinatal psychiatrists, nurses, counsellors, case managers (who do home visits), and peer counselors.

Outpatient civil commitment, in which patients are mandated to accept treatment, is an option in some jurisdictions and could help ensure that patients receive treatment consistently.

Table 4

Discharge planning for safety of mother and infant

Notify child protective services (CPS) depending on the risk to the child. Case-by-case review is needed to assess whether the infant should be removed. CPS may put in place a plan for safety, short of removal. The plan may require that the woman continue psychiatric care
Meet with the patient and family to discuss her diagnosis, the risks, the importance of continued medication adherence, and the need for family or social supports to assist with child care
Consider engaging visiting nurses or doulas to provide help and support at home
Schedule frequent outpatient appointments for the mother after discharge
Consider family therapy after the mother has improved because of her risk for affective episodes outside the postpartum28
 

 

Related resources

  • Altshuler L, Richards M, Yonkers K. Treating bipolar disorder during pregnancy. Current Psychiatry. 2003;2(7):14-26. www.CurrentPsychiatry.com.
  • Gentile S. Infant safety with antipsychotic therapy in breastfeeding: a systematic review. J Clin Psychiatry. 2008;69(4):666-673.
  • Miller LJ. Postpartum mood disorders. Washington, DC: American Psychiatric Publishing, Inc; 1999.
  • Stowe ZN. The use of mood stabilizers during breastfeeding. J Clin Psychiatry. 2007;68(suppl 9):22-28.
  • Toxicology Data Network (Toxnet). Literature on reproductive risks associated with psychotropics. National Library of Medicine. http://toxnet.nlm.nih.gov.
Drug brand names

  • Haloperidol • Haldol
  • Lithium • various
  • Olanzapine • Zyprexa
Disclosures

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

Acknowledgement

Dr. Resnick, a forensic psychiatrist and coauthor of this article, testified for the defense in both trials of Andrea Yates.

References

1. Resnick PJ. The Andrea Yates case: insanity on trial. Cleveland State Law Review. 2007;55(2):147-156.

2. Altshuler LL, Hendrick V, Cohen LS. Course of mood and anxiety disorders during pregnancy and the postpartum period. J Clin Psychiatry. 1998;59(suppl. 2):29-33.

3. Knops GG. Postpartum mood disorders. Postgrad Med. 1993;93:103-116.

4. Resnick PJ. Child murder by parents: a psychiatric review of filicide. Am J Psychiatry. 1969;126:73-82.

5. Friedman SH, Horwitz SM, Resnick PJ. Child murder by mothers: a critical analysis of the current state of knowledge and a research agenda. Am J Psychiatry. 2005;162:1578-1587.

6. Friedman SH, Resnick PJ. Neonaticide: phenomenology and considerations for prevention. Int J Law Psychiatry. In press.

7. Wisner K, Peindl K, Hanusa BH. Symptomatology of affective and psychotic illnesses related to childbearing. J Affect Disord. 1994;30:77-87.

8. Chandra PS, Venkatasubramanian G, Thomas T. Infanticidal ideas and infanticidal behaviour in Indian women with severe postpartum psychiatric disorders. J Nerv Ment Dis. 2002;190(7):457-461.

9. Lindahl V, Pearson JL, Colpe L. Prevalence of suicidality during pregnancy and the postpartum. Arch Womens Ment Health. 2005;8(2):77-87.

10. Sit D, Rothschild AJ, Wisner KL. A review of postpartum psychosis. J Women’s Health. 2006;15(4):352-368.

11. Attia E, Downey J, Oberman M. Postpartum psychoses. In: Miller LJ, ed. Postpartum mood disorders. Washington, DC: American Psychiatric Publishing Inc.; 1999:99-117.

12. Diagnostic and Statistical Manual of Mental Disorders, 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.

13. Heron J, McGuinness M, Blackmore ER, et al. Early postpartum symptoms in puerperal psychosis. BJOG. 2008;115(3):348-353.

14. Meltzer-Brody S, Payne J, Rubinow D. Postpartum depression: what to tell patients who breast-feed. Current Psychiatry. 2008;7(5):87-95.

15. Jennings KD, Ross S, Popper S, et al. Thoughts of harming infants in depressed and nondepressed mothers. J Affect Disord. 1999;54:21-28.

16. Wisner KL, Gracious BL, Piontek CM, et al. Postpartum disorders: phenomenology, treatment approaches, and relationship to infanticide. In: Spinelli MG, ed. Infanticide: psychosocial and legal perspectives on mothers who kill. Washington, DC: American Psychiatric Publishing, Inc.; 2003.

17. Fairbrother N, Abramowitz JS. New parenthood as a risk factors for the development of obsessional problems. Behav Res Ther. 2007;45(9):2155-2163.

18. Cohen LS, Sichel DA, Robertson LM, et al. Postpartum prophylaxis for women with bipolar disorder. Am J Psychiatry. 1995;152(11):1641-1645.

19. Stewart DE, Klompenhouwer JL, Kendell RE, et al. Prophylactic lithium in puerperal psychosis. Br J Psychiatry. 1991;158:393-397.

20. Sharma V, Smith A, Mazmanian D. Olanzapine in the prevention of postpartum psychosis and mood episodes in bipolar disorder. Bipolar Disord. 2006;8(4):400-404.

21. Pfuhlmann B, Stoeber G, Beckmann H. Postpartum psychoses: prognosis, risk factors, and treatment. Curr Psychiatry Rep. 2002;4(3):185-190.

22. Sharma V, Mazmanian D. Sleep loss and postpartum psychosis. Bipolar Disord. 2003;5(2):98-105.

23. Lindahl V, Pearson JL, Colpe L. Prevalence of suicidality during pregnancy and the postpartum. Arch Womens Ment Health. 2005;8(2):77-87.

24. Connell M. The postpartum psychosis defense and feminism: more or less justice for women? Case Western Reserve Law Review. 2002;53:143.-

25. Forray A, Ostroff RB. The use of electroconvulsive therapy in postpartum affective disorders. J ECT. 2007;23(3):188-193.

26. Engqvist I, Nilsson A, Nilsson K, et al. Strategies in caring for women with postpartum psychosis—an interview study with psychiatric nurses. J Clin Nurs. 2007;16(7):1333-1342.

27. Robertson E, Lyons A. Living with puerperal psychosis: a qualitative analysis. Psychol Psychother. 2003;76(4):411-431.

28. Robertson E, Jones I, Haque S, et al. Risk of puerperal and non-puerperal recurrence of illness following bipolar affective puerperal (post-partum) psychosis. Br J Psychiatry. 2005;186:258-259.

Article PDF
Author and Disclosure Information

Susan Hatters Friedman, MD
Senior instructor, Departments of psychiatry and pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH

Phillip J. Resnick, MD
Professor, Department of psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH

Miriam B. Rosenthal, MD
Associate professor emeriti, Departments of psychiatry and obstetrics and gynecology, Case Western Reserve University School of Medicine, Cleveland, OH

Issue
Current Psychiatry - 08(02)
Publications
Page Number
40-45
Legacy Keywords
postpartum psychosis; PPP; infanticide; Andrea Yates; postpartum depression; Susan Hatters Friedman; Phillip J Resnick; Miriam B rosenthal
Sections
Author and Disclosure Information

Susan Hatters Friedman, MD
Senior instructor, Departments of psychiatry and pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH

Phillip J. Resnick, MD
Professor, Department of psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH

Miriam B. Rosenthal, MD
Associate professor emeriti, Departments of psychiatry and obstetrics and gynecology, Case Western Reserve University School of Medicine, Cleveland, OH

Author and Disclosure Information

Susan Hatters Friedman, MD
Senior instructor, Departments of psychiatry and pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH

Phillip J. Resnick, MD
Professor, Department of psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH

Miriam B. Rosenthal, MD
Associate professor emeriti, Departments of psychiatry and obstetrics and gynecology, Case Western Reserve University School of Medicine, Cleveland, OH

Article PDF
Article PDF

Comment on this article

In June 2001, Andrea Yates drowned her 5 children ages 6 months to 7 years in the bathtub of their home. She had delusions that her house was bugged and television cameras were monitoring her mothering skills. She came to believe that “the one and only Satan” was within her, and that her children would burn in hell if she did not save their souls while they were still innocent.

Her conviction of capital murder in her first trial was overturned on appeal. She was found not guilty by reason of insanity at her retrial in 2006 and committed to a Texas state mental hospital.1

Postpartum psychosis (PPP) presents dramatically days to weeks after delivery, with wide-ranging symptoms that can include dysphoric mania and delirium. Because untreated PPP has an estimated 4% risk of infanticide (murder of the infant in the first year of life),2 and a 5% risk of suicide,3 psychiatric hospitalization usually is required to protect the mother and her baby.

The diagnosis may be missed, however, because postpartum psychotic symptoms wax and wane and suspiciousness or poor insight cause some women—such as Andrea Yates—to hide their delusional thinking from their families. This article discusses the risk factors, prevention, and treatment of PPP, including a review of:

  • infanticide and suicide risks in the postpartum period
  • increased susceptibility to PPP in women with bipolar disorder and other psychiatric disorders
  • hospitalization for support and safety of the mother and her infant.

Risks of infanticide and suicide

A number of motives exist for infanticide (Table 1).4 Psychiatric literature shows that mothers who kill their children often have experienced psychosis, suicidality, depression, and considerable life stress.5 Common factors include alcohol use, limited social support, and a personal history of abuse. Studies on infanticide found a significant increase in common psychiatric disorders and financial stress among the mothers. Neonaticide (murder of the infant in the first day of life) generally is not related to PPP because PPP usually does not begin until after the day of delivery.6

Among women who develop psychiatric illness, homicidal ideation is more frequent in those with a perinatal onset of psychopathology.7 Infanticidal ideas and behavior are associated with psychotic ideas about the infant.8 Suicide is the cause of up to 20% of postpartum deaths.9

Table 1

Motives for infanticide: Mental illness or something else?

MotivesExamples
Likely related to postpartum psychosis or depression
AltruisticA depressed or psychotic mother may believe she is sending her baby to heaven to prevent suffering on earth
A suicidal mother may kill her infant along with herself rather than leave the child alone
Acutely psychoticA mother kills her baby for no comprehensible reason, such as in response to command hallucinations or the confusion of delirium
Rarely related to postpartum psychosis
Fatal maltreatment‘Battered child’ syndrome is the most common cause of infanticide; death often occurs after chronic abuse or neglect
A minority of perpetrators are psychotic; a mother out of touch with reality may have difficulty providing for her infant’s needs
Not likely related to postpartum psychosis
Unwanted childParent does not want child because of inconvenience or out-of-wedlock birth
Spouse revengeMurder of a child to cause emotional suffering for the other parent is the least frequent motive for infanticide
Source: Reference 4

The bipolar connection

Many factors can elevate the risk of PPP, including sleep deprivation in susceptible women, the hormonal shifts after birth, and psychiatric comorbidity (Table 2). Nearly three-fourths (>72%) of mothers with PPP have bipolar disorder or schizoaffective disorder, whereas 12% have schizophrenia.10 Some authors consider PPP to be bipolar disorder until proven otherwise. Mothers with a history of bipolar disorder or PPP have a 100-fold increase in rates of psychiatric hospitalization in the postpartum period.11

PPP is not categorized as a distinct disorder in DSM-IV-TR, and lack of a consistent terminology has led to differing definitions. Brief psychotic disorder, psychotic disorder not otherwise specified, and affective disorders are sometimes proffered.12 Some DSM disorders permit the specifier “with postpartum onset” if the symptoms occur in mothers within 4 weeks of birth.

Presentation. PPP is relatively rare, occurring at a rate of 1 to 3 cases per 1,000 births. Symptoms often have an abrupt onset, within days to weeks of delivery.10 In at least one-half of cases, symptoms begin by the third postpartum day,13 when many mothers have been discharged home and may be solely responsible for their infants.

Symptoms include confusion, bizarre behaviors, hallucinations (including rarer types such as tactile and olfactory), mood lability (ranging from euphoria to depression), decreased need for sleep or insomnia, restlessness, agitation, disorganized thinking, and bizarre delusions of relatively rapid onset.13 One mother might believe God wants her baby to be sacrificed as the second coming of the Messiah, a second may believe she has special powers, and a third that her baby is defective.

 

 

Table 2

Postpartum psychosis: Risk factors supported by evidence

Sleep deprivation in susceptible women
Hormonal shifts after birth (primarily the rapid drop in estrogen)
Psychosocial stressors such as marital problems, older age, single motherhood, lower socioeconomic status
Bipolar disorder or schizoaffective disorder
Past history of postpartum psychosis
Family history of postpartum psychosis
Previous psychiatric hospitalization, especially during the prenatal period for a bipolar or psychotic condition
Menstruation or cessation of lactation
Obstetric factors that can cause a small increase in relative risk:
  • first pregnancy
  • delivery complications
  • preterm birth
  • acute Caesarean section
  • long duration of labor
Source: For bibliographic citations

Differential diagnosis

When evaluating a postpartum woman with psychotic symptoms, stay in contact with her obstetrician and the child’s pediatrician. Rule out delirium and organic causes of the mother’s symptoms (Box).11

The psychiatric differential diagnosis includes “baby blues”—mild, transient mood swings, sadness, irritability, anxiety, and insomnia that most new mothers experience in the first postpartum week. Schizophrenia’s delusional thinking and hallucinations have a more gradual onset, compared with those of postpartum psychosis.

Postpartum depression (PPD) occurs in approximately 10% to 15% of new mothers.14 Depressive symptoms occur within weeks to months after delivery and often coexist with anxious symptoms. Some women with severe depression may present with psychotic symptoms. A mother may experience insomnia, sometimes not being able to sleep when the baby is sleeping. She may lack interest in caring for her baby and experience difficulty bonding.

At times it can be difficult to distinguish PPD from PPP. When evaluating a mother who is referred for “postpartum depression,” consider PPP in the differential diagnosis. A woman with PPD or PPP may report depressed mood, but in PPP this symptom usually is related to rapid mood changes. Other clinical features that point toward PPP are abnormal hallucinations (such as olfactory or tactile), hypomanic or mixed mood symptoms, and confusion.

Box

Medical workup in differential diagnosis of postpartum psychosis

When evaluating a postpartum woman with psychotic symptoms, stay in contact with her obstetrician and the child’s pediatrician. Rule out delirium and organic causes of the mother’s symptoms, giving special consideration to metabolic, neurologic, cardiovascular, infectious, and substance- or medication-induced origins. The extensive differential diagnosis includes:

  • thyroiditis
  • tumor
  • CNS infection
  • head injury
  • embolism
  • eclampsia
  • substance withdrawal
  • medication-induced (such as corticosteroids)
  • electrolyte anomalies
  • anoxia
  • vitamin B12 deficiency.11
Suicidal thoughts or thoughts of harming the infant may be present in either PPD or PPP. Both elevate the risk of infanticide; one study found that 41 out of 100 depressed mothers acknowledged having thoughts of harming their infants.15

Psychosis vs OCD. Psychotic thinking and behaviors also must be differentiated from obsessive thoughts and compulsions.10,16 Obsessive compulsive disorder (OCD) may be exacerbated or emerge for the first time during the perinatal period.17

In postpartum OCD, women may experience intrusive thoughts of accidental or purposeful harm to their baby. As opposed to women with PPP, mothers with OCD are not out of touch with reality and their thoughts are ego-dystonic.17 When these mothers have thoughts of their infants being harmed, they realize that these thoughts are not plans but fears and they try to avoid the thoughts.

Preventing PPP

Bipolar disorder is one of the most difficult disorders to treat during pregnancy because the serious risks of untreated illness must be balanced against the potential teratogenic risk of medications. Nevertheless, proactively managing bipolar disorder during pregnancy may reduce the risk of PPP.10

Closely monitor women with a history of bipolar disorder or PPP. During pregnancy, counsel them—and their partners—to:

  • anticipate that depressive or psychotic symptoms could develop within days after delivery18
  • seek treatment immediately if this occurs.
Some women will prefer to remain off medication during the first trimester—which is critical in organogenesis—and then restart medication later in pregnancy. This approach is not without risks, however (see Related Resources).

Postpartum medication. Whether or not a woman with bipolar disorder takes medication during pregnancy, consider treatment with mood stabilizers or atypical antipsychotics in the postpartum to prevent PPP (Table 3). Evidence is limited, but a search of PubMed found 1 study in which prophylactic lithium was given late in the third trimester or immediately after delivery to 21 women with a history of bipolar disorder or PPP. Only 2 patients had a psychotic recurrence while on prophylactic lithium; 1 unexplained stillbirth occurred.19

A retrospective study examined the course of women with bipolar disorder, some of whom were given prophylactic mood stabilizers immediately in the postpartum. One of 14 who received antimanic agents relapsed within the first 3 months postpartum, compared with 8 of 13 who were not so treated.18

 

 

Compared with antiepileptics, less information is available about the use of atypical antipsychotics in pregnancy and lactation. Antipsychotics’ potential advantage in women at risk for PPP is that these agents may help prevent or treat both manic and psychotic symptoms.

In a small, naturalistic, prospective study, 11 women at risk for PPP received olanzapine alone or with an antidepressant or mood stabilizer for at least 4 weeks after delivery. Two (18%) experienced a postpartum mood episode, compared with 8 (57%) of 14 other at-risk women who received antidepressants, mood stabilizers, or no medication.20

Breast-feeding. Consider treatment effects on lactation and discuss this with the mother and the baby’s pediatrician, when possible. For useful reviews of risks and benefits of mood stabilizers and antipsychotics during breast-feeding, see Related Resources.

When you discuss breast-feeding, consider possible risks to the neonate as well as potential sleep interruption for the mother. If a mother has a supportive partner, the partner might be put in charge of night-time feedings in a routine combining breast-feeding and bottle-feeding. In some cases you may need to recommend cessation of lactation.21

Table 3

Treating postpartum psychosis: Consider 3 components

ComponentRecommendations
Hospitalization vs home careHospitalize in most cases because of emergent severe symptoms and fluctuating course; base decision on risk evaluation/safety issues for patient and infant
After discharge, visiting nurses are useful to help monitor the mother’s condition at home
PsychoeducationEducate patient, family, and social support network; address risks to mother and infant and risks in future pregnancies
MedicationWhen prescribing mood stabilizers and/or antipsychotics, consider:
  • whether mother is breast-feeding (discuss with patient, family, and pediatrician)
  • maternal side effects, including sedation

Managing PPP

Early symptoms. Because of its severity and rapid evolution, PPP often presents as a psychiatric emergency. Monitor atrisk patients’ sleep patterns and mood for early signs of psychosis.22 Watch especially for hypomanic symptoms such as elevated or mixed mood and decreased judgment, which are common early in PPP.13

A mother with few signs of abnormal mood, good social support, and close follow-up may potentially be safely managed as an outpatient. Initial evaluation and management of PPP usually requires hospitalization, however, because of the risks of suicide, infanticide, and child maltreatment.23

Hospitalization. Mother-infant bonding is important, but safety is paramount if a mother is psychotic—especially if she is experiencing psychotic thoughts about her infant. If possible, the infant should remain with family members during the mother’s hospitalization. Supervised mother-infant visits are often arranged, as appropriate.

Mood-stabilizing medications, including antipsychotics, are mainstays of treatment.24 In some cases, conventional antipsychotics such as haloperidol may be useful because of a lower risk of weight gain or of sedation that could impair a mother’s ability to respond to her infant. Electroconvulsive therapy often yields rapid symptomatic improvement for mothers with postpartum mood or psychotic symptoms.25

During the mother’s hospitalization, encourage the staff to be supportive and convey hopefulness.26 In an interview study, women who had been treated for PPP said they experienced anger and frustration while hospitalized because they believed that they and their families received inadequate information and support.27

Discharge planning. Assuming that the mother adheres to prescribed treatment, discharge may occur within 1 week. Plan discharge arrangements carefully (Table 4).28 A team approach can be very useful within the outpatient clinic. In the model of the Perinatal Psychiatry Clinic of Connections in suburban Cleveland, OH, the mother’s treatment team includes perinatal psychiatrists, nurses, counsellors, case managers (who do home visits), and peer counselors.

Outpatient civil commitment, in which patients are mandated to accept treatment, is an option in some jurisdictions and could help ensure that patients receive treatment consistently.

Table 4

Discharge planning for safety of mother and infant

Notify child protective services (CPS) depending on the risk to the child. Case-by-case review is needed to assess whether the infant should be removed. CPS may put in place a plan for safety, short of removal. The plan may require that the woman continue psychiatric care
Meet with the patient and family to discuss her diagnosis, the risks, the importance of continued medication adherence, and the need for family or social supports to assist with child care
Consider engaging visiting nurses or doulas to provide help and support at home
Schedule frequent outpatient appointments for the mother after discharge
Consider family therapy after the mother has improved because of her risk for affective episodes outside the postpartum28
 

 

Related resources

  • Altshuler L, Richards M, Yonkers K. Treating bipolar disorder during pregnancy. Current Psychiatry. 2003;2(7):14-26. www.CurrentPsychiatry.com.
  • Gentile S. Infant safety with antipsychotic therapy in breastfeeding: a systematic review. J Clin Psychiatry. 2008;69(4):666-673.
  • Miller LJ. Postpartum mood disorders. Washington, DC: American Psychiatric Publishing, Inc; 1999.
  • Stowe ZN. The use of mood stabilizers during breastfeeding. J Clin Psychiatry. 2007;68(suppl 9):22-28.
  • Toxicology Data Network (Toxnet). Literature on reproductive risks associated with psychotropics. National Library of Medicine. http://toxnet.nlm.nih.gov.
Drug brand names

  • Haloperidol • Haldol
  • Lithium • various
  • Olanzapine • Zyprexa
Disclosures

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

Acknowledgement

Dr. Resnick, a forensic psychiatrist and coauthor of this article, testified for the defense in both trials of Andrea Yates.

Comment on this article

In June 2001, Andrea Yates drowned her 5 children ages 6 months to 7 years in the bathtub of their home. She had delusions that her house was bugged and television cameras were monitoring her mothering skills. She came to believe that “the one and only Satan” was within her, and that her children would burn in hell if she did not save their souls while they were still innocent.

Her conviction of capital murder in her first trial was overturned on appeal. She was found not guilty by reason of insanity at her retrial in 2006 and committed to a Texas state mental hospital.1

Postpartum psychosis (PPP) presents dramatically days to weeks after delivery, with wide-ranging symptoms that can include dysphoric mania and delirium. Because untreated PPP has an estimated 4% risk of infanticide (murder of the infant in the first year of life),2 and a 5% risk of suicide,3 psychiatric hospitalization usually is required to protect the mother and her baby.

The diagnosis may be missed, however, because postpartum psychotic symptoms wax and wane and suspiciousness or poor insight cause some women—such as Andrea Yates—to hide their delusional thinking from their families. This article discusses the risk factors, prevention, and treatment of PPP, including a review of:

  • infanticide and suicide risks in the postpartum period
  • increased susceptibility to PPP in women with bipolar disorder and other psychiatric disorders
  • hospitalization for support and safety of the mother and her infant.

Risks of infanticide and suicide

A number of motives exist for infanticide (Table 1).4 Psychiatric literature shows that mothers who kill their children often have experienced psychosis, suicidality, depression, and considerable life stress.5 Common factors include alcohol use, limited social support, and a personal history of abuse. Studies on infanticide found a significant increase in common psychiatric disorders and financial stress among the mothers. Neonaticide (murder of the infant in the first day of life) generally is not related to PPP because PPP usually does not begin until after the day of delivery.6

Among women who develop psychiatric illness, homicidal ideation is more frequent in those with a perinatal onset of psychopathology.7 Infanticidal ideas and behavior are associated with psychotic ideas about the infant.8 Suicide is the cause of up to 20% of postpartum deaths.9

Table 1

Motives for infanticide: Mental illness or something else?

MotivesExamples
Likely related to postpartum psychosis or depression
AltruisticA depressed or psychotic mother may believe she is sending her baby to heaven to prevent suffering on earth
A suicidal mother may kill her infant along with herself rather than leave the child alone
Acutely psychoticA mother kills her baby for no comprehensible reason, such as in response to command hallucinations or the confusion of delirium
Rarely related to postpartum psychosis
Fatal maltreatment‘Battered child’ syndrome is the most common cause of infanticide; death often occurs after chronic abuse or neglect
A minority of perpetrators are psychotic; a mother out of touch with reality may have difficulty providing for her infant’s needs
Not likely related to postpartum psychosis
Unwanted childParent does not want child because of inconvenience or out-of-wedlock birth
Spouse revengeMurder of a child to cause emotional suffering for the other parent is the least frequent motive for infanticide
Source: Reference 4

The bipolar connection

Many factors can elevate the risk of PPP, including sleep deprivation in susceptible women, the hormonal shifts after birth, and psychiatric comorbidity (Table 2). Nearly three-fourths (>72%) of mothers with PPP have bipolar disorder or schizoaffective disorder, whereas 12% have schizophrenia.10 Some authors consider PPP to be bipolar disorder until proven otherwise. Mothers with a history of bipolar disorder or PPP have a 100-fold increase in rates of psychiatric hospitalization in the postpartum period.11

PPP is not categorized as a distinct disorder in DSM-IV-TR, and lack of a consistent terminology has led to differing definitions. Brief psychotic disorder, psychotic disorder not otherwise specified, and affective disorders are sometimes proffered.12 Some DSM disorders permit the specifier “with postpartum onset” if the symptoms occur in mothers within 4 weeks of birth.

Presentation. PPP is relatively rare, occurring at a rate of 1 to 3 cases per 1,000 births. Symptoms often have an abrupt onset, within days to weeks of delivery.10 In at least one-half of cases, symptoms begin by the third postpartum day,13 when many mothers have been discharged home and may be solely responsible for their infants.

Symptoms include confusion, bizarre behaviors, hallucinations (including rarer types such as tactile and olfactory), mood lability (ranging from euphoria to depression), decreased need for sleep or insomnia, restlessness, agitation, disorganized thinking, and bizarre delusions of relatively rapid onset.13 One mother might believe God wants her baby to be sacrificed as the second coming of the Messiah, a second may believe she has special powers, and a third that her baby is defective.

 

 

Table 2

Postpartum psychosis: Risk factors supported by evidence

Sleep deprivation in susceptible women
Hormonal shifts after birth (primarily the rapid drop in estrogen)
Psychosocial stressors such as marital problems, older age, single motherhood, lower socioeconomic status
Bipolar disorder or schizoaffective disorder
Past history of postpartum psychosis
Family history of postpartum psychosis
Previous psychiatric hospitalization, especially during the prenatal period for a bipolar or psychotic condition
Menstruation or cessation of lactation
Obstetric factors that can cause a small increase in relative risk:
  • first pregnancy
  • delivery complications
  • preterm birth
  • acute Caesarean section
  • long duration of labor
Source: For bibliographic citations

Differential diagnosis

When evaluating a postpartum woman with psychotic symptoms, stay in contact with her obstetrician and the child’s pediatrician. Rule out delirium and organic causes of the mother’s symptoms (Box).11

The psychiatric differential diagnosis includes “baby blues”—mild, transient mood swings, sadness, irritability, anxiety, and insomnia that most new mothers experience in the first postpartum week. Schizophrenia’s delusional thinking and hallucinations have a more gradual onset, compared with those of postpartum psychosis.

Postpartum depression (PPD) occurs in approximately 10% to 15% of new mothers.14 Depressive symptoms occur within weeks to months after delivery and often coexist with anxious symptoms. Some women with severe depression may present with psychotic symptoms. A mother may experience insomnia, sometimes not being able to sleep when the baby is sleeping. She may lack interest in caring for her baby and experience difficulty bonding.

At times it can be difficult to distinguish PPD from PPP. When evaluating a mother who is referred for “postpartum depression,” consider PPP in the differential diagnosis. A woman with PPD or PPP may report depressed mood, but in PPP this symptom usually is related to rapid mood changes. Other clinical features that point toward PPP are abnormal hallucinations (such as olfactory or tactile), hypomanic or mixed mood symptoms, and confusion.

Box

Medical workup in differential diagnosis of postpartum psychosis

When evaluating a postpartum woman with psychotic symptoms, stay in contact with her obstetrician and the child’s pediatrician. Rule out delirium and organic causes of the mother’s symptoms, giving special consideration to metabolic, neurologic, cardiovascular, infectious, and substance- or medication-induced origins. The extensive differential diagnosis includes:

  • thyroiditis
  • tumor
  • CNS infection
  • head injury
  • embolism
  • eclampsia
  • substance withdrawal
  • medication-induced (such as corticosteroids)
  • electrolyte anomalies
  • anoxia
  • vitamin B12 deficiency.11
Suicidal thoughts or thoughts of harming the infant may be present in either PPD or PPP. Both elevate the risk of infanticide; one study found that 41 out of 100 depressed mothers acknowledged having thoughts of harming their infants.15

Psychosis vs OCD. Psychotic thinking and behaviors also must be differentiated from obsessive thoughts and compulsions.10,16 Obsessive compulsive disorder (OCD) may be exacerbated or emerge for the first time during the perinatal period.17

In postpartum OCD, women may experience intrusive thoughts of accidental or purposeful harm to their baby. As opposed to women with PPP, mothers with OCD are not out of touch with reality and their thoughts are ego-dystonic.17 When these mothers have thoughts of their infants being harmed, they realize that these thoughts are not plans but fears and they try to avoid the thoughts.

Preventing PPP

Bipolar disorder is one of the most difficult disorders to treat during pregnancy because the serious risks of untreated illness must be balanced against the potential teratogenic risk of medications. Nevertheless, proactively managing bipolar disorder during pregnancy may reduce the risk of PPP.10

Closely monitor women with a history of bipolar disorder or PPP. During pregnancy, counsel them—and their partners—to:

  • anticipate that depressive or psychotic symptoms could develop within days after delivery18
  • seek treatment immediately if this occurs.
Some women will prefer to remain off medication during the first trimester—which is critical in organogenesis—and then restart medication later in pregnancy. This approach is not without risks, however (see Related Resources).

Postpartum medication. Whether or not a woman with bipolar disorder takes medication during pregnancy, consider treatment with mood stabilizers or atypical antipsychotics in the postpartum to prevent PPP (Table 3). Evidence is limited, but a search of PubMed found 1 study in which prophylactic lithium was given late in the third trimester or immediately after delivery to 21 women with a history of bipolar disorder or PPP. Only 2 patients had a psychotic recurrence while on prophylactic lithium; 1 unexplained stillbirth occurred.19

A retrospective study examined the course of women with bipolar disorder, some of whom were given prophylactic mood stabilizers immediately in the postpartum. One of 14 who received antimanic agents relapsed within the first 3 months postpartum, compared with 8 of 13 who were not so treated.18

 

 

Compared with antiepileptics, less information is available about the use of atypical antipsychotics in pregnancy and lactation. Antipsychotics’ potential advantage in women at risk for PPP is that these agents may help prevent or treat both manic and psychotic symptoms.

In a small, naturalistic, prospective study, 11 women at risk for PPP received olanzapine alone or with an antidepressant or mood stabilizer for at least 4 weeks after delivery. Two (18%) experienced a postpartum mood episode, compared with 8 (57%) of 14 other at-risk women who received antidepressants, mood stabilizers, or no medication.20

Breast-feeding. Consider treatment effects on lactation and discuss this with the mother and the baby’s pediatrician, when possible. For useful reviews of risks and benefits of mood stabilizers and antipsychotics during breast-feeding, see Related Resources.

When you discuss breast-feeding, consider possible risks to the neonate as well as potential sleep interruption for the mother. If a mother has a supportive partner, the partner might be put in charge of night-time feedings in a routine combining breast-feeding and bottle-feeding. In some cases you may need to recommend cessation of lactation.21

Table 3

Treating postpartum psychosis: Consider 3 components

ComponentRecommendations
Hospitalization vs home careHospitalize in most cases because of emergent severe symptoms and fluctuating course; base decision on risk evaluation/safety issues for patient and infant
After discharge, visiting nurses are useful to help monitor the mother’s condition at home
PsychoeducationEducate patient, family, and social support network; address risks to mother and infant and risks in future pregnancies
MedicationWhen prescribing mood stabilizers and/or antipsychotics, consider:
  • whether mother is breast-feeding (discuss with patient, family, and pediatrician)
  • maternal side effects, including sedation

Managing PPP

Early symptoms. Because of its severity and rapid evolution, PPP often presents as a psychiatric emergency. Monitor atrisk patients’ sleep patterns and mood for early signs of psychosis.22 Watch especially for hypomanic symptoms such as elevated or mixed mood and decreased judgment, which are common early in PPP.13

A mother with few signs of abnormal mood, good social support, and close follow-up may potentially be safely managed as an outpatient. Initial evaluation and management of PPP usually requires hospitalization, however, because of the risks of suicide, infanticide, and child maltreatment.23

Hospitalization. Mother-infant bonding is important, but safety is paramount if a mother is psychotic—especially if she is experiencing psychotic thoughts about her infant. If possible, the infant should remain with family members during the mother’s hospitalization. Supervised mother-infant visits are often arranged, as appropriate.

Mood-stabilizing medications, including antipsychotics, are mainstays of treatment.24 In some cases, conventional antipsychotics such as haloperidol may be useful because of a lower risk of weight gain or of sedation that could impair a mother’s ability to respond to her infant. Electroconvulsive therapy often yields rapid symptomatic improvement for mothers with postpartum mood or psychotic symptoms.25

During the mother’s hospitalization, encourage the staff to be supportive and convey hopefulness.26 In an interview study, women who had been treated for PPP said they experienced anger and frustration while hospitalized because they believed that they and their families received inadequate information and support.27

Discharge planning. Assuming that the mother adheres to prescribed treatment, discharge may occur within 1 week. Plan discharge arrangements carefully (Table 4).28 A team approach can be very useful within the outpatient clinic. In the model of the Perinatal Psychiatry Clinic of Connections in suburban Cleveland, OH, the mother’s treatment team includes perinatal psychiatrists, nurses, counsellors, case managers (who do home visits), and peer counselors.

Outpatient civil commitment, in which patients are mandated to accept treatment, is an option in some jurisdictions and could help ensure that patients receive treatment consistently.

Table 4

Discharge planning for safety of mother and infant

Notify child protective services (CPS) depending on the risk to the child. Case-by-case review is needed to assess whether the infant should be removed. CPS may put in place a plan for safety, short of removal. The plan may require that the woman continue psychiatric care
Meet with the patient and family to discuss her diagnosis, the risks, the importance of continued medication adherence, and the need for family or social supports to assist with child care
Consider engaging visiting nurses or doulas to provide help and support at home
Schedule frequent outpatient appointments for the mother after discharge
Consider family therapy after the mother has improved because of her risk for affective episodes outside the postpartum28
 

 

Related resources

  • Altshuler L, Richards M, Yonkers K. Treating bipolar disorder during pregnancy. Current Psychiatry. 2003;2(7):14-26. www.CurrentPsychiatry.com.
  • Gentile S. Infant safety with antipsychotic therapy in breastfeeding: a systematic review. J Clin Psychiatry. 2008;69(4):666-673.
  • Miller LJ. Postpartum mood disorders. Washington, DC: American Psychiatric Publishing, Inc; 1999.
  • Stowe ZN. The use of mood stabilizers during breastfeeding. J Clin Psychiatry. 2007;68(suppl 9):22-28.
  • Toxicology Data Network (Toxnet). Literature on reproductive risks associated with psychotropics. National Library of Medicine. http://toxnet.nlm.nih.gov.
Drug brand names

  • Haloperidol • Haldol
  • Lithium • various
  • Olanzapine • Zyprexa
Disclosures

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

Acknowledgement

Dr. Resnick, a forensic psychiatrist and coauthor of this article, testified for the defense in both trials of Andrea Yates.

References

1. Resnick PJ. The Andrea Yates case: insanity on trial. Cleveland State Law Review. 2007;55(2):147-156.

2. Altshuler LL, Hendrick V, Cohen LS. Course of mood and anxiety disorders during pregnancy and the postpartum period. J Clin Psychiatry. 1998;59(suppl. 2):29-33.

3. Knops GG. Postpartum mood disorders. Postgrad Med. 1993;93:103-116.

4. Resnick PJ. Child murder by parents: a psychiatric review of filicide. Am J Psychiatry. 1969;126:73-82.

5. Friedman SH, Horwitz SM, Resnick PJ. Child murder by mothers: a critical analysis of the current state of knowledge and a research agenda. Am J Psychiatry. 2005;162:1578-1587.

6. Friedman SH, Resnick PJ. Neonaticide: phenomenology and considerations for prevention. Int J Law Psychiatry. In press.

7. Wisner K, Peindl K, Hanusa BH. Symptomatology of affective and psychotic illnesses related to childbearing. J Affect Disord. 1994;30:77-87.

8. Chandra PS, Venkatasubramanian G, Thomas T. Infanticidal ideas and infanticidal behaviour in Indian women with severe postpartum psychiatric disorders. J Nerv Ment Dis. 2002;190(7):457-461.

9. Lindahl V, Pearson JL, Colpe L. Prevalence of suicidality during pregnancy and the postpartum. Arch Womens Ment Health. 2005;8(2):77-87.

10. Sit D, Rothschild AJ, Wisner KL. A review of postpartum psychosis. J Women’s Health. 2006;15(4):352-368.

11. Attia E, Downey J, Oberman M. Postpartum psychoses. In: Miller LJ, ed. Postpartum mood disorders. Washington, DC: American Psychiatric Publishing Inc.; 1999:99-117.

12. Diagnostic and Statistical Manual of Mental Disorders, 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.

13. Heron J, McGuinness M, Blackmore ER, et al. Early postpartum symptoms in puerperal psychosis. BJOG. 2008;115(3):348-353.

14. Meltzer-Brody S, Payne J, Rubinow D. Postpartum depression: what to tell patients who breast-feed. Current Psychiatry. 2008;7(5):87-95.

15. Jennings KD, Ross S, Popper S, et al. Thoughts of harming infants in depressed and nondepressed mothers. J Affect Disord. 1999;54:21-28.

16. Wisner KL, Gracious BL, Piontek CM, et al. Postpartum disorders: phenomenology, treatment approaches, and relationship to infanticide. In: Spinelli MG, ed. Infanticide: psychosocial and legal perspectives on mothers who kill. Washington, DC: American Psychiatric Publishing, Inc.; 2003.

17. Fairbrother N, Abramowitz JS. New parenthood as a risk factors for the development of obsessional problems. Behav Res Ther. 2007;45(9):2155-2163.

18. Cohen LS, Sichel DA, Robertson LM, et al. Postpartum prophylaxis for women with bipolar disorder. Am J Psychiatry. 1995;152(11):1641-1645.

19. Stewart DE, Klompenhouwer JL, Kendell RE, et al. Prophylactic lithium in puerperal psychosis. Br J Psychiatry. 1991;158:393-397.

20. Sharma V, Smith A, Mazmanian D. Olanzapine in the prevention of postpartum psychosis and mood episodes in bipolar disorder. Bipolar Disord. 2006;8(4):400-404.

21. Pfuhlmann B, Stoeber G, Beckmann H. Postpartum psychoses: prognosis, risk factors, and treatment. Curr Psychiatry Rep. 2002;4(3):185-190.

22. Sharma V, Mazmanian D. Sleep loss and postpartum psychosis. Bipolar Disord. 2003;5(2):98-105.

23. Lindahl V, Pearson JL, Colpe L. Prevalence of suicidality during pregnancy and the postpartum. Arch Womens Ment Health. 2005;8(2):77-87.

24. Connell M. The postpartum psychosis defense and feminism: more or less justice for women? Case Western Reserve Law Review. 2002;53:143.-

25. Forray A, Ostroff RB. The use of electroconvulsive therapy in postpartum affective disorders. J ECT. 2007;23(3):188-193.

26. Engqvist I, Nilsson A, Nilsson K, et al. Strategies in caring for women with postpartum psychosis—an interview study with psychiatric nurses. J Clin Nurs. 2007;16(7):1333-1342.

27. Robertson E, Lyons A. Living with puerperal psychosis: a qualitative analysis. Psychol Psychother. 2003;76(4):411-431.

28. Robertson E, Jones I, Haque S, et al. Risk of puerperal and non-puerperal recurrence of illness following bipolar affective puerperal (post-partum) psychosis. Br J Psychiatry. 2005;186:258-259.

References

1. Resnick PJ. The Andrea Yates case: insanity on trial. Cleveland State Law Review. 2007;55(2):147-156.

2. Altshuler LL, Hendrick V, Cohen LS. Course of mood and anxiety disorders during pregnancy and the postpartum period. J Clin Psychiatry. 1998;59(suppl. 2):29-33.

3. Knops GG. Postpartum mood disorders. Postgrad Med. 1993;93:103-116.

4. Resnick PJ. Child murder by parents: a psychiatric review of filicide. Am J Psychiatry. 1969;126:73-82.

5. Friedman SH, Horwitz SM, Resnick PJ. Child murder by mothers: a critical analysis of the current state of knowledge and a research agenda. Am J Psychiatry. 2005;162:1578-1587.

6. Friedman SH, Resnick PJ. Neonaticide: phenomenology and considerations for prevention. Int J Law Psychiatry. In press.

7. Wisner K, Peindl K, Hanusa BH. Symptomatology of affective and psychotic illnesses related to childbearing. J Affect Disord. 1994;30:77-87.

8. Chandra PS, Venkatasubramanian G, Thomas T. Infanticidal ideas and infanticidal behaviour in Indian women with severe postpartum psychiatric disorders. J Nerv Ment Dis. 2002;190(7):457-461.

9. Lindahl V, Pearson JL, Colpe L. Prevalence of suicidality during pregnancy and the postpartum. Arch Womens Ment Health. 2005;8(2):77-87.

10. Sit D, Rothschild AJ, Wisner KL. A review of postpartum psychosis. J Women’s Health. 2006;15(4):352-368.

11. Attia E, Downey J, Oberman M. Postpartum psychoses. In: Miller LJ, ed. Postpartum mood disorders. Washington, DC: American Psychiatric Publishing Inc.; 1999:99-117.

12. Diagnostic and Statistical Manual of Mental Disorders, 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.

13. Heron J, McGuinness M, Blackmore ER, et al. Early postpartum symptoms in puerperal psychosis. BJOG. 2008;115(3):348-353.

14. Meltzer-Brody S, Payne J, Rubinow D. Postpartum depression: what to tell patients who breast-feed. Current Psychiatry. 2008;7(5):87-95.

15. Jennings KD, Ross S, Popper S, et al. Thoughts of harming infants in depressed and nondepressed mothers. J Affect Disord. 1999;54:21-28.

16. Wisner KL, Gracious BL, Piontek CM, et al. Postpartum disorders: phenomenology, treatment approaches, and relationship to infanticide. In: Spinelli MG, ed. Infanticide: psychosocial and legal perspectives on mothers who kill. Washington, DC: American Psychiatric Publishing, Inc.; 2003.

17. Fairbrother N, Abramowitz JS. New parenthood as a risk factors for the development of obsessional problems. Behav Res Ther. 2007;45(9):2155-2163.

18. Cohen LS, Sichel DA, Robertson LM, et al. Postpartum prophylaxis for women with bipolar disorder. Am J Psychiatry. 1995;152(11):1641-1645.

19. Stewart DE, Klompenhouwer JL, Kendell RE, et al. Prophylactic lithium in puerperal psychosis. Br J Psychiatry. 1991;158:393-397.

20. Sharma V, Smith A, Mazmanian D. Olanzapine in the prevention of postpartum psychosis and mood episodes in bipolar disorder. Bipolar Disord. 2006;8(4):400-404.

21. Pfuhlmann B, Stoeber G, Beckmann H. Postpartum psychoses: prognosis, risk factors, and treatment. Curr Psychiatry Rep. 2002;4(3):185-190.

22. Sharma V, Mazmanian D. Sleep loss and postpartum psychosis. Bipolar Disord. 2003;5(2):98-105.

23. Lindahl V, Pearson JL, Colpe L. Prevalence of suicidality during pregnancy and the postpartum. Arch Womens Ment Health. 2005;8(2):77-87.

24. Connell M. The postpartum psychosis defense and feminism: more or less justice for women? Case Western Reserve Law Review. 2002;53:143.-

25. Forray A, Ostroff RB. The use of electroconvulsive therapy in postpartum affective disorders. J ECT. 2007;23(3):188-193.

26. Engqvist I, Nilsson A, Nilsson K, et al. Strategies in caring for women with postpartum psychosis—an interview study with psychiatric nurses. J Clin Nurs. 2007;16(7):1333-1342.

27. Robertson E, Lyons A. Living with puerperal psychosis: a qualitative analysis. Psychol Psychother. 2003;76(4):411-431.

28. Robertson E, Jones I, Haque S, et al. Risk of puerperal and non-puerperal recurrence of illness following bipolar affective puerperal (post-partum) psychosis. Br J Psychiatry. 2005;186:258-259.

Issue
Current Psychiatry - 08(02)
Issue
Current Psychiatry - 08(02)
Page Number
40-45
Page Number
40-45
Publications
Publications
Article Type
Display Headline
Postpartum psychosis: Strategies to protect infant and mother from harm
Display Headline
Postpartum psychosis: Strategies to protect infant and mother from harm
Legacy Keywords
postpartum psychosis; PPP; infanticide; Andrea Yates; postpartum depression; Susan Hatters Friedman; Phillip J Resnick; Miriam B rosenthal
Legacy Keywords
postpartum psychosis; PPP; infanticide; Andrea Yates; postpartum depression; Susan Hatters Friedman; Phillip J Resnick; Miriam B rosenthal
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Controversies in bipolar disorder: Trust evidence or experience?

Article Type
Changed
Wed, 05/08/2019 - 15:41
Display Headline
Controversies in bipolar disorder: Trust evidence or experience?

Today’s buzzword in health care is evidence-based medicine. Most clinicians would agree that evidence from clinical research should guide decisions about treating bipolar disorder. In theory, randomized controlled trials should tell us how to manage bipolar patients and achieve therapeutic success. page 40.)

We rarely have encountered a patient with postpartum depression or psychosis who does not have a history of (often undiagnosed and untreated) recurrent mood episodes. For most of these patients, a mood stabilizer may be a better choice than an antidepressant.

The role of thyroid hormones

Adding a thyroid hormone—usually liothyronine—to an antidepressant has been demonstrated to accelerate, page 47.)

Atypical depression and the bipolar spectrum

Depressive episodes are considered either “typical” (a category that includes melancholic depression—in DSM-IV-TR, major depression with melancholic features) or “atypical” (in DSM-IV-TR, major depression with atypical features). Atypical features were originally associated with response to monoamine oxidase inhibitor antidepressants, whereas non atypical depression was thought more likely to respond to tricyclic antidepressants.34 The depression of bipolar disorder is usually atypical ( Box 4 ), especially in patients with softer variants of the illness.35

 

We believe that depressed patients with atypical symptoms aggregate into groups according to the presence, severity, and character of interdepressive manic or hypomanic episodes. Some patients experience recurrent depressive episodes with intervening euthymia (recurrent major depression), whereas others experience depressive episodes punctuated by brief subthreshold hypomanic episodes. Patients in these groups occasionally tolerate or even benefit from cautiously managed antidepressant monotherapy. Patients with atypical depressive episodes alternating with frank hypomanic, manic, mixed, or manic-psychotic episodes usually require a mood stabilizer and may benefit from cotreatment with an atypical antipsychotic.

Akiskol and Benazzi35 suggest that atypical depression may be a subtype of the bipolar spectrum. Our experience suggests that the bipolar spectrum is a continuum of degrees of risk for mood instability in persons with recurrent atypical depression.

Box 4

 

Atypical depression: Who sees ‘leaden paralysis’?

DSM-IV-TR defines atypical depression as depression characterized by mood reactivity and at least 2 of these 4 features:

 

  • hypersomnia
  • increased appetite or weight gain
  • leaden paralysis
  • sensitivity to interpersonal rejection.

The term ‘hypersomnia’ is misleading. Many of these patients do not sleep excessively because work or school attendance prevents oversleeping. Instead, they experience an increased sleep requirement manifested by difficulty getting up in the morning and increased daytime sleepiness.

Increased appetite and weight gain (hyperphagia) often are present, but almost as often our patients report no change in appetite or weight or even anorexia and weight loss.

We rarely see a condition one would term ‘leaden paralysis.’ We also find that ‘sensitivity to interpersonal rejection’ is too narrow a construct. Our patients with atypical depression experience increased sensitivity to every stressor in their lives—work, school, family, and social stressors—not just interpersonal rejection.

Related resources

 

  • Lieber AL. Bipolar spectrum disorder: an overview of the soft bipolar spectrum. www.psycom.net/depression.central.lieber.html.
  • Phelps J. Why am I still depressed? Recognizing and managing the ups and downs of bipolar II and soft bipolar disorder. www.psycheducation.org.
  • Maier T. Evidence-based psychiatry: understanding the limitations of a method. J Eval Clin Pract. 2006;12(3):325.

Drug brand names

 

  • Liothyronine • Cytomel
  • Sertraline • Zoloft

Disclosure

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

References

 

1. Goldberg JF. What constitutes evidence-based pharmacotherapy for bipolar disorder? Part 1: First-line treatments. J Clin Psychiatry. 2007;68:1982-1983.

2. Goldberg JF. What constitutes evidence-based pharmacotherapy for bipolar disorder? Part 2: Complex presentations and clinical context. J Clin Psychiatry. 2008;69:495-496.

3. Levine R, Fink M. Why evidence-based medicine cannot be applied to psychiatry. Psychiatric Times. 2008;25(4):10.

4. Akiskol HS, Benazzi F. The DSM-IV and ICD-10 categories of recurrent [major] depressive and bipolar II disorders: evidence that they lie on a dimensional spectrum. J Affect Disord. 2006;92:45-54.

5. Goodwin FK, Jamison KR. Manic-depressive illness: bipolar disorders and recurrent depression. 2nd ed. New York, NY: Oxford University Press; 2007:3–27.

6. Hirschfeld RMA, Lewis L, Vornik L. Perceptions and impact of bipolar disorder: how far have we really come? Results of the National Depressive and Manic-Depressive Association 2000 survey of individuals with bipolar disorder. J Clin Psychiatry. 2003;64(2):161-167.

7. Blanco C, Laje G, Olfson M, et al. Trends in the treatment of bipolar disorder by outpatient psychiatrists. Am J Psychiatry. 2002;159:1005-1010.

8. Ghaemi SN, Lenox MS, Baldessarini RJ. Effectiveness and safety of long-term antidepressant treatment in bipolar disorder. J Clin Psychiatry. 2001;62:565-569.

9. Ghaemi SN, Boiman EE, Goodwin FK. Diagnosing bipolar disorder and the effect of antidepressants: a naturalistic study. J Clin Psychiatry. 2000;61:804-808.

10. Gijsman HF, Geddes JR, Rendell JM, et al. Antidepressants for bipolar depression: a systematic review of randomized, controlled trials. Am J Psychiatry. 2005;161:1537-1547.

11. Ghaemi SN, Sachs GS, Chiou AM, et al. Is bipolar disorder still underdiagnosed? Are antidepressants overutilized? J Affect Disord. 1999;52:134-144.

12. Sachs GS, Nierenberg AA, Calabrese JR, et al. Effectiveness of adjunctive antidepressant treatment for bipolar depression. N Engl J Med. 2007;356:1711-1722.

13. Altshuler L, Suppes T, Black D, et al. Impact of antidepressant discontinuation after acute bipolar depression remission on rates of depressive relapse at 1-year follow-up. Am J Psychiatry. 2003;160:1252-1262.

14. Akiskol HS. Developmental pathways to bipolarity: are juvenile-onset depressions pre-bipolar? J Am Acad Child Adolesc Psychiatry. 1995;34(6):754-763.

15. Geller B, Zimmerman B, Williams M, et al. Bipolar disorder at prospective follow-up of adults who had prepubertal major depressive disorder. Am J Psychiatry. 2001;158:125-127.

16. Food and Drug Administration: Center for Drug Evaluation and Research. Revisions to product labeling. Available at: http://www.FDA.gov/cder/drug/antidepressants/default.htm. Accessed January 12, 2009.

17. McElroy S, Strakowski S, West S, et al. Phenomenology of adolescent and adult mania in hospitalized patients with bipolar disorder. Am J Psychiatry. 1997;154:44-49.

18. Olfson M, Marcus SC. A case-control study of antidepressants and attempted suicide during early phase treatment of major depressive episodes. J Clin Psychiatry. 2008;69:425-432.

19. Keck PE, Jr, McElroy SL, Havens JR, et al. Psychosis in bipolar disorder: phenomenology and impact on morbidity and course of illness. Compr Psychiatry. 2003;44:263-269.

20. Jones I, Craddock N. Familiarity of the puerperal trigger in bipolar disorder: results of a family study. Am J Psychiatry. 2001;158:913-917.

21. Chaudron LH, Pies RW. The relationship between postpartum psychosis and bipolar disorder: a review. J Clin Psychiatry. 2003;64:1284-1292.

22. Wisner KL, Peindl KS, Hanusa BH. Psychiatric episodes in women and young children. J Affect Disord. 1995;34:1-11.

23. Sharma V. A cautionary note on the use of antidepressants in postpartum depression. Bipolar Disord. 2006;8:411-414.

24. O’Malley S. “Are you there alone?” The unspeakable crime of Andrea Yates. New York, NY: Simon and Schuster; 2004.

25. Altshuler LL, Bauer M, Frye MA, et al. Does thyroid supplementation accelerate tricyclic antidepressant response? A review and meta-analysis of the literature. Am J Psychiatry. 2001;158:1617-1622.

26. Joffe RT. The use of thyroid supplements to augment antidepressant medication. J Clin Psychiatry. 2008;59:26-29.

27. Cooper-Kazaz R, Apter JT, Cohen R, et al. Combined treatment with sertraline and liothyronine in major depression: a randomized, double-blind, placebo-controlled trial. Arch Gen Psychiatry. 2007;64:679-688.

28. Gold MS, Pottash AL, Extein I. Hypothyroidism and depression: evidence from complete thyroid function evaluation. JAMA. 1981;245:28-31.

29. Kupka RW, Nolen WA, Post RM, et al. High rate of autoimmune thyroiditis in bipolar disorder: lack of association with lithium exposure. Biol Psychiatry. 2002;51:305-311.

30. Szuba MP, Amsterdam JD. Rapid antidepressant response after nocturnal TRH administration in patients with bipolar I and bipolar type II major depression. J Clin Psychopharmacol. 2005;25:325-330.

31. Extein I, Pottash AL, Gold MS. Does subclinical hypothyroidism predispose to tricyclic-induced rapid mood cycles? J Clin Psychiatry. 1982;43:32-33.

32. American Association of Clinical Endocrinologists. Medical guidelines for clinical practice for the evaluation and treatment of hyperthyroidism and hypothyroidism. Endocr Prac. 2002;8:457-469.

33. El-Mallakh RS, Karippott A. Antidepressant-associated chronic irritable dysphoria (ACID) in bipolar disorder. J Affect Disord. 2005;84:267-272.

34. Henkl V, Mergl R, Antje-Kathrin A, et al. Treatment of depression with atypical features: a meta-analytic approach. Psychiatry Res. 2006;141(1):89-101.

35. Perugi G, Akiskal HS, Lattanzi D, et al. The high prevalence of “soft” bipolar (II) features in atypical depression. Compr Psychiatry. 1998;39(2):63-71.

Article PDF
Author and Disclosure Information

 

Gary E. Miller, MD

Clinical professor of psychiatry, University of Texas Health Science Center, Houston, TX
Richard L. Noel, MD
Assistant clinical professor of psychiatry, University of Texas Health Science Center, Houston, TX

Comment on this article


Hear Dr. Miller discuss this article
 

Issue
Current Psychiatry - 08(02)
Publications
Topics
Page Number
27-28,31-33,39
Legacy Keywords
bipolar disorder; evidence-based medicine; bipolar spectrum; subthreshold hypomania; manic switches; antidepressant monotherapy; Gary E Miller; Richard L noel
Sections
Author and Disclosure Information

 

Gary E. Miller, MD

Clinical professor of psychiatry, University of Texas Health Science Center, Houston, TX
Richard L. Noel, MD
Assistant clinical professor of psychiatry, University of Texas Health Science Center, Houston, TX

Comment on this article


Hear Dr. Miller discuss this article
 

Author and Disclosure Information

 

Gary E. Miller, MD

Clinical professor of psychiatry, University of Texas Health Science Center, Houston, TX
Richard L. Noel, MD
Assistant clinical professor of psychiatry, University of Texas Health Science Center, Houston, TX

Comment on this article


Hear Dr. Miller discuss this article
 

Article PDF
Article PDF

Today’s buzzword in health care is evidence-based medicine. Most clinicians would agree that evidence from clinical research should guide decisions about treating bipolar disorder. In theory, randomized controlled trials should tell us how to manage bipolar patients and achieve therapeutic success. page 40.)

We rarely have encountered a patient with postpartum depression or psychosis who does not have a history of (often undiagnosed and untreated) recurrent mood episodes. For most of these patients, a mood stabilizer may be a better choice than an antidepressant.

The role of thyroid hormones

Adding a thyroid hormone—usually liothyronine—to an antidepressant has been demonstrated to accelerate, page 47.)

Atypical depression and the bipolar spectrum

Depressive episodes are considered either “typical” (a category that includes melancholic depression—in DSM-IV-TR, major depression with melancholic features) or “atypical” (in DSM-IV-TR, major depression with atypical features). Atypical features were originally associated with response to monoamine oxidase inhibitor antidepressants, whereas non atypical depression was thought more likely to respond to tricyclic antidepressants.34 The depression of bipolar disorder is usually atypical ( Box 4 ), especially in patients with softer variants of the illness.35

 

We believe that depressed patients with atypical symptoms aggregate into groups according to the presence, severity, and character of interdepressive manic or hypomanic episodes. Some patients experience recurrent depressive episodes with intervening euthymia (recurrent major depression), whereas others experience depressive episodes punctuated by brief subthreshold hypomanic episodes. Patients in these groups occasionally tolerate or even benefit from cautiously managed antidepressant monotherapy. Patients with atypical depressive episodes alternating with frank hypomanic, manic, mixed, or manic-psychotic episodes usually require a mood stabilizer and may benefit from cotreatment with an atypical antipsychotic.

Akiskol and Benazzi35 suggest that atypical depression may be a subtype of the bipolar spectrum. Our experience suggests that the bipolar spectrum is a continuum of degrees of risk for mood instability in persons with recurrent atypical depression.

Box 4

 

Atypical depression: Who sees ‘leaden paralysis’?

DSM-IV-TR defines atypical depression as depression characterized by mood reactivity and at least 2 of these 4 features:

 

  • hypersomnia
  • increased appetite or weight gain
  • leaden paralysis
  • sensitivity to interpersonal rejection.

The term ‘hypersomnia’ is misleading. Many of these patients do not sleep excessively because work or school attendance prevents oversleeping. Instead, they experience an increased sleep requirement manifested by difficulty getting up in the morning and increased daytime sleepiness.

Increased appetite and weight gain (hyperphagia) often are present, but almost as often our patients report no change in appetite or weight or even anorexia and weight loss.

We rarely see a condition one would term ‘leaden paralysis.’ We also find that ‘sensitivity to interpersonal rejection’ is too narrow a construct. Our patients with atypical depression experience increased sensitivity to every stressor in their lives—work, school, family, and social stressors—not just interpersonal rejection.

Related resources

 

  • Lieber AL. Bipolar spectrum disorder: an overview of the soft bipolar spectrum. www.psycom.net/depression.central.lieber.html.
  • Phelps J. Why am I still depressed? Recognizing and managing the ups and downs of bipolar II and soft bipolar disorder. www.psycheducation.org.
  • Maier T. Evidence-based psychiatry: understanding the limitations of a method. J Eval Clin Pract. 2006;12(3):325.

Drug brand names

 

  • Liothyronine • Cytomel
  • Sertraline • Zoloft

Disclosure

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

Today’s buzzword in health care is evidence-based medicine. Most clinicians would agree that evidence from clinical research should guide decisions about treating bipolar disorder. In theory, randomized controlled trials should tell us how to manage bipolar patients and achieve therapeutic success. page 40.)

We rarely have encountered a patient with postpartum depression or psychosis who does not have a history of (often undiagnosed and untreated) recurrent mood episodes. For most of these patients, a mood stabilizer may be a better choice than an antidepressant.

The role of thyroid hormones

Adding a thyroid hormone—usually liothyronine—to an antidepressant has been demonstrated to accelerate, page 47.)

Atypical depression and the bipolar spectrum

Depressive episodes are considered either “typical” (a category that includes melancholic depression—in DSM-IV-TR, major depression with melancholic features) or “atypical” (in DSM-IV-TR, major depression with atypical features). Atypical features were originally associated with response to monoamine oxidase inhibitor antidepressants, whereas non atypical depression was thought more likely to respond to tricyclic antidepressants.34 The depression of bipolar disorder is usually atypical ( Box 4 ), especially in patients with softer variants of the illness.35

 

We believe that depressed patients with atypical symptoms aggregate into groups according to the presence, severity, and character of interdepressive manic or hypomanic episodes. Some patients experience recurrent depressive episodes with intervening euthymia (recurrent major depression), whereas others experience depressive episodes punctuated by brief subthreshold hypomanic episodes. Patients in these groups occasionally tolerate or even benefit from cautiously managed antidepressant monotherapy. Patients with atypical depressive episodes alternating with frank hypomanic, manic, mixed, or manic-psychotic episodes usually require a mood stabilizer and may benefit from cotreatment with an atypical antipsychotic.

Akiskol and Benazzi35 suggest that atypical depression may be a subtype of the bipolar spectrum. Our experience suggests that the bipolar spectrum is a continuum of degrees of risk for mood instability in persons with recurrent atypical depression.

Box 4

 

Atypical depression: Who sees ‘leaden paralysis’?

DSM-IV-TR defines atypical depression as depression characterized by mood reactivity and at least 2 of these 4 features:

 

  • hypersomnia
  • increased appetite or weight gain
  • leaden paralysis
  • sensitivity to interpersonal rejection.

The term ‘hypersomnia’ is misleading. Many of these patients do not sleep excessively because work or school attendance prevents oversleeping. Instead, they experience an increased sleep requirement manifested by difficulty getting up in the morning and increased daytime sleepiness.

Increased appetite and weight gain (hyperphagia) often are present, but almost as often our patients report no change in appetite or weight or even anorexia and weight loss.

We rarely see a condition one would term ‘leaden paralysis.’ We also find that ‘sensitivity to interpersonal rejection’ is too narrow a construct. Our patients with atypical depression experience increased sensitivity to every stressor in their lives—work, school, family, and social stressors—not just interpersonal rejection.

Related resources

 

  • Lieber AL. Bipolar spectrum disorder: an overview of the soft bipolar spectrum. www.psycom.net/depression.central.lieber.html.
  • Phelps J. Why am I still depressed? Recognizing and managing the ups and downs of bipolar II and soft bipolar disorder. www.psycheducation.org.
  • Maier T. Evidence-based psychiatry: understanding the limitations of a method. J Eval Clin Pract. 2006;12(3):325.

Drug brand names

 

  • Liothyronine • Cytomel
  • Sertraline • Zoloft

Disclosure

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

References

 

1. Goldberg JF. What constitutes evidence-based pharmacotherapy for bipolar disorder? Part 1: First-line treatments. J Clin Psychiatry. 2007;68:1982-1983.

2. Goldberg JF. What constitutes evidence-based pharmacotherapy for bipolar disorder? Part 2: Complex presentations and clinical context. J Clin Psychiatry. 2008;69:495-496.

3. Levine R, Fink M. Why evidence-based medicine cannot be applied to psychiatry. Psychiatric Times. 2008;25(4):10.

4. Akiskol HS, Benazzi F. The DSM-IV and ICD-10 categories of recurrent [major] depressive and bipolar II disorders: evidence that they lie on a dimensional spectrum. J Affect Disord. 2006;92:45-54.

5. Goodwin FK, Jamison KR. Manic-depressive illness: bipolar disorders and recurrent depression. 2nd ed. New York, NY: Oxford University Press; 2007:3–27.

6. Hirschfeld RMA, Lewis L, Vornik L. Perceptions and impact of bipolar disorder: how far have we really come? Results of the National Depressive and Manic-Depressive Association 2000 survey of individuals with bipolar disorder. J Clin Psychiatry. 2003;64(2):161-167.

7. Blanco C, Laje G, Olfson M, et al. Trends in the treatment of bipolar disorder by outpatient psychiatrists. Am J Psychiatry. 2002;159:1005-1010.

8. Ghaemi SN, Lenox MS, Baldessarini RJ. Effectiveness and safety of long-term antidepressant treatment in bipolar disorder. J Clin Psychiatry. 2001;62:565-569.

9. Ghaemi SN, Boiman EE, Goodwin FK. Diagnosing bipolar disorder and the effect of antidepressants: a naturalistic study. J Clin Psychiatry. 2000;61:804-808.

10. Gijsman HF, Geddes JR, Rendell JM, et al. Antidepressants for bipolar depression: a systematic review of randomized, controlled trials. Am J Psychiatry. 2005;161:1537-1547.

11. Ghaemi SN, Sachs GS, Chiou AM, et al. Is bipolar disorder still underdiagnosed? Are antidepressants overutilized? J Affect Disord. 1999;52:134-144.

12. Sachs GS, Nierenberg AA, Calabrese JR, et al. Effectiveness of adjunctive antidepressant treatment for bipolar depression. N Engl J Med. 2007;356:1711-1722.

13. Altshuler L, Suppes T, Black D, et al. Impact of antidepressant discontinuation after acute bipolar depression remission on rates of depressive relapse at 1-year follow-up. Am J Psychiatry. 2003;160:1252-1262.

14. Akiskol HS. Developmental pathways to bipolarity: are juvenile-onset depressions pre-bipolar? J Am Acad Child Adolesc Psychiatry. 1995;34(6):754-763.

15. Geller B, Zimmerman B, Williams M, et al. Bipolar disorder at prospective follow-up of adults who had prepubertal major depressive disorder. Am J Psychiatry. 2001;158:125-127.

16. Food and Drug Administration: Center for Drug Evaluation and Research. Revisions to product labeling. Available at: http://www.FDA.gov/cder/drug/antidepressants/default.htm. Accessed January 12, 2009.

17. McElroy S, Strakowski S, West S, et al. Phenomenology of adolescent and adult mania in hospitalized patients with bipolar disorder. Am J Psychiatry. 1997;154:44-49.

18. Olfson M, Marcus SC. A case-control study of antidepressants and attempted suicide during early phase treatment of major depressive episodes. J Clin Psychiatry. 2008;69:425-432.

19. Keck PE, Jr, McElroy SL, Havens JR, et al. Psychosis in bipolar disorder: phenomenology and impact on morbidity and course of illness. Compr Psychiatry. 2003;44:263-269.

20. Jones I, Craddock N. Familiarity of the puerperal trigger in bipolar disorder: results of a family study. Am J Psychiatry. 2001;158:913-917.

21. Chaudron LH, Pies RW. The relationship between postpartum psychosis and bipolar disorder: a review. J Clin Psychiatry. 2003;64:1284-1292.

22. Wisner KL, Peindl KS, Hanusa BH. Psychiatric episodes in women and young children. J Affect Disord. 1995;34:1-11.

23. Sharma V. A cautionary note on the use of antidepressants in postpartum depression. Bipolar Disord. 2006;8:411-414.

24. O’Malley S. “Are you there alone?” The unspeakable crime of Andrea Yates. New York, NY: Simon and Schuster; 2004.

25. Altshuler LL, Bauer M, Frye MA, et al. Does thyroid supplementation accelerate tricyclic antidepressant response? A review and meta-analysis of the literature. Am J Psychiatry. 2001;158:1617-1622.

26. Joffe RT. The use of thyroid supplements to augment antidepressant medication. J Clin Psychiatry. 2008;59:26-29.

27. Cooper-Kazaz R, Apter JT, Cohen R, et al. Combined treatment with sertraline and liothyronine in major depression: a randomized, double-blind, placebo-controlled trial. Arch Gen Psychiatry. 2007;64:679-688.

28. Gold MS, Pottash AL, Extein I. Hypothyroidism and depression: evidence from complete thyroid function evaluation. JAMA. 1981;245:28-31.

29. Kupka RW, Nolen WA, Post RM, et al. High rate of autoimmune thyroiditis in bipolar disorder: lack of association with lithium exposure. Biol Psychiatry. 2002;51:305-311.

30. Szuba MP, Amsterdam JD. Rapid antidepressant response after nocturnal TRH administration in patients with bipolar I and bipolar type II major depression. J Clin Psychopharmacol. 2005;25:325-330.

31. Extein I, Pottash AL, Gold MS. Does subclinical hypothyroidism predispose to tricyclic-induced rapid mood cycles? J Clin Psychiatry. 1982;43:32-33.

32. American Association of Clinical Endocrinologists. Medical guidelines for clinical practice for the evaluation and treatment of hyperthyroidism and hypothyroidism. Endocr Prac. 2002;8:457-469.

33. El-Mallakh RS, Karippott A. Antidepressant-associated chronic irritable dysphoria (ACID) in bipolar disorder. J Affect Disord. 2005;84:267-272.

34. Henkl V, Mergl R, Antje-Kathrin A, et al. Treatment of depression with atypical features: a meta-analytic approach. Psychiatry Res. 2006;141(1):89-101.

35. Perugi G, Akiskal HS, Lattanzi D, et al. The high prevalence of “soft” bipolar (II) features in atypical depression. Compr Psychiatry. 1998;39(2):63-71.

References

 

1. Goldberg JF. What constitutes evidence-based pharmacotherapy for bipolar disorder? Part 1: First-line treatments. J Clin Psychiatry. 2007;68:1982-1983.

2. Goldberg JF. What constitutes evidence-based pharmacotherapy for bipolar disorder? Part 2: Complex presentations and clinical context. J Clin Psychiatry. 2008;69:495-496.

3. Levine R, Fink M. Why evidence-based medicine cannot be applied to psychiatry. Psychiatric Times. 2008;25(4):10.

4. Akiskol HS, Benazzi F. The DSM-IV and ICD-10 categories of recurrent [major] depressive and bipolar II disorders: evidence that they lie on a dimensional spectrum. J Affect Disord. 2006;92:45-54.

5. Goodwin FK, Jamison KR. Manic-depressive illness: bipolar disorders and recurrent depression. 2nd ed. New York, NY: Oxford University Press; 2007:3–27.

6. Hirschfeld RMA, Lewis L, Vornik L. Perceptions and impact of bipolar disorder: how far have we really come? Results of the National Depressive and Manic-Depressive Association 2000 survey of individuals with bipolar disorder. J Clin Psychiatry. 2003;64(2):161-167.

7. Blanco C, Laje G, Olfson M, et al. Trends in the treatment of bipolar disorder by outpatient psychiatrists. Am J Psychiatry. 2002;159:1005-1010.

8. Ghaemi SN, Lenox MS, Baldessarini RJ. Effectiveness and safety of long-term antidepressant treatment in bipolar disorder. J Clin Psychiatry. 2001;62:565-569.

9. Ghaemi SN, Boiman EE, Goodwin FK. Diagnosing bipolar disorder and the effect of antidepressants: a naturalistic study. J Clin Psychiatry. 2000;61:804-808.

10. Gijsman HF, Geddes JR, Rendell JM, et al. Antidepressants for bipolar depression: a systematic review of randomized, controlled trials. Am J Psychiatry. 2005;161:1537-1547.

11. Ghaemi SN, Sachs GS, Chiou AM, et al. Is bipolar disorder still underdiagnosed? Are antidepressants overutilized? J Affect Disord. 1999;52:134-144.

12. Sachs GS, Nierenberg AA, Calabrese JR, et al. Effectiveness of adjunctive antidepressant treatment for bipolar depression. N Engl J Med. 2007;356:1711-1722.

13. Altshuler L, Suppes T, Black D, et al. Impact of antidepressant discontinuation after acute bipolar depression remission on rates of depressive relapse at 1-year follow-up. Am J Psychiatry. 2003;160:1252-1262.

14. Akiskol HS. Developmental pathways to bipolarity: are juvenile-onset depressions pre-bipolar? J Am Acad Child Adolesc Psychiatry. 1995;34(6):754-763.

15. Geller B, Zimmerman B, Williams M, et al. Bipolar disorder at prospective follow-up of adults who had prepubertal major depressive disorder. Am J Psychiatry. 2001;158:125-127.

16. Food and Drug Administration: Center for Drug Evaluation and Research. Revisions to product labeling. Available at: http://www.FDA.gov/cder/drug/antidepressants/default.htm. Accessed January 12, 2009.

17. McElroy S, Strakowski S, West S, et al. Phenomenology of adolescent and adult mania in hospitalized patients with bipolar disorder. Am J Psychiatry. 1997;154:44-49.

18. Olfson M, Marcus SC. A case-control study of antidepressants and attempted suicide during early phase treatment of major depressive episodes. J Clin Psychiatry. 2008;69:425-432.

19. Keck PE, Jr, McElroy SL, Havens JR, et al. Psychosis in bipolar disorder: phenomenology and impact on morbidity and course of illness. Compr Psychiatry. 2003;44:263-269.

20. Jones I, Craddock N. Familiarity of the puerperal trigger in bipolar disorder: results of a family study. Am J Psychiatry. 2001;158:913-917.

21. Chaudron LH, Pies RW. The relationship between postpartum psychosis and bipolar disorder: a review. J Clin Psychiatry. 2003;64:1284-1292.

22. Wisner KL, Peindl KS, Hanusa BH. Psychiatric episodes in women and young children. J Affect Disord. 1995;34:1-11.

23. Sharma V. A cautionary note on the use of antidepressants in postpartum depression. Bipolar Disord. 2006;8:411-414.

24. O’Malley S. “Are you there alone?” The unspeakable crime of Andrea Yates. New York, NY: Simon and Schuster; 2004.

25. Altshuler LL, Bauer M, Frye MA, et al. Does thyroid supplementation accelerate tricyclic antidepressant response? A review and meta-analysis of the literature. Am J Psychiatry. 2001;158:1617-1622.

26. Joffe RT. The use of thyroid supplements to augment antidepressant medication. J Clin Psychiatry. 2008;59:26-29.

27. Cooper-Kazaz R, Apter JT, Cohen R, et al. Combined treatment with sertraline and liothyronine in major depression: a randomized, double-blind, placebo-controlled trial. Arch Gen Psychiatry. 2007;64:679-688.

28. Gold MS, Pottash AL, Extein I. Hypothyroidism and depression: evidence from complete thyroid function evaluation. JAMA. 1981;245:28-31.

29. Kupka RW, Nolen WA, Post RM, et al. High rate of autoimmune thyroiditis in bipolar disorder: lack of association with lithium exposure. Biol Psychiatry. 2002;51:305-311.

30. Szuba MP, Amsterdam JD. Rapid antidepressant response after nocturnal TRH administration in patients with bipolar I and bipolar type II major depression. J Clin Psychopharmacol. 2005;25:325-330.

31. Extein I, Pottash AL, Gold MS. Does subclinical hypothyroidism predispose to tricyclic-induced rapid mood cycles? J Clin Psychiatry. 1982;43:32-33.

32. American Association of Clinical Endocrinologists. Medical guidelines for clinical practice for the evaluation and treatment of hyperthyroidism and hypothyroidism. Endocr Prac. 2002;8:457-469.

33. El-Mallakh RS, Karippott A. Antidepressant-associated chronic irritable dysphoria (ACID) in bipolar disorder. J Affect Disord. 2005;84:267-272.

34. Henkl V, Mergl R, Antje-Kathrin A, et al. Treatment of depression with atypical features: a meta-analytic approach. Psychiatry Res. 2006;141(1):89-101.

35. Perugi G, Akiskal HS, Lattanzi D, et al. The high prevalence of “soft” bipolar (II) features in atypical depression. Compr Psychiatry. 1998;39(2):63-71.

Issue
Current Psychiatry - 08(02)
Issue
Current Psychiatry - 08(02)
Page Number
27-28,31-33,39
Page Number
27-28,31-33,39
Publications
Publications
Topics
Article Type
Display Headline
Controversies in bipolar disorder: Trust evidence or experience?
Display Headline
Controversies in bipolar disorder: Trust evidence or experience?
Legacy Keywords
bipolar disorder; evidence-based medicine; bipolar spectrum; subthreshold hypomania; manic switches; antidepressant monotherapy; Gary E Miller; Richard L noel
Legacy Keywords
bipolar disorder; evidence-based medicine; bipolar spectrum; subthreshold hypomania; manic switches; antidepressant monotherapy; Gary E Miller; Richard L noel
Sections
Disallow All Ads
Alternative CME
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Subclinical hypothyroidism: Merely monitor or time to treat?

Article Type
Changed
Tue, 12/11/2018 - 15:06
Display Headline
Subclinical hypothyroidism: Merely monitor or time to treat?

Principal Source: Roberts LM, Pattison H, Roalfe A, et al. Is subclinical thyroid dysfunction in the elderly associated with depression or cognitive dysfunction? Ann Intern Med. 2006;145:573-581.—Discussant: Y Pritham Raj, MD

Practice Points

  • Subclinical thyroid dysfunction is largely a laboratory diagnosis that merits observation but not necessarily treatment.
  • Watchful waiting is preferable in patients age ≥65 with mild subclinical hypothyroidism (TSH <10 mU/L) unless they have prominent mood, cognitive, or medical conditions—such as congestive heart failure or hyperlipidemia—that could benefit from early thyroid replacement.
  • In adults age <65, consider TSH 4.5 to 10 mU/L as a threshold for initiating thyroid replacement, particularly if anti-TPO antibodies are present (although prevailing recommendations still favor the watchful waiting approach).6

Thyroid dysfunction enters the differential diagnosis for most mood, anxiety, thought, and cognitive disorders. Because more than one-half of the estimated 27 million Americans with hyperthyroidism or hypothyroidism are undiagnosed,1 the American Thyroid Association recommends universal screening for thyroid dysfunction after age 35, with a recheck every 5 years. Although some clinicians feel this recommendation is excessive, strategic screening with a thyroid-stimulating hormone (TSH) test is important for patients with psychiatric illnesses.

If a patient’s TSH is abnormal, repeating the test while measuring the free thyroxine (T4) and in most cases the antithyroid peroxidase antibody (anti-TPO) has good clinical value. Anti-TPO antibodies are a useful biomarker for autoimmune thyroid disease, such as Hashimoto’s thyroiditis or Graves’ disease. If laboratory findings suggest the hypothyroid spectrum, a fasting lipid profile may help determine risk of adverse cardiovascular outcomes.

Therapy

Symptoms of hypothyroidism—indicated by an elevated TSH (usually >20 mU/L) and low T4—overlap with psychiatric illness (Table) but are easy to treat. Psychiatrists who are accustomed to calculating weight-based dosing of medications such as lithium and valproic acid may have little difficulty initiating levothyroxine replacement (typically 1.6 mcg/kg/day) for patients with overt hypothyroidism. Treating hyperthyroidism (low TSH and high T4) can be more complex and generally is left to an internist or endocrinologist. But how should you treat subclinical thyroid dysfunction?

Table

Hypothyroidism symptoms that indicate treatment

With psychiatric overlap
Fatigue
Hypersomnolence
Cognitive impairment (forgetfulness)
Difficulty concentrating/learning
Weight gain/fluid retention
Somatic symptoms
Dry, itchy skin
Brittle nails and hair
Constipation
Myalgias
Heavy and/or irregular menses
Increased miscarriage risk
Cold sensitivity

Treat or wait?

Subclinical hypothyroidism (SH)—in which T4 is normal—usually is a laboratory diagnosis defined in a spectrum:

  • TSH of 4.5 to 10 mU/L is mild SH (80% of cases)
  • TSH of 10 to 20 mU/L is more severe SH.

SH is a well-established risk factor for depression. One study found a nearly 3-fold higher lifetime prevalence of depression in young and middle-aged women with SH.2 To the practicing psychiatrist, these results may sound like a mandate to treat all patients with SH—particularly those with depression. Consider, however, that in a prospective observational study the TSH of >37% of patients with SH returned to normal with observation alone.3 In fact, <27% of patients with SH went on to develop overt hypothyroidism during the study period, on average within 31.7 months.

A second study that would argue against treating patients with mild SH noted decreased cardiovascular and noncardio-vascular mortality among elderly patients with elevated TSH,4 implying that SH may be protective compared with the euthyroid state, at least among octogenarians.

Still, do mood, anxiety, or cognitive symptoms in SH patients merit earlier, more aggressive treatment? This question was addressed by a recent cross-sectional study that demonstrated no correlation between mood and SH.5 Although statistically significant associations were seen among anxiety, cognition, and elevated TSH, the magnitude of the associations lacked clinical relevance. This study was designed to further assess an earlier inconclusive review.6

Ultimately, treating SH—although easy to do—may have little impact on your patient’s overall mood and cognition until TSH is ≥10 mU/L.

Drug brand names

  • Levothyroxine • Levoxyl, Synthroid
  • Lithium • various
  • Valproic acid • Depakene

Disclosure

Dr. Raj is a consultant to Alpharma and a speaker for AstraZeneca.

References

1. American Association of Clinical Endocrinologists. Thyroid fact sheet. Available at: http://www.medem.com/medlib/article/ZZZNIEIUKIE. Accessed January 14, 2009.

2. Haggerty JJ, Stern RA, Mason GA, et al. Subclinical hypothyroidism: a modifiable risk factor for depression? Am J Psychiatry. 1993;150:508-510.

3. Diez JJ, Iglesias P. Spontaneous subclinical hypothyroidism in patients older than 55 years: an analysis of natural course and risk factors for the development of overt thyroid failure. J Clin Endocrinol Metab. 2004;89(10):4890-4897.

4. Gussekloo J, van Exel E, de Craen AJM, et al. Thyroid status, disability and cognitive function, and survival in old age. JAMA. 2004;292:2591-2599.

5. Roberts LM, Pattison H, Roalfe A, et al. Is subclinical thyroid dysfunction in the elderly associated with depression or cognitive dysfunction? Ann Intern Med. 2006;145:573-581.

6. Surks MI, Ortiz E, Daniels GH, et al. Subclinical thyroid disease scientific review and guidelines for diagnosis and management. JAMA. 2004;291(2):228-238.

Article PDF
Author and Disclosure Information


Robert M. McCarron, DO
Series Editor
Dr. Raj is assistant professor of internal medicine and psychiatry, Oregon Health and Science University, Portland.

Issue
Current Psychiatry - 08(02)
Publications
Page Number
47-48
Legacy Keywords
thyroid dysfunction; hypothyroidism; subclinical hypothyroidism; thyroid-stimulating hormone; Y Pritham Raj; Robert M McCarron
Sections
Author and Disclosure Information


Robert M. McCarron, DO
Series Editor
Dr. Raj is assistant professor of internal medicine and psychiatry, Oregon Health and Science University, Portland.

Author and Disclosure Information


Robert M. McCarron, DO
Series Editor
Dr. Raj is assistant professor of internal medicine and psychiatry, Oregon Health and Science University, Portland.

Article PDF
Article PDF

Principal Source: Roberts LM, Pattison H, Roalfe A, et al. Is subclinical thyroid dysfunction in the elderly associated with depression or cognitive dysfunction? Ann Intern Med. 2006;145:573-581.—Discussant: Y Pritham Raj, MD

Practice Points

  • Subclinical thyroid dysfunction is largely a laboratory diagnosis that merits observation but not necessarily treatment.
  • Watchful waiting is preferable in patients age ≥65 with mild subclinical hypothyroidism (TSH <10 mU/L) unless they have prominent mood, cognitive, or medical conditions—such as congestive heart failure or hyperlipidemia—that could benefit from early thyroid replacement.
  • In adults age <65, consider TSH 4.5 to 10 mU/L as a threshold for initiating thyroid replacement, particularly if anti-TPO antibodies are present (although prevailing recommendations still favor the watchful waiting approach).6

Thyroid dysfunction enters the differential diagnosis for most mood, anxiety, thought, and cognitive disorders. Because more than one-half of the estimated 27 million Americans with hyperthyroidism or hypothyroidism are undiagnosed,1 the American Thyroid Association recommends universal screening for thyroid dysfunction after age 35, with a recheck every 5 years. Although some clinicians feel this recommendation is excessive, strategic screening with a thyroid-stimulating hormone (TSH) test is important for patients with psychiatric illnesses.

If a patient’s TSH is abnormal, repeating the test while measuring the free thyroxine (T4) and in most cases the antithyroid peroxidase antibody (anti-TPO) has good clinical value. Anti-TPO antibodies are a useful biomarker for autoimmune thyroid disease, such as Hashimoto’s thyroiditis or Graves’ disease. If laboratory findings suggest the hypothyroid spectrum, a fasting lipid profile may help determine risk of adverse cardiovascular outcomes.

Therapy

Symptoms of hypothyroidism—indicated by an elevated TSH (usually >20 mU/L) and low T4—overlap with psychiatric illness (Table) but are easy to treat. Psychiatrists who are accustomed to calculating weight-based dosing of medications such as lithium and valproic acid may have little difficulty initiating levothyroxine replacement (typically 1.6 mcg/kg/day) for patients with overt hypothyroidism. Treating hyperthyroidism (low TSH and high T4) can be more complex and generally is left to an internist or endocrinologist. But how should you treat subclinical thyroid dysfunction?

Table

Hypothyroidism symptoms that indicate treatment

With psychiatric overlap
Fatigue
Hypersomnolence
Cognitive impairment (forgetfulness)
Difficulty concentrating/learning
Weight gain/fluid retention
Somatic symptoms
Dry, itchy skin
Brittle nails and hair
Constipation
Myalgias
Heavy and/or irregular menses
Increased miscarriage risk
Cold sensitivity

Treat or wait?

Subclinical hypothyroidism (SH)—in which T4 is normal—usually is a laboratory diagnosis defined in a spectrum:

  • TSH of 4.5 to 10 mU/L is mild SH (80% of cases)
  • TSH of 10 to 20 mU/L is more severe SH.

SH is a well-established risk factor for depression. One study found a nearly 3-fold higher lifetime prevalence of depression in young and middle-aged women with SH.2 To the practicing psychiatrist, these results may sound like a mandate to treat all patients with SH—particularly those with depression. Consider, however, that in a prospective observational study the TSH of >37% of patients with SH returned to normal with observation alone.3 In fact, <27% of patients with SH went on to develop overt hypothyroidism during the study period, on average within 31.7 months.

A second study that would argue against treating patients with mild SH noted decreased cardiovascular and noncardio-vascular mortality among elderly patients with elevated TSH,4 implying that SH may be protective compared with the euthyroid state, at least among octogenarians.

Still, do mood, anxiety, or cognitive symptoms in SH patients merit earlier, more aggressive treatment? This question was addressed by a recent cross-sectional study that demonstrated no correlation between mood and SH.5 Although statistically significant associations were seen among anxiety, cognition, and elevated TSH, the magnitude of the associations lacked clinical relevance. This study was designed to further assess an earlier inconclusive review.6

Ultimately, treating SH—although easy to do—may have little impact on your patient’s overall mood and cognition until TSH is ≥10 mU/L.

Drug brand names

  • Levothyroxine • Levoxyl, Synthroid
  • Lithium • various
  • Valproic acid • Depakene

Disclosure

Dr. Raj is a consultant to Alpharma and a speaker for AstraZeneca.

Principal Source: Roberts LM, Pattison H, Roalfe A, et al. Is subclinical thyroid dysfunction in the elderly associated with depression or cognitive dysfunction? Ann Intern Med. 2006;145:573-581.—Discussant: Y Pritham Raj, MD

Practice Points

  • Subclinical thyroid dysfunction is largely a laboratory diagnosis that merits observation but not necessarily treatment.
  • Watchful waiting is preferable in patients age ≥65 with mild subclinical hypothyroidism (TSH <10 mU/L) unless they have prominent mood, cognitive, or medical conditions—such as congestive heart failure or hyperlipidemia—that could benefit from early thyroid replacement.
  • In adults age <65, consider TSH 4.5 to 10 mU/L as a threshold for initiating thyroid replacement, particularly if anti-TPO antibodies are present (although prevailing recommendations still favor the watchful waiting approach).6

Thyroid dysfunction enters the differential diagnosis for most mood, anxiety, thought, and cognitive disorders. Because more than one-half of the estimated 27 million Americans with hyperthyroidism or hypothyroidism are undiagnosed,1 the American Thyroid Association recommends universal screening for thyroid dysfunction after age 35, with a recheck every 5 years. Although some clinicians feel this recommendation is excessive, strategic screening with a thyroid-stimulating hormone (TSH) test is important for patients with psychiatric illnesses.

If a patient’s TSH is abnormal, repeating the test while measuring the free thyroxine (T4) and in most cases the antithyroid peroxidase antibody (anti-TPO) has good clinical value. Anti-TPO antibodies are a useful biomarker for autoimmune thyroid disease, such as Hashimoto’s thyroiditis or Graves’ disease. If laboratory findings suggest the hypothyroid spectrum, a fasting lipid profile may help determine risk of adverse cardiovascular outcomes.

Therapy

Symptoms of hypothyroidism—indicated by an elevated TSH (usually >20 mU/L) and low T4—overlap with psychiatric illness (Table) but are easy to treat. Psychiatrists who are accustomed to calculating weight-based dosing of medications such as lithium and valproic acid may have little difficulty initiating levothyroxine replacement (typically 1.6 mcg/kg/day) for patients with overt hypothyroidism. Treating hyperthyroidism (low TSH and high T4) can be more complex and generally is left to an internist or endocrinologist. But how should you treat subclinical thyroid dysfunction?

Table

Hypothyroidism symptoms that indicate treatment

With psychiatric overlap
Fatigue
Hypersomnolence
Cognitive impairment (forgetfulness)
Difficulty concentrating/learning
Weight gain/fluid retention
Somatic symptoms
Dry, itchy skin
Brittle nails and hair
Constipation
Myalgias
Heavy and/or irregular menses
Increased miscarriage risk
Cold sensitivity

Treat or wait?

Subclinical hypothyroidism (SH)—in which T4 is normal—usually is a laboratory diagnosis defined in a spectrum:

  • TSH of 4.5 to 10 mU/L is mild SH (80% of cases)
  • TSH of 10 to 20 mU/L is more severe SH.

SH is a well-established risk factor for depression. One study found a nearly 3-fold higher lifetime prevalence of depression in young and middle-aged women with SH.2 To the practicing psychiatrist, these results may sound like a mandate to treat all patients with SH—particularly those with depression. Consider, however, that in a prospective observational study the TSH of >37% of patients with SH returned to normal with observation alone.3 In fact, <27% of patients with SH went on to develop overt hypothyroidism during the study period, on average within 31.7 months.

A second study that would argue against treating patients with mild SH noted decreased cardiovascular and noncardio-vascular mortality among elderly patients with elevated TSH,4 implying that SH may be protective compared with the euthyroid state, at least among octogenarians.

Still, do mood, anxiety, or cognitive symptoms in SH patients merit earlier, more aggressive treatment? This question was addressed by a recent cross-sectional study that demonstrated no correlation between mood and SH.5 Although statistically significant associations were seen among anxiety, cognition, and elevated TSH, the magnitude of the associations lacked clinical relevance. This study was designed to further assess an earlier inconclusive review.6

Ultimately, treating SH—although easy to do—may have little impact on your patient’s overall mood and cognition until TSH is ≥10 mU/L.

Drug brand names

  • Levothyroxine • Levoxyl, Synthroid
  • Lithium • various
  • Valproic acid • Depakene

Disclosure

Dr. Raj is a consultant to Alpharma and a speaker for AstraZeneca.

References

1. American Association of Clinical Endocrinologists. Thyroid fact sheet. Available at: http://www.medem.com/medlib/article/ZZZNIEIUKIE. Accessed January 14, 2009.

2. Haggerty JJ, Stern RA, Mason GA, et al. Subclinical hypothyroidism: a modifiable risk factor for depression? Am J Psychiatry. 1993;150:508-510.

3. Diez JJ, Iglesias P. Spontaneous subclinical hypothyroidism in patients older than 55 years: an analysis of natural course and risk factors for the development of overt thyroid failure. J Clin Endocrinol Metab. 2004;89(10):4890-4897.

4. Gussekloo J, van Exel E, de Craen AJM, et al. Thyroid status, disability and cognitive function, and survival in old age. JAMA. 2004;292:2591-2599.

5. Roberts LM, Pattison H, Roalfe A, et al. Is subclinical thyroid dysfunction in the elderly associated with depression or cognitive dysfunction? Ann Intern Med. 2006;145:573-581.

6. Surks MI, Ortiz E, Daniels GH, et al. Subclinical thyroid disease scientific review and guidelines for diagnosis and management. JAMA. 2004;291(2):228-238.

References

1. American Association of Clinical Endocrinologists. Thyroid fact sheet. Available at: http://www.medem.com/medlib/article/ZZZNIEIUKIE. Accessed January 14, 2009.

2. Haggerty JJ, Stern RA, Mason GA, et al. Subclinical hypothyroidism: a modifiable risk factor for depression? Am J Psychiatry. 1993;150:508-510.

3. Diez JJ, Iglesias P. Spontaneous subclinical hypothyroidism in patients older than 55 years: an analysis of natural course and risk factors for the development of overt thyroid failure. J Clin Endocrinol Metab. 2004;89(10):4890-4897.

4. Gussekloo J, van Exel E, de Craen AJM, et al. Thyroid status, disability and cognitive function, and survival in old age. JAMA. 2004;292:2591-2599.

5. Roberts LM, Pattison H, Roalfe A, et al. Is subclinical thyroid dysfunction in the elderly associated with depression or cognitive dysfunction? Ann Intern Med. 2006;145:573-581.

6. Surks MI, Ortiz E, Daniels GH, et al. Subclinical thyroid disease scientific review and guidelines for diagnosis and management. JAMA. 2004;291(2):228-238.

Issue
Current Psychiatry - 08(02)
Issue
Current Psychiatry - 08(02)
Page Number
47-48
Page Number
47-48
Publications
Publications
Article Type
Display Headline
Subclinical hypothyroidism: Merely monitor or time to treat?
Display Headline
Subclinical hypothyroidism: Merely monitor or time to treat?
Legacy Keywords
thyroid dysfunction; hypothyroidism; subclinical hypothyroidism; thyroid-stimulating hormone; Y Pritham Raj; Robert M McCarron
Legacy Keywords
thyroid dysfunction; hypothyroidism; subclinical hypothyroidism; thyroid-stimulating hormone; Y Pritham Raj; Robert M McCarron
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Loss of enzyme induction: Ups and downs of a hidden drug-drug interaction

Article Type
Changed
Tue, 12/11/2018 - 15:06
Display Headline
Loss of enzyme induction: Ups and downs of a hidden drug-drug interaction

Mr. P, age 35 with schizophrenia and seizure disorder, has been maintained on risperidone, 6 mg qhs, and phenytoin, 300 mg qhs. For clinical reasons, the treating neurologist changes the anticonvulsant to divalproex. One week later, Mr. P presents to the emergency room complaining of jaw and neck stiffness.

Ms. K, age 43 with a history of schizoaffective disorder, bipolar type, and erratic medication adherence, is being treated with quetiapine, 600 mg at bedtime, and carbamazepine, 1,000 mg/d. Between appointments she stops taking carbamazepine, believing it is causing her to hear voices from her television. Two weeks later, the manager of Ms. K’s independent living facility tells the psychiatrist that the patient appears excessively sedated and has fallen twice in the past few days.

Mrs. T, a 39-year-old state hospital resident with schizoaffective disorder, bipolar type, has been treated with clozapine, 250 mg bid for 6 months; her most recent trough serum level was 492 ng/mL. She smokes 15 cigarettes/d. Two weeks after the hospital institutes a no-smoking policy, Mrs. T complains of excessive drooling and lightheadedness. Her trough clozapine level is now 875 ng/mL.

Discontinuing a medication that has enzyme-inducing effects presents a hidden problem for patients receiving antipsychotic pharmacotherapy. Certain hepatic enzymes responsible for antipsychotic metabolism—as well those involved in intercellular drug transport—are induced by medication or environmental exposures.1,2 Adding a medication that induces these enzymes to the regimen of a patient receiving antipsychotic therapy can result in markedly reduced serum antipsychotic levels, and discontinuing an inducing agent can result in increased antipsychotic levels.

Drug-drug interactions (DDIs) are a substantial contributor to adverse drug reactions (Box).3-7 Antipsychotic prescribing information highlights potential DDIs from the use of enzyme inhibitors and inducers but identifies only effects caused by adding a second agent. The prescriber remains the sole line of defense for monitoring for DDIs when discontinuing a medication that has inducing or inhibiting effects.

Most psychiatrists are aware that certain medications have clinically significant effects on cytochrome P450 (CYP) activity and of the potential for CYP inhibitors to generate DDIs. Clinicians often are aware of antidepressant medications’ CYP-inhibiting effects, know that levels of other medications will change when discontinuing a potent P450 inhibitor, and understand the need to increase dosages of medications influenced by such agents.8

However, few studies have evaluated the effects of enzyme induction on antipsychotic drug levels,9,10 and the literature rarely discusses changes in serum drug levels after loss of enzyme or drug transport induction.11 If unrecognized, these changes may have significant clinical consequences.

Box

Drug interactions: A common cause of nonadherence

Drug-drug interactions (DDIs) are a common and often preventable cause of morbidity and mortality. National surveillance data showed 700,000 emergency room visits related to adverse drug reactions (ADRs) in the 2 years from January 2004 through 2005.3 ADRs are particularly concerning for psychiatrists managing polypharmacy regimens for patients with severe mental disorders such as schizophrenia.

Literature on DDIs with antipsychotics focuses primarily on kinetic interactions that generate supratherapeutic drug levels.4,5 Because development of side effects is associated with reduced adherence, these kinetic interactions may increase the risk of adverse effects and lead to patients stopping the antipsychotic treatment.6,7

Two induction pathways

The primary mechanism underlying clinically significant DDIs occurs during CYP-mediated phase I metabolism. Molecules undergo oxidative conversion into metabolites that can be conjugated by phase II enzymes, generating more soluble forms that facilitate excretion.

The workhorse of human CYP metabolism is 3A4 (Table 1),12,13 which comprises 30% of hepatic activity and 70% of gut cytochrome activity.14 CYP 1A2 is responsible for 10% to 15% of CYP activity.

Both CYP 3A4 and 1A2 are inducible. A wide variety of medications induce 3A4 activity. The list of 1A2 inducers is shorter; the most common are aryl hydrocarbons from cigarette smoke and proton pump inhibitors.

CYP 2D6 accounts for 20% of hepatic cytochrome activity but is not inducible. CYP 2D6 is well known to psychiatrists because some selective serotonin reuptake inhibitors (SSRIs) and the non-SSRI antidepressant bupropion are potent inhibitors of this enzyme.15,16

P-glycoprotein (PGP) induction. Transmembrane shuttles such as P-glycoprotein (PGP) are an important component of drug disposition. PGP belongs to the family of ATP binding cassette (ABC) transporters that bring molecules across cellular barriers.17,18 It was first described in cancer cells that developed multiple drug resistance (MDR) and is often referred to as MDR1.19 PGP is encoded on human chromosome 7 and expressed in normal tissues, particularly in areas where cells seek to limit drug influx, such as those lining the luminal surface of the small and large intestine and those lining the blood-brain barrier and blood-testis barrier. The expression of PGP in hepatic cells promotes drug clearance by enhancing biliary drug excretion.

 

 

PGP is encoded on the same chromosome as CYP 3A4, and these 2 proteins frequently are expressed in the same cells, particularly in the intestinal lining and liver. Moreover, PGP is inducible, and there is substantial overlap between medications that are substrates for—or inducers of—PGP and CYP 3A4 activity. This makes it challenging to determine whether the kinetic effects of a second medication are the result of interference of 3A4, PGP, or both.

Polymorphisms in PGP activity may influence the penetration of psychotropic medications into the CNS. Studies indicate an association between certain PGP polymorphisms and treatment outcomes.17,18

Table 1

What induces CYP 1A2 and 3A4?

EnzymeDescriptionInducers*
CYP 1A2
  • Responsible for 10% to 15% of all CYP P450 activity
  • Located on chromosome 15
  • Low affinity/high capacity enzyme
  • Low affinity/high capacity enzyme
  • Prevalence of genetic polymorphisms conferring poor metabolizer status: 12% to 13%
Aryl hydrocarbons (smoking), protonpump inhibitors (omeprazole > lansoprazole > pantoprazole), modafinil, St. John’s wort, chargrilled meat, cruciferous vegetables such as broccoli and cabbage, flavones, protein supplements
CYP 3A4
  • Responsible for 30% of hepatic CYP 450 activity, 70% of gut cytochrome activity
  • Located on chromosome 7 (same as PGP)
  • Low affinity/high capacity enzyme
  • Little evidence for significant functional polymorphisms
Carbamazepine, phenytoin, phenobarbital, rifampin, oxcarbazepine, efavirenz, glucocorticoids, modafinil, nevirapine, pioglitazone, St. John’s wort
* Listed in order from strongest to weakest induction
CYP: cytochrome P450; PGP: P-glycoprotein
Source: References 12,13

Stopping an inducer

In general, inducers of CYP enzymes stimulate gene transcription within hours of exposure; maximum transcriptional activity occurs after 10 to 12 hours of exposure. As transcription increases, the concentration of the CYP mRNA transcript steadily accumulates, as does concentration of CYP protein.

After an inducer is discontinued, transcription returns to basal levels within 18 hours; however, the degradation of CYP proteins is a first-order process, with a half-life of 8 to 30 hours. As a result, the decrease in cellular CYP concentration—and the level of activity—lags behind the decreased synthesis from reduced mRNA levels.

As with other first-order kinetic processes, the expected decrease in CYP activity will require 5 half-lives to reach the new steady state (ie, back to baseline CYP activity). This suggests that drug levels previously decreased by CYP induction will reach their peak on average 1 to 2 weeks after the inducer is discontinued.20

Interactions with antipsychotics

Effects on serum antipsychotic levels caused by discontinuing a CYP or PGP inducer can be predicted from data on decreases in antipsychotic levels following inducer exposure. Except for ziprasidone and paliperidone, most atypical antipsychotics are prone to substantial decreases during concomitant inducer use (Table 2).21

The effect of enzyme inducers on risperidone is particularly interesting. Conversion of risperidone to its active metabolite 9-OH risperidone (paliperidone) occurs primarily via 2D6,22 yet concurrent use of carbamazepine—a potent CYP 3A4 inducer—results in a 50% decrease in the concentration of the active moiety (risperidone plus 9-OH risperidone). This finding and other early investigations suggested that CYP 3A had a role in risperidone metabolism,23,24 but these early studies and case series often involved molecules that had activity at both 3A4 and PGP. Further research clarified that effects on PGP—and not 3A4—are responsible for the changes in risperidone metabolism observed with the use of carbamazepine and other medications.25,26

Induction in case patients: Follow-up. Regardless of whether induction is mediated by ≥1 metabolic pathways, the loss of the inducer will result in serum antipsychotic increases that are proportional to the initial decrease.20 For example, with risperidone, the expected decrease is 50%. Therefore, after Mr. P stopped taking phenytoin, his serum risperidone level would be expected to double, which resulted in extrapyramidal side effects.

Quetiapine clearance is increased 5-fold by inducer exposure, so a clinician treating Ms. K would expect a marked increase in somnolence—and possibly orthostasis—as serum quetiapine levels peak 1 to 2 weeks following carbamazepine discontinuation.

The effects of smoking cessation on serum clozapine levels have been well-documented.1,27 Clinicians should anticipate median increases in serum clozapine levels of 55% after a patient discontinues smoking (aryl hydrocarbon exposure), but changes vary substantially among individuals. Mrs. T’s serum clozapine increased approximately 78%.

Careful clinical monitoring and slow downward adjustment of antipsychotic doses could have prevented the adverse effects these 3 patients experienced after loss of CYP/PGP induction and the consequences those side effects present for future medication adherence. When loss of induction is unplanned—as when Ms. K stopped taking carbamazepine but continued quetiapine—clinicians need to be alert to the fact that the patient was prescribed an inducer and include the loss of induction as a hypothesis for the patient’s somnolence.

 

 

Table 2

Effects of CYP/PGP induction on atypical antipsychotics

AntipsychoticMetabolic pathwaysEffect of induction
Aripiprazole2D6 and 3A4 convert aripiprazole to active metabolite dehydro-aripiprazole3A4 induction decreases maximum concentration of aripiprazole and metabolite by 70%
ClozapineMultiple enzymes convert clozapine to N-desmethylclozapine; mean contributions of CYP 1A2, 2C19, 3A4, 2C9, and 2D6 are 30%, 24%, 22%, 12%, and 6%, respectively, with CYP 1A2 predominantly involved at low concentrationsLoss of smoking-related 1A2 induction results in 50% increase in serum levels
OlanzapineDirect glucuronidation or 1A2-mediated oxidation to N-desmethlyolanzapineCarbamazepine use increases clearance by 50%. Olanzapine concentration:dose ratio is about 5-fold lower in smokers (7.9 +/- 2.6) than in nonsmokers (1.56 +/- 1.1; P
Paliperidone59% excreted unchanged in urine; phase I metabolism accounts for ≤10% of drug clearanceUnlikely to significantly impact levels, but impact of PGP induction is unknown
Quetiapine3A4-mediated sulfoxidation to inactive metabolite is primary pathway, but numerous metabolites noted, with 1 active metabolite (norquetiapine)Phenytoin increases clearance 5-fold
Risperidone2D6 converts risperidone to active metabolite 9-OH risperidoneIn a drug interaction study of risperidone, 6 mg/d for 3 weeks, followed by 3 weeks of carbamazepine, active moiety concentration was decreased by about 50%
Ziprasidone3A4 (~1/3); aldehyde oxidase (~2/3)Approximately 35% decrease in ziprasidone exposure by carbamazepine
CYP: cytochrome P450; PGP: P-glycoprotein
Source: Reference 21

Clinical considerations

In the absence of detailed data on antipsychotic metabolism, clinicians can make intelligent decisions regarding potential DDIs by:

  • knowing the extent of induction by common offenders (such as carbamazepine or phenytoin) documented in the medication’s prescribing information or demonstrated through convincing case reports or case series
  • memorizing the list of CYP 1A2 and CYP 3A4/PGP inducers.

Although the list of CYP 1A2 and CYP 3A4/PGP inducers is short, it is essential for clinicians to consult a readily available source of this information that is periodically updated to account for newer medications, such as the online table maintained by Flockhart (see Related Resources).28

Patients who may be susceptible to effects from loss of enzyme induction (including smokers receiving olanzapine or clozapine or others taking 3A4/PGP inducers) must be identified, and plans made for dosage adjustments if inducing agents are discontinued for a sufficient time (≥1 week) to result in downregulation of CYP or PGP activity. A slow taper of the antipsychotic over 1 to 2 weeks to the new target dose should compensate for loss of enzyme or PGP induction.

For newer antipsychotic medications with limited data, the proposed discontinuation of an inducer should, at the minimum, prompt a discussion between the psychiatrist and patient regarding the expected increase in serum antipsychotic levels and potential adverse effects that may result. Clinicians also must make every attempt to stay apprised of a patient’s current medications, bearing in mind that another provider may prescribe an inducer. Patients with schizophrenia always should be educated to contact the psychiatrist following any change in medication regimen, placing particular emphasis on the 1 or 2 medications that are known to be implicated in DDIs with the patient’s current antipsychotic.

Related Resources

  • Flockhart DA. Drug interactions: Cytochrome P450 drug interaction table. Indiana University School of Medicine. 2007. http://medicine.iupui.edu/flockhart/table.htm.
  • Cozza KL, Armstrong SC, Oesterheld JR. Concise guide to drug interaction principles for medical practice: Cytochrome P450s, UGTS, p-glycoproteins. Washington, DC: American Psychiatric Press, Inc; 2003.
Drug brand names

  • Aripiprazole • Abilify
  • Bupropion • Wellbutrin
  • Carbamazepine • Carbatrol, Tegretol
  • Clozapine • Clozaril
  • Divalproex • Depakote
  • Efavirenz • Sustiva
  • Lansoprazole • Prevacid
  • Modafinil • Provigil
  • Nevirapine • Viramune
  • Olanzapine • Zyprexa
  • Omeprazole • Prilosec
  • Oxcarbazepine • Trileptal
  • Paliperidone • Invega
  • Pantoprazole • Protonix
  • Phenobarbital • Barbita, Luminal, others
  • Phenytoin • Dilantin
  • Pioglitazone • Actos
  • Quetiapine • Seroquel
  • Rifampin • Rifadin, Rimactane
  • Risperidone • Risperdal
  • Ziprasidone • Geodon
Disclosure

Dr. Meyer receives grant/research support from the National Institute of Mental Health, Pfizer Inc., and the University of California. He is a consultant to Bristol-Myers Squibb, Organon, Vanda Pharmaceuticals, and Wyeth, and a speaker for AstraZeneca, Bristol-Myers Squibb, Dainippon Sumitomo Pharma, and Pfizer Inc.

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

References

1. Meyer JM. Individual changes in clozapine levels after smoking cessation: results and a predictive model. J Clin Psychopharmacol. 2001;21:569-574.

2. Wong YW, Yeh C, Thyrum PT. The effects of concomitant phenytoin administration on the steady-state pharmacokinetics of quetiapine. J Clin Psychopharmacol. 2001;21:89-93.

3. Budnitz DS, Pollock DA, Weidenbach KN, et al. National surveillance of emergency department visits for outpatient adverse drug events. JAMA. 2006;296:1858-1866.

4. Prior TI, Baker GB. Interactions between the cytochrome P450 system and the second-generation antipsychotics. J Psychiatry Neurosci. 2003;28:99-112.

5. Spina E, de Leon J. Metabolic drug interactions with newer antipsychotics: a comparative review. Basic Clin Pharmacol Toxicol. 2007;100:4-22.

6. Preskorn SH. Drug-drug interactions: proof of relevance (part II): cause of tolerability problems or noncompliance. J Psychiatr Pract. 2005;11:397-401.

7. Weiden PJ, Mackell JA, McDonnell D. Obesity as a risk factor for antipsychotic noncompliance. Schizophr Res. 2004;66:51-7.

8. Preskorn SH, Flockhart D. 2006 guide to psychiatric drug interactions. Prim Psychiatry. 2006;13:35-64.

9. Spina E, Perucca E. Clinical significance of pharmacokinetic interactions between antiepileptic and psychotropic drugs. Epilepsia. 2002;43(suppl 2):37-44.

10. Meyer JM. Drug-drug interactions with antipsychotics. CNS Spectr. 2007;12:6-9.

11. Takahashi H, Yoshida K, Higuchi H, et al. Development of parkinsonian symptoms after discontinuation of carbamazepine in patients concurrently treated with risperidone: two case reports. Clin Neuropharmacol. 2001;24:358-360.

12. Rendic S. Summary of information on human CYP enzymes: human P450 metabolism data. Drug Metab Rev. 2002;34:83-448.

13. Hong CC, Tang BK, Hammond GL, et al. Cytochrome P450 1A2 (CYP1A2) activity and risk factors for breast cancer: a cross-sectional study. Breast Cancer Res. 2004;6:R352-365.

14. Cozza KL, Armstrong SC, Oesterheld JR. Concise guide to drug interaction principles for medical practice: cytochrome P450s, UGTS, p-glycoproteins. Washington, DC: American Psychiatric Press, Inc; 2003.

15. Kirchheiner J, Seeringer A. Clinical implications of pharmacogenetics of cytochrome P450 drug metabolizing enzymes. Biochim Biophys Acta. 2007;1770:489-494.

16. Kotlyar M, Brauer LH, Tracy TS, et al. Inhibition of CYP2D6 activity by bupropion. J Clin Psychopharmacol. 2005;25:226-229.

17. Uhr M, Tontsch A, Namendorf C, et al. Polymorphisms in the drug transporter gene ABCB1 predict antidepressant treatment response in depression. Neuron. 2008;57:203-209.

18. Bozina N, Kuzman MR, Medved V, et al. Associations between MDR1 gene polymorphisms and schizophrenia and therapeutic response to olanzapine in female schizophrenic patients. J Psychiatr Res. 2008;42:89-97.

19. Kim RB. Drugs as p-glycoprotein substrates, inhibitors, and inducers. Drug Metab Rev. 2002;34:47-54.

20. Hollenberg PF. Characteristics and common properties of inhibitors, inducers, and activators of CYP enzymes. Drug Metab Rev. 2002;34:17-35.

21. Physicians’ desk reference. 62nd ed. Montvale, NJ: Thomson Healthcare Inc.; 2007.

22. Heykants J, Huang ML, Mannens G, et al. The pharmacokinetics of risperidone in humans: a summary. J Clin Psychiatry. 1994;55 (suppl):13-7.

23. de Leon J, Bork J. Risperidone and cytochrome P450 3A. J Clin Psychiatry. 1997;58:450.-

24. Lane HY, Chang WH. Risperidone-carbamazepine interactions: is cytochrome P450 3A involved? J Clin Psychiatry. 1998;59:430-431.

25. Ejsing TB, Pedersen AD, Linnet K. P-glycoprotein interaction with risperidone and 9-OH-risperidone studied in vitro, in knock-out mice and in drug-drug interaction experiments. Hum Psychopharmacol. 2005;20:493-500.

26. Cousein E, Barthelemy C, Poullain S, et al. P-glycoprotein and cytochrome P450 3A4 involvement in risperidone transport using an in vitro Caco-2/TC7 model and an in vivo model. Prog Neuropsychopharmacol Biol Psychiatry. 2007;31:878-886.

27. Rostami-Hodjegan A, Amin AM, et al. Influence of dose, cigarette smoking, age, sex, and metabolic activity on plasma clozapine concentrations: a predictive model and nomograms to aid clozapine dose adjustment and to assess compliance in individual patients. J Clin Psychopharmacol. 2004;24:70-78.

28. Flockhart DA. Drug interactions: cytochrome P450 drug interaction table. Indiana University School of Medicine. 2007. Available at: http://medicine.iupui.edu/flockhart/table.htm. Accessed October 22, 2008.

Article PDF
Author and Disclosure Information

Jonathan M. Meyer, MD
Assistant professor, department of psychiatry, University of California, San Diego, staff psychiatrist, VA San Diego Healthcare System

Susan G. Leckband, RPh, BCPP
Assistant clinical professor, school of pharmacy, University of California, San Diego, psychiatric clinical pharmacist specialist, VA San Diego Healthcare System

Issue
Current Psychiatry - 08(01)
Publications
Page Number
47-53
Legacy Keywords
enzyme induction; antipsychotics; drug interactions; DDIs; p-glycoprotein; PGP; P450 CYP; CYP1A2; CYP 3A4; Jonathan Meyer; Susan Leckband
Sections
Author and Disclosure Information

Jonathan M. Meyer, MD
Assistant professor, department of psychiatry, University of California, San Diego, staff psychiatrist, VA San Diego Healthcare System

Susan G. Leckband, RPh, BCPP
Assistant clinical professor, school of pharmacy, University of California, San Diego, psychiatric clinical pharmacist specialist, VA San Diego Healthcare System

Author and Disclosure Information

Jonathan M. Meyer, MD
Assistant professor, department of psychiatry, University of California, San Diego, staff psychiatrist, VA San Diego Healthcare System

Susan G. Leckband, RPh, BCPP
Assistant clinical professor, school of pharmacy, University of California, San Diego, psychiatric clinical pharmacist specialist, VA San Diego Healthcare System

Article PDF
Article PDF

Mr. P, age 35 with schizophrenia and seizure disorder, has been maintained on risperidone, 6 mg qhs, and phenytoin, 300 mg qhs. For clinical reasons, the treating neurologist changes the anticonvulsant to divalproex. One week later, Mr. P presents to the emergency room complaining of jaw and neck stiffness.

Ms. K, age 43 with a history of schizoaffective disorder, bipolar type, and erratic medication adherence, is being treated with quetiapine, 600 mg at bedtime, and carbamazepine, 1,000 mg/d. Between appointments she stops taking carbamazepine, believing it is causing her to hear voices from her television. Two weeks later, the manager of Ms. K’s independent living facility tells the psychiatrist that the patient appears excessively sedated and has fallen twice in the past few days.

Mrs. T, a 39-year-old state hospital resident with schizoaffective disorder, bipolar type, has been treated with clozapine, 250 mg bid for 6 months; her most recent trough serum level was 492 ng/mL. She smokes 15 cigarettes/d. Two weeks after the hospital institutes a no-smoking policy, Mrs. T complains of excessive drooling and lightheadedness. Her trough clozapine level is now 875 ng/mL.

Discontinuing a medication that has enzyme-inducing effects presents a hidden problem for patients receiving antipsychotic pharmacotherapy. Certain hepatic enzymes responsible for antipsychotic metabolism—as well those involved in intercellular drug transport—are induced by medication or environmental exposures.1,2 Adding a medication that induces these enzymes to the regimen of a patient receiving antipsychotic therapy can result in markedly reduced serum antipsychotic levels, and discontinuing an inducing agent can result in increased antipsychotic levels.

Drug-drug interactions (DDIs) are a substantial contributor to adverse drug reactions (Box).3-7 Antipsychotic prescribing information highlights potential DDIs from the use of enzyme inhibitors and inducers but identifies only effects caused by adding a second agent. The prescriber remains the sole line of defense for monitoring for DDIs when discontinuing a medication that has inducing or inhibiting effects.

Most psychiatrists are aware that certain medications have clinically significant effects on cytochrome P450 (CYP) activity and of the potential for CYP inhibitors to generate DDIs. Clinicians often are aware of antidepressant medications’ CYP-inhibiting effects, know that levels of other medications will change when discontinuing a potent P450 inhibitor, and understand the need to increase dosages of medications influenced by such agents.8

However, few studies have evaluated the effects of enzyme induction on antipsychotic drug levels,9,10 and the literature rarely discusses changes in serum drug levels after loss of enzyme or drug transport induction.11 If unrecognized, these changes may have significant clinical consequences.

Box

Drug interactions: A common cause of nonadherence

Drug-drug interactions (DDIs) are a common and often preventable cause of morbidity and mortality. National surveillance data showed 700,000 emergency room visits related to adverse drug reactions (ADRs) in the 2 years from January 2004 through 2005.3 ADRs are particularly concerning for psychiatrists managing polypharmacy regimens for patients with severe mental disorders such as schizophrenia.

Literature on DDIs with antipsychotics focuses primarily on kinetic interactions that generate supratherapeutic drug levels.4,5 Because development of side effects is associated with reduced adherence, these kinetic interactions may increase the risk of adverse effects and lead to patients stopping the antipsychotic treatment.6,7

Two induction pathways

The primary mechanism underlying clinically significant DDIs occurs during CYP-mediated phase I metabolism. Molecules undergo oxidative conversion into metabolites that can be conjugated by phase II enzymes, generating more soluble forms that facilitate excretion.

The workhorse of human CYP metabolism is 3A4 (Table 1),12,13 which comprises 30% of hepatic activity and 70% of gut cytochrome activity.14 CYP 1A2 is responsible for 10% to 15% of CYP activity.

Both CYP 3A4 and 1A2 are inducible. A wide variety of medications induce 3A4 activity. The list of 1A2 inducers is shorter; the most common are aryl hydrocarbons from cigarette smoke and proton pump inhibitors.

CYP 2D6 accounts for 20% of hepatic cytochrome activity but is not inducible. CYP 2D6 is well known to psychiatrists because some selective serotonin reuptake inhibitors (SSRIs) and the non-SSRI antidepressant bupropion are potent inhibitors of this enzyme.15,16

P-glycoprotein (PGP) induction. Transmembrane shuttles such as P-glycoprotein (PGP) are an important component of drug disposition. PGP belongs to the family of ATP binding cassette (ABC) transporters that bring molecules across cellular barriers.17,18 It was first described in cancer cells that developed multiple drug resistance (MDR) and is often referred to as MDR1.19 PGP is encoded on human chromosome 7 and expressed in normal tissues, particularly in areas where cells seek to limit drug influx, such as those lining the luminal surface of the small and large intestine and those lining the blood-brain barrier and blood-testis barrier. The expression of PGP in hepatic cells promotes drug clearance by enhancing biliary drug excretion.

 

 

PGP is encoded on the same chromosome as CYP 3A4, and these 2 proteins frequently are expressed in the same cells, particularly in the intestinal lining and liver. Moreover, PGP is inducible, and there is substantial overlap between medications that are substrates for—or inducers of—PGP and CYP 3A4 activity. This makes it challenging to determine whether the kinetic effects of a second medication are the result of interference of 3A4, PGP, or both.

Polymorphisms in PGP activity may influence the penetration of psychotropic medications into the CNS. Studies indicate an association between certain PGP polymorphisms and treatment outcomes.17,18

Table 1

What induces CYP 1A2 and 3A4?

EnzymeDescriptionInducers*
CYP 1A2
  • Responsible for 10% to 15% of all CYP P450 activity
  • Located on chromosome 15
  • Low affinity/high capacity enzyme
  • Low affinity/high capacity enzyme
  • Prevalence of genetic polymorphisms conferring poor metabolizer status: 12% to 13%
Aryl hydrocarbons (smoking), protonpump inhibitors (omeprazole > lansoprazole > pantoprazole), modafinil, St. John’s wort, chargrilled meat, cruciferous vegetables such as broccoli and cabbage, flavones, protein supplements
CYP 3A4
  • Responsible for 30% of hepatic CYP 450 activity, 70% of gut cytochrome activity
  • Located on chromosome 7 (same as PGP)
  • Low affinity/high capacity enzyme
  • Little evidence for significant functional polymorphisms
Carbamazepine, phenytoin, phenobarbital, rifampin, oxcarbazepine, efavirenz, glucocorticoids, modafinil, nevirapine, pioglitazone, St. John’s wort
* Listed in order from strongest to weakest induction
CYP: cytochrome P450; PGP: P-glycoprotein
Source: References 12,13

Stopping an inducer

In general, inducers of CYP enzymes stimulate gene transcription within hours of exposure; maximum transcriptional activity occurs after 10 to 12 hours of exposure. As transcription increases, the concentration of the CYP mRNA transcript steadily accumulates, as does concentration of CYP protein.

After an inducer is discontinued, transcription returns to basal levels within 18 hours; however, the degradation of CYP proteins is a first-order process, with a half-life of 8 to 30 hours. As a result, the decrease in cellular CYP concentration—and the level of activity—lags behind the decreased synthesis from reduced mRNA levels.

As with other first-order kinetic processes, the expected decrease in CYP activity will require 5 half-lives to reach the new steady state (ie, back to baseline CYP activity). This suggests that drug levels previously decreased by CYP induction will reach their peak on average 1 to 2 weeks after the inducer is discontinued.20

Interactions with antipsychotics

Effects on serum antipsychotic levels caused by discontinuing a CYP or PGP inducer can be predicted from data on decreases in antipsychotic levels following inducer exposure. Except for ziprasidone and paliperidone, most atypical antipsychotics are prone to substantial decreases during concomitant inducer use (Table 2).21

The effect of enzyme inducers on risperidone is particularly interesting. Conversion of risperidone to its active metabolite 9-OH risperidone (paliperidone) occurs primarily via 2D6,22 yet concurrent use of carbamazepine—a potent CYP 3A4 inducer—results in a 50% decrease in the concentration of the active moiety (risperidone plus 9-OH risperidone). This finding and other early investigations suggested that CYP 3A had a role in risperidone metabolism,23,24 but these early studies and case series often involved molecules that had activity at both 3A4 and PGP. Further research clarified that effects on PGP—and not 3A4—are responsible for the changes in risperidone metabolism observed with the use of carbamazepine and other medications.25,26

Induction in case patients: Follow-up. Regardless of whether induction is mediated by ≥1 metabolic pathways, the loss of the inducer will result in serum antipsychotic increases that are proportional to the initial decrease.20 For example, with risperidone, the expected decrease is 50%. Therefore, after Mr. P stopped taking phenytoin, his serum risperidone level would be expected to double, which resulted in extrapyramidal side effects.

Quetiapine clearance is increased 5-fold by inducer exposure, so a clinician treating Ms. K would expect a marked increase in somnolence—and possibly orthostasis—as serum quetiapine levels peak 1 to 2 weeks following carbamazepine discontinuation.

The effects of smoking cessation on serum clozapine levels have been well-documented.1,27 Clinicians should anticipate median increases in serum clozapine levels of 55% after a patient discontinues smoking (aryl hydrocarbon exposure), but changes vary substantially among individuals. Mrs. T’s serum clozapine increased approximately 78%.

Careful clinical monitoring and slow downward adjustment of antipsychotic doses could have prevented the adverse effects these 3 patients experienced after loss of CYP/PGP induction and the consequences those side effects present for future medication adherence. When loss of induction is unplanned—as when Ms. K stopped taking carbamazepine but continued quetiapine—clinicians need to be alert to the fact that the patient was prescribed an inducer and include the loss of induction as a hypothesis for the patient’s somnolence.

 

 

Table 2

Effects of CYP/PGP induction on atypical antipsychotics

AntipsychoticMetabolic pathwaysEffect of induction
Aripiprazole2D6 and 3A4 convert aripiprazole to active metabolite dehydro-aripiprazole3A4 induction decreases maximum concentration of aripiprazole and metabolite by 70%
ClozapineMultiple enzymes convert clozapine to N-desmethylclozapine; mean contributions of CYP 1A2, 2C19, 3A4, 2C9, and 2D6 are 30%, 24%, 22%, 12%, and 6%, respectively, with CYP 1A2 predominantly involved at low concentrationsLoss of smoking-related 1A2 induction results in 50% increase in serum levels
OlanzapineDirect glucuronidation or 1A2-mediated oxidation to N-desmethlyolanzapineCarbamazepine use increases clearance by 50%. Olanzapine concentration:dose ratio is about 5-fold lower in smokers (7.9 +/- 2.6) than in nonsmokers (1.56 +/- 1.1; P
Paliperidone59% excreted unchanged in urine; phase I metabolism accounts for ≤10% of drug clearanceUnlikely to significantly impact levels, but impact of PGP induction is unknown
Quetiapine3A4-mediated sulfoxidation to inactive metabolite is primary pathway, but numerous metabolites noted, with 1 active metabolite (norquetiapine)Phenytoin increases clearance 5-fold
Risperidone2D6 converts risperidone to active metabolite 9-OH risperidoneIn a drug interaction study of risperidone, 6 mg/d for 3 weeks, followed by 3 weeks of carbamazepine, active moiety concentration was decreased by about 50%
Ziprasidone3A4 (~1/3); aldehyde oxidase (~2/3)Approximately 35% decrease in ziprasidone exposure by carbamazepine
CYP: cytochrome P450; PGP: P-glycoprotein
Source: Reference 21

Clinical considerations

In the absence of detailed data on antipsychotic metabolism, clinicians can make intelligent decisions regarding potential DDIs by:

  • knowing the extent of induction by common offenders (such as carbamazepine or phenytoin) documented in the medication’s prescribing information or demonstrated through convincing case reports or case series
  • memorizing the list of CYP 1A2 and CYP 3A4/PGP inducers.

Although the list of CYP 1A2 and CYP 3A4/PGP inducers is short, it is essential for clinicians to consult a readily available source of this information that is periodically updated to account for newer medications, such as the online table maintained by Flockhart (see Related Resources).28

Patients who may be susceptible to effects from loss of enzyme induction (including smokers receiving olanzapine or clozapine or others taking 3A4/PGP inducers) must be identified, and plans made for dosage adjustments if inducing agents are discontinued for a sufficient time (≥1 week) to result in downregulation of CYP or PGP activity. A slow taper of the antipsychotic over 1 to 2 weeks to the new target dose should compensate for loss of enzyme or PGP induction.

For newer antipsychotic medications with limited data, the proposed discontinuation of an inducer should, at the minimum, prompt a discussion between the psychiatrist and patient regarding the expected increase in serum antipsychotic levels and potential adverse effects that may result. Clinicians also must make every attempt to stay apprised of a patient’s current medications, bearing in mind that another provider may prescribe an inducer. Patients with schizophrenia always should be educated to contact the psychiatrist following any change in medication regimen, placing particular emphasis on the 1 or 2 medications that are known to be implicated in DDIs with the patient’s current antipsychotic.

Related Resources

  • Flockhart DA. Drug interactions: Cytochrome P450 drug interaction table. Indiana University School of Medicine. 2007. http://medicine.iupui.edu/flockhart/table.htm.
  • Cozza KL, Armstrong SC, Oesterheld JR. Concise guide to drug interaction principles for medical practice: Cytochrome P450s, UGTS, p-glycoproteins. Washington, DC: American Psychiatric Press, Inc; 2003.
Drug brand names

  • Aripiprazole • Abilify
  • Bupropion • Wellbutrin
  • Carbamazepine • Carbatrol, Tegretol
  • Clozapine • Clozaril
  • Divalproex • Depakote
  • Efavirenz • Sustiva
  • Lansoprazole • Prevacid
  • Modafinil • Provigil
  • Nevirapine • Viramune
  • Olanzapine • Zyprexa
  • Omeprazole • Prilosec
  • Oxcarbazepine • Trileptal
  • Paliperidone • Invega
  • Pantoprazole • Protonix
  • Phenobarbital • Barbita, Luminal, others
  • Phenytoin • Dilantin
  • Pioglitazone • Actos
  • Quetiapine • Seroquel
  • Rifampin • Rifadin, Rimactane
  • Risperidone • Risperdal
  • Ziprasidone • Geodon
Disclosure

Dr. Meyer receives grant/research support from the National Institute of Mental Health, Pfizer Inc., and the University of California. He is a consultant to Bristol-Myers Squibb, Organon, Vanda Pharmaceuticals, and Wyeth, and a speaker for AstraZeneca, Bristol-Myers Squibb, Dainippon Sumitomo Pharma, and Pfizer Inc.

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

Mr. P, age 35 with schizophrenia and seizure disorder, has been maintained on risperidone, 6 mg qhs, and phenytoin, 300 mg qhs. For clinical reasons, the treating neurologist changes the anticonvulsant to divalproex. One week later, Mr. P presents to the emergency room complaining of jaw and neck stiffness.

Ms. K, age 43 with a history of schizoaffective disorder, bipolar type, and erratic medication adherence, is being treated with quetiapine, 600 mg at bedtime, and carbamazepine, 1,000 mg/d. Between appointments she stops taking carbamazepine, believing it is causing her to hear voices from her television. Two weeks later, the manager of Ms. K’s independent living facility tells the psychiatrist that the patient appears excessively sedated and has fallen twice in the past few days.

Mrs. T, a 39-year-old state hospital resident with schizoaffective disorder, bipolar type, has been treated with clozapine, 250 mg bid for 6 months; her most recent trough serum level was 492 ng/mL. She smokes 15 cigarettes/d. Two weeks after the hospital institutes a no-smoking policy, Mrs. T complains of excessive drooling and lightheadedness. Her trough clozapine level is now 875 ng/mL.

Discontinuing a medication that has enzyme-inducing effects presents a hidden problem for patients receiving antipsychotic pharmacotherapy. Certain hepatic enzymes responsible for antipsychotic metabolism—as well those involved in intercellular drug transport—are induced by medication or environmental exposures.1,2 Adding a medication that induces these enzymes to the regimen of a patient receiving antipsychotic therapy can result in markedly reduced serum antipsychotic levels, and discontinuing an inducing agent can result in increased antipsychotic levels.

Drug-drug interactions (DDIs) are a substantial contributor to adverse drug reactions (Box).3-7 Antipsychotic prescribing information highlights potential DDIs from the use of enzyme inhibitors and inducers but identifies only effects caused by adding a second agent. The prescriber remains the sole line of defense for monitoring for DDIs when discontinuing a medication that has inducing or inhibiting effects.

Most psychiatrists are aware that certain medications have clinically significant effects on cytochrome P450 (CYP) activity and of the potential for CYP inhibitors to generate DDIs. Clinicians often are aware of antidepressant medications’ CYP-inhibiting effects, know that levels of other medications will change when discontinuing a potent P450 inhibitor, and understand the need to increase dosages of medications influenced by such agents.8

However, few studies have evaluated the effects of enzyme induction on antipsychotic drug levels,9,10 and the literature rarely discusses changes in serum drug levels after loss of enzyme or drug transport induction.11 If unrecognized, these changes may have significant clinical consequences.

Box

Drug interactions: A common cause of nonadherence

Drug-drug interactions (DDIs) are a common and often preventable cause of morbidity and mortality. National surveillance data showed 700,000 emergency room visits related to adverse drug reactions (ADRs) in the 2 years from January 2004 through 2005.3 ADRs are particularly concerning for psychiatrists managing polypharmacy regimens for patients with severe mental disorders such as schizophrenia.

Literature on DDIs with antipsychotics focuses primarily on kinetic interactions that generate supratherapeutic drug levels.4,5 Because development of side effects is associated with reduced adherence, these kinetic interactions may increase the risk of adverse effects and lead to patients stopping the antipsychotic treatment.6,7

Two induction pathways

The primary mechanism underlying clinically significant DDIs occurs during CYP-mediated phase I metabolism. Molecules undergo oxidative conversion into metabolites that can be conjugated by phase II enzymes, generating more soluble forms that facilitate excretion.

The workhorse of human CYP metabolism is 3A4 (Table 1),12,13 which comprises 30% of hepatic activity and 70% of gut cytochrome activity.14 CYP 1A2 is responsible for 10% to 15% of CYP activity.

Both CYP 3A4 and 1A2 are inducible. A wide variety of medications induce 3A4 activity. The list of 1A2 inducers is shorter; the most common are aryl hydrocarbons from cigarette smoke and proton pump inhibitors.

CYP 2D6 accounts for 20% of hepatic cytochrome activity but is not inducible. CYP 2D6 is well known to psychiatrists because some selective serotonin reuptake inhibitors (SSRIs) and the non-SSRI antidepressant bupropion are potent inhibitors of this enzyme.15,16

P-glycoprotein (PGP) induction. Transmembrane shuttles such as P-glycoprotein (PGP) are an important component of drug disposition. PGP belongs to the family of ATP binding cassette (ABC) transporters that bring molecules across cellular barriers.17,18 It was first described in cancer cells that developed multiple drug resistance (MDR) and is often referred to as MDR1.19 PGP is encoded on human chromosome 7 and expressed in normal tissues, particularly in areas where cells seek to limit drug influx, such as those lining the luminal surface of the small and large intestine and those lining the blood-brain barrier and blood-testis barrier. The expression of PGP in hepatic cells promotes drug clearance by enhancing biliary drug excretion.

 

 

PGP is encoded on the same chromosome as CYP 3A4, and these 2 proteins frequently are expressed in the same cells, particularly in the intestinal lining and liver. Moreover, PGP is inducible, and there is substantial overlap between medications that are substrates for—or inducers of—PGP and CYP 3A4 activity. This makes it challenging to determine whether the kinetic effects of a second medication are the result of interference of 3A4, PGP, or both.

Polymorphisms in PGP activity may influence the penetration of psychotropic medications into the CNS. Studies indicate an association between certain PGP polymorphisms and treatment outcomes.17,18

Table 1

What induces CYP 1A2 and 3A4?

EnzymeDescriptionInducers*
CYP 1A2
  • Responsible for 10% to 15% of all CYP P450 activity
  • Located on chromosome 15
  • Low affinity/high capacity enzyme
  • Low affinity/high capacity enzyme
  • Prevalence of genetic polymorphisms conferring poor metabolizer status: 12% to 13%
Aryl hydrocarbons (smoking), protonpump inhibitors (omeprazole > lansoprazole > pantoprazole), modafinil, St. John’s wort, chargrilled meat, cruciferous vegetables such as broccoli and cabbage, flavones, protein supplements
CYP 3A4
  • Responsible for 30% of hepatic CYP 450 activity, 70% of gut cytochrome activity
  • Located on chromosome 7 (same as PGP)
  • Low affinity/high capacity enzyme
  • Little evidence for significant functional polymorphisms
Carbamazepine, phenytoin, phenobarbital, rifampin, oxcarbazepine, efavirenz, glucocorticoids, modafinil, nevirapine, pioglitazone, St. John’s wort
* Listed in order from strongest to weakest induction
CYP: cytochrome P450; PGP: P-glycoprotein
Source: References 12,13

Stopping an inducer

In general, inducers of CYP enzymes stimulate gene transcription within hours of exposure; maximum transcriptional activity occurs after 10 to 12 hours of exposure. As transcription increases, the concentration of the CYP mRNA transcript steadily accumulates, as does concentration of CYP protein.

After an inducer is discontinued, transcription returns to basal levels within 18 hours; however, the degradation of CYP proteins is a first-order process, with a half-life of 8 to 30 hours. As a result, the decrease in cellular CYP concentration—and the level of activity—lags behind the decreased synthesis from reduced mRNA levels.

As with other first-order kinetic processes, the expected decrease in CYP activity will require 5 half-lives to reach the new steady state (ie, back to baseline CYP activity). This suggests that drug levels previously decreased by CYP induction will reach their peak on average 1 to 2 weeks after the inducer is discontinued.20

Interactions with antipsychotics

Effects on serum antipsychotic levels caused by discontinuing a CYP or PGP inducer can be predicted from data on decreases in antipsychotic levels following inducer exposure. Except for ziprasidone and paliperidone, most atypical antipsychotics are prone to substantial decreases during concomitant inducer use (Table 2).21

The effect of enzyme inducers on risperidone is particularly interesting. Conversion of risperidone to its active metabolite 9-OH risperidone (paliperidone) occurs primarily via 2D6,22 yet concurrent use of carbamazepine—a potent CYP 3A4 inducer—results in a 50% decrease in the concentration of the active moiety (risperidone plus 9-OH risperidone). This finding and other early investigations suggested that CYP 3A had a role in risperidone metabolism,23,24 but these early studies and case series often involved molecules that had activity at both 3A4 and PGP. Further research clarified that effects on PGP—and not 3A4—are responsible for the changes in risperidone metabolism observed with the use of carbamazepine and other medications.25,26

Induction in case patients: Follow-up. Regardless of whether induction is mediated by ≥1 metabolic pathways, the loss of the inducer will result in serum antipsychotic increases that are proportional to the initial decrease.20 For example, with risperidone, the expected decrease is 50%. Therefore, after Mr. P stopped taking phenytoin, his serum risperidone level would be expected to double, which resulted in extrapyramidal side effects.

Quetiapine clearance is increased 5-fold by inducer exposure, so a clinician treating Ms. K would expect a marked increase in somnolence—and possibly orthostasis—as serum quetiapine levels peak 1 to 2 weeks following carbamazepine discontinuation.

The effects of smoking cessation on serum clozapine levels have been well-documented.1,27 Clinicians should anticipate median increases in serum clozapine levels of 55% after a patient discontinues smoking (aryl hydrocarbon exposure), but changes vary substantially among individuals. Mrs. T’s serum clozapine increased approximately 78%.

Careful clinical monitoring and slow downward adjustment of antipsychotic doses could have prevented the adverse effects these 3 patients experienced after loss of CYP/PGP induction and the consequences those side effects present for future medication adherence. When loss of induction is unplanned—as when Ms. K stopped taking carbamazepine but continued quetiapine—clinicians need to be alert to the fact that the patient was prescribed an inducer and include the loss of induction as a hypothesis for the patient’s somnolence.

 

 

Table 2

Effects of CYP/PGP induction on atypical antipsychotics

AntipsychoticMetabolic pathwaysEffect of induction
Aripiprazole2D6 and 3A4 convert aripiprazole to active metabolite dehydro-aripiprazole3A4 induction decreases maximum concentration of aripiprazole and metabolite by 70%
ClozapineMultiple enzymes convert clozapine to N-desmethylclozapine; mean contributions of CYP 1A2, 2C19, 3A4, 2C9, and 2D6 are 30%, 24%, 22%, 12%, and 6%, respectively, with CYP 1A2 predominantly involved at low concentrationsLoss of smoking-related 1A2 induction results in 50% increase in serum levels
OlanzapineDirect glucuronidation or 1A2-mediated oxidation to N-desmethlyolanzapineCarbamazepine use increases clearance by 50%. Olanzapine concentration:dose ratio is about 5-fold lower in smokers (7.9 +/- 2.6) than in nonsmokers (1.56 +/- 1.1; P
Paliperidone59% excreted unchanged in urine; phase I metabolism accounts for ≤10% of drug clearanceUnlikely to significantly impact levels, but impact of PGP induction is unknown
Quetiapine3A4-mediated sulfoxidation to inactive metabolite is primary pathway, but numerous metabolites noted, with 1 active metabolite (norquetiapine)Phenytoin increases clearance 5-fold
Risperidone2D6 converts risperidone to active metabolite 9-OH risperidoneIn a drug interaction study of risperidone, 6 mg/d for 3 weeks, followed by 3 weeks of carbamazepine, active moiety concentration was decreased by about 50%
Ziprasidone3A4 (~1/3); aldehyde oxidase (~2/3)Approximately 35% decrease in ziprasidone exposure by carbamazepine
CYP: cytochrome P450; PGP: P-glycoprotein
Source: Reference 21

Clinical considerations

In the absence of detailed data on antipsychotic metabolism, clinicians can make intelligent decisions regarding potential DDIs by:

  • knowing the extent of induction by common offenders (such as carbamazepine or phenytoin) documented in the medication’s prescribing information or demonstrated through convincing case reports or case series
  • memorizing the list of CYP 1A2 and CYP 3A4/PGP inducers.

Although the list of CYP 1A2 and CYP 3A4/PGP inducers is short, it is essential for clinicians to consult a readily available source of this information that is periodically updated to account for newer medications, such as the online table maintained by Flockhart (see Related Resources).28

Patients who may be susceptible to effects from loss of enzyme induction (including smokers receiving olanzapine or clozapine or others taking 3A4/PGP inducers) must be identified, and plans made for dosage adjustments if inducing agents are discontinued for a sufficient time (≥1 week) to result in downregulation of CYP or PGP activity. A slow taper of the antipsychotic over 1 to 2 weeks to the new target dose should compensate for loss of enzyme or PGP induction.

For newer antipsychotic medications with limited data, the proposed discontinuation of an inducer should, at the minimum, prompt a discussion between the psychiatrist and patient regarding the expected increase in serum antipsychotic levels and potential adverse effects that may result. Clinicians also must make every attempt to stay apprised of a patient’s current medications, bearing in mind that another provider may prescribe an inducer. Patients with schizophrenia always should be educated to contact the psychiatrist following any change in medication regimen, placing particular emphasis on the 1 or 2 medications that are known to be implicated in DDIs with the patient’s current antipsychotic.

Related Resources

  • Flockhart DA. Drug interactions: Cytochrome P450 drug interaction table. Indiana University School of Medicine. 2007. http://medicine.iupui.edu/flockhart/table.htm.
  • Cozza KL, Armstrong SC, Oesterheld JR. Concise guide to drug interaction principles for medical practice: Cytochrome P450s, UGTS, p-glycoproteins. Washington, DC: American Psychiatric Press, Inc; 2003.
Drug brand names

  • Aripiprazole • Abilify
  • Bupropion • Wellbutrin
  • Carbamazepine • Carbatrol, Tegretol
  • Clozapine • Clozaril
  • Divalproex • Depakote
  • Efavirenz • Sustiva
  • Lansoprazole • Prevacid
  • Modafinil • Provigil
  • Nevirapine • Viramune
  • Olanzapine • Zyprexa
  • Omeprazole • Prilosec
  • Oxcarbazepine • Trileptal
  • Paliperidone • Invega
  • Pantoprazole • Protonix
  • Phenobarbital • Barbita, Luminal, others
  • Phenytoin • Dilantin
  • Pioglitazone • Actos
  • Quetiapine • Seroquel
  • Rifampin • Rifadin, Rimactane
  • Risperidone • Risperdal
  • Ziprasidone • Geodon
Disclosure

Dr. Meyer receives grant/research support from the National Institute of Mental Health, Pfizer Inc., and the University of California. He is a consultant to Bristol-Myers Squibb, Organon, Vanda Pharmaceuticals, and Wyeth, and a speaker for AstraZeneca, Bristol-Myers Squibb, Dainippon Sumitomo Pharma, and Pfizer Inc.

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

References

1. Meyer JM. Individual changes in clozapine levels after smoking cessation: results and a predictive model. J Clin Psychopharmacol. 2001;21:569-574.

2. Wong YW, Yeh C, Thyrum PT. The effects of concomitant phenytoin administration on the steady-state pharmacokinetics of quetiapine. J Clin Psychopharmacol. 2001;21:89-93.

3. Budnitz DS, Pollock DA, Weidenbach KN, et al. National surveillance of emergency department visits for outpatient adverse drug events. JAMA. 2006;296:1858-1866.

4. Prior TI, Baker GB. Interactions between the cytochrome P450 system and the second-generation antipsychotics. J Psychiatry Neurosci. 2003;28:99-112.

5. Spina E, de Leon J. Metabolic drug interactions with newer antipsychotics: a comparative review. Basic Clin Pharmacol Toxicol. 2007;100:4-22.

6. Preskorn SH. Drug-drug interactions: proof of relevance (part II): cause of tolerability problems or noncompliance. J Psychiatr Pract. 2005;11:397-401.

7. Weiden PJ, Mackell JA, McDonnell D. Obesity as a risk factor for antipsychotic noncompliance. Schizophr Res. 2004;66:51-7.

8. Preskorn SH, Flockhart D. 2006 guide to psychiatric drug interactions. Prim Psychiatry. 2006;13:35-64.

9. Spina E, Perucca E. Clinical significance of pharmacokinetic interactions between antiepileptic and psychotropic drugs. Epilepsia. 2002;43(suppl 2):37-44.

10. Meyer JM. Drug-drug interactions with antipsychotics. CNS Spectr. 2007;12:6-9.

11. Takahashi H, Yoshida K, Higuchi H, et al. Development of parkinsonian symptoms after discontinuation of carbamazepine in patients concurrently treated with risperidone: two case reports. Clin Neuropharmacol. 2001;24:358-360.

12. Rendic S. Summary of information on human CYP enzymes: human P450 metabolism data. Drug Metab Rev. 2002;34:83-448.

13. Hong CC, Tang BK, Hammond GL, et al. Cytochrome P450 1A2 (CYP1A2) activity and risk factors for breast cancer: a cross-sectional study. Breast Cancer Res. 2004;6:R352-365.

14. Cozza KL, Armstrong SC, Oesterheld JR. Concise guide to drug interaction principles for medical practice: cytochrome P450s, UGTS, p-glycoproteins. Washington, DC: American Psychiatric Press, Inc; 2003.

15. Kirchheiner J, Seeringer A. Clinical implications of pharmacogenetics of cytochrome P450 drug metabolizing enzymes. Biochim Biophys Acta. 2007;1770:489-494.

16. Kotlyar M, Brauer LH, Tracy TS, et al. Inhibition of CYP2D6 activity by bupropion. J Clin Psychopharmacol. 2005;25:226-229.

17. Uhr M, Tontsch A, Namendorf C, et al. Polymorphisms in the drug transporter gene ABCB1 predict antidepressant treatment response in depression. Neuron. 2008;57:203-209.

18. Bozina N, Kuzman MR, Medved V, et al. Associations between MDR1 gene polymorphisms and schizophrenia and therapeutic response to olanzapine in female schizophrenic patients. J Psychiatr Res. 2008;42:89-97.

19. Kim RB. Drugs as p-glycoprotein substrates, inhibitors, and inducers. Drug Metab Rev. 2002;34:47-54.

20. Hollenberg PF. Characteristics and common properties of inhibitors, inducers, and activators of CYP enzymes. Drug Metab Rev. 2002;34:17-35.

21. Physicians’ desk reference. 62nd ed. Montvale, NJ: Thomson Healthcare Inc.; 2007.

22. Heykants J, Huang ML, Mannens G, et al. The pharmacokinetics of risperidone in humans: a summary. J Clin Psychiatry. 1994;55 (suppl):13-7.

23. de Leon J, Bork J. Risperidone and cytochrome P450 3A. J Clin Psychiatry. 1997;58:450.-

24. Lane HY, Chang WH. Risperidone-carbamazepine interactions: is cytochrome P450 3A involved? J Clin Psychiatry. 1998;59:430-431.

25. Ejsing TB, Pedersen AD, Linnet K. P-glycoprotein interaction with risperidone and 9-OH-risperidone studied in vitro, in knock-out mice and in drug-drug interaction experiments. Hum Psychopharmacol. 2005;20:493-500.

26. Cousein E, Barthelemy C, Poullain S, et al. P-glycoprotein and cytochrome P450 3A4 involvement in risperidone transport using an in vitro Caco-2/TC7 model and an in vivo model. Prog Neuropsychopharmacol Biol Psychiatry. 2007;31:878-886.

27. Rostami-Hodjegan A, Amin AM, et al. Influence of dose, cigarette smoking, age, sex, and metabolic activity on plasma clozapine concentrations: a predictive model and nomograms to aid clozapine dose adjustment and to assess compliance in individual patients. J Clin Psychopharmacol. 2004;24:70-78.

28. Flockhart DA. Drug interactions: cytochrome P450 drug interaction table. Indiana University School of Medicine. 2007. Available at: http://medicine.iupui.edu/flockhart/table.htm. Accessed October 22, 2008.

References

1. Meyer JM. Individual changes in clozapine levels after smoking cessation: results and a predictive model. J Clin Psychopharmacol. 2001;21:569-574.

2. Wong YW, Yeh C, Thyrum PT. The effects of concomitant phenytoin administration on the steady-state pharmacokinetics of quetiapine. J Clin Psychopharmacol. 2001;21:89-93.

3. Budnitz DS, Pollock DA, Weidenbach KN, et al. National surveillance of emergency department visits for outpatient adverse drug events. JAMA. 2006;296:1858-1866.

4. Prior TI, Baker GB. Interactions between the cytochrome P450 system and the second-generation antipsychotics. J Psychiatry Neurosci. 2003;28:99-112.

5. Spina E, de Leon J. Metabolic drug interactions with newer antipsychotics: a comparative review. Basic Clin Pharmacol Toxicol. 2007;100:4-22.

6. Preskorn SH. Drug-drug interactions: proof of relevance (part II): cause of tolerability problems or noncompliance. J Psychiatr Pract. 2005;11:397-401.

7. Weiden PJ, Mackell JA, McDonnell D. Obesity as a risk factor for antipsychotic noncompliance. Schizophr Res. 2004;66:51-7.

8. Preskorn SH, Flockhart D. 2006 guide to psychiatric drug interactions. Prim Psychiatry. 2006;13:35-64.

9. Spina E, Perucca E. Clinical significance of pharmacokinetic interactions between antiepileptic and psychotropic drugs. Epilepsia. 2002;43(suppl 2):37-44.

10. Meyer JM. Drug-drug interactions with antipsychotics. CNS Spectr. 2007;12:6-9.

11. Takahashi H, Yoshida K, Higuchi H, et al. Development of parkinsonian symptoms after discontinuation of carbamazepine in patients concurrently treated with risperidone: two case reports. Clin Neuropharmacol. 2001;24:358-360.

12. Rendic S. Summary of information on human CYP enzymes: human P450 metabolism data. Drug Metab Rev. 2002;34:83-448.

13. Hong CC, Tang BK, Hammond GL, et al. Cytochrome P450 1A2 (CYP1A2) activity and risk factors for breast cancer: a cross-sectional study. Breast Cancer Res. 2004;6:R352-365.

14. Cozza KL, Armstrong SC, Oesterheld JR. Concise guide to drug interaction principles for medical practice: cytochrome P450s, UGTS, p-glycoproteins. Washington, DC: American Psychiatric Press, Inc; 2003.

15. Kirchheiner J, Seeringer A. Clinical implications of pharmacogenetics of cytochrome P450 drug metabolizing enzymes. Biochim Biophys Acta. 2007;1770:489-494.

16. Kotlyar M, Brauer LH, Tracy TS, et al. Inhibition of CYP2D6 activity by bupropion. J Clin Psychopharmacol. 2005;25:226-229.

17. Uhr M, Tontsch A, Namendorf C, et al. Polymorphisms in the drug transporter gene ABCB1 predict antidepressant treatment response in depression. Neuron. 2008;57:203-209.

18. Bozina N, Kuzman MR, Medved V, et al. Associations between MDR1 gene polymorphisms and schizophrenia and therapeutic response to olanzapine in female schizophrenic patients. J Psychiatr Res. 2008;42:89-97.

19. Kim RB. Drugs as p-glycoprotein substrates, inhibitors, and inducers. Drug Metab Rev. 2002;34:47-54.

20. Hollenberg PF. Characteristics and common properties of inhibitors, inducers, and activators of CYP enzymes. Drug Metab Rev. 2002;34:17-35.

21. Physicians’ desk reference. 62nd ed. Montvale, NJ: Thomson Healthcare Inc.; 2007.

22. Heykants J, Huang ML, Mannens G, et al. The pharmacokinetics of risperidone in humans: a summary. J Clin Psychiatry. 1994;55 (suppl):13-7.

23. de Leon J, Bork J. Risperidone and cytochrome P450 3A. J Clin Psychiatry. 1997;58:450.-

24. Lane HY, Chang WH. Risperidone-carbamazepine interactions: is cytochrome P450 3A involved? J Clin Psychiatry. 1998;59:430-431.

25. Ejsing TB, Pedersen AD, Linnet K. P-glycoprotein interaction with risperidone and 9-OH-risperidone studied in vitro, in knock-out mice and in drug-drug interaction experiments. Hum Psychopharmacol. 2005;20:493-500.

26. Cousein E, Barthelemy C, Poullain S, et al. P-glycoprotein and cytochrome P450 3A4 involvement in risperidone transport using an in vitro Caco-2/TC7 model and an in vivo model. Prog Neuropsychopharmacol Biol Psychiatry. 2007;31:878-886.

27. Rostami-Hodjegan A, Amin AM, et al. Influence of dose, cigarette smoking, age, sex, and metabolic activity on plasma clozapine concentrations: a predictive model and nomograms to aid clozapine dose adjustment and to assess compliance in individual patients. J Clin Psychopharmacol. 2004;24:70-78.

28. Flockhart DA. Drug interactions: cytochrome P450 drug interaction table. Indiana University School of Medicine. 2007. Available at: http://medicine.iupui.edu/flockhart/table.htm. Accessed October 22, 2008.

Issue
Current Psychiatry - 08(01)
Issue
Current Psychiatry - 08(01)
Page Number
47-53
Page Number
47-53
Publications
Publications
Article Type
Display Headline
Loss of enzyme induction: Ups and downs of a hidden drug-drug interaction
Display Headline
Loss of enzyme induction: Ups and downs of a hidden drug-drug interaction
Legacy Keywords
enzyme induction; antipsychotics; drug interactions; DDIs; p-glycoprotein; PGP; P450 CYP; CYP1A2; CYP 3A4; Jonathan Meyer; Susan Leckband
Legacy Keywords
enzyme induction; antipsychotics; drug interactions; DDIs; p-glycoprotein; PGP; P450 CYP; CYP1A2; CYP 3A4; Jonathan Meyer; Susan Leckband
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media