User login
Welcome to Current Psychiatry, a leading source of information, online and in print, for practitioners of psychiatry and its related subspecialties, including addiction psychiatry, child and adolescent psychiatry, and geriatric psychiatry. This Web site contains evidence-based reviews of the prevention, diagnosis, and treatment of mental illness and psychological disorders; case reports; updates on psychopharmacology; news about the specialty of psychiatry; pearls for practice; and other topics of interest and use to this audience.
Dear Drupal User: You're seeing this because you're logged in to Drupal, and not redirected to MDedge.com/psychiatry.
Depression
adolescent depression
adolescent major depressive disorder
adolescent schizophrenia
adolescent with major depressive disorder
animals
autism
baby
brexpiprazole
child
child bipolar
child depression
child schizophrenia
children with bipolar disorder
children with depression
children with major depressive disorder
compulsive behaviors
cure
elderly bipolar
elderly depression
elderly major depressive disorder
elderly schizophrenia
elderly with dementia
first break
first episode
gambling
gaming
geriatric depression
geriatric major depressive disorder
geriatric schizophrenia
infant
kid
major depressive disorder
major depressive disorder in adolescents
major depressive disorder in children
parenting
pediatric
pediatric bipolar
pediatric depression
pediatric major depressive disorder
pediatric schizophrenia
pregnancy
pregnant
rexulti
skin care
teen
wine
section[contains(@class, 'nav-hidden')]
footer[@id='footer']
div[contains(@class, 'pane-pub-article-current-psychiatry')]
div[contains(@class, 'pane-pub-home-current-psychiatry')]
div[contains(@class, 'pane-pub-topic-current-psychiatry')]
div[contains(@class, 'panel-panel-inner')]
div[contains(@class, 'pane-node-field-article-topics')]
section[contains(@class, 'footer-nav-section-wrapper')]
Why off-label antipsychotics remain first-choice drugs for delirium
Delirium is a medical emergency that needs to be identified and treated vigorously. Antipsychotics—including haloperidol and atypical agents—effectively manage a wide spectrum of delirium symptoms and are an essential component in the standard multimodal approach.1 Even so, antipsychotics are not FDA-approved for treating delirium, and evidence on their safety in medically ill patients is limited—particularly in the elderly, in whom delirium occurs most often.
The FDA has warned of increased risk of death when atypical antipsychotics are used to treat behavioral disturbances in elderly patients with dementia.2 Similarly, a retrospective study of elderly patients taking antipsychotics found higher mortality rates associated with typical antipsychotics than with atypicals.3
This article discusses the risks and benefits of using antipsychotics to manage delirium. Based on the literature and clinical experience, we offer recommendations on choosing among the available agents and avoiding side effects.
A challenging diagnosis
Delirium is a neuropsychiatric syndrome precipitated by an underlying medical condition or a medication effect on the brain. Its characteristic symptoms—abrupt onset of disturbed consciousness, attention, cognition, and perception—tend to fluctuate during the day. Delirium most often occurs in elderly patients (Box)1,4-7—particularly with dementia—but also occurs in younger patients with serious illnesses such as cancer or HIV-AIDS.
Delirium is underdiagnosed and under-treated in medical settings,4,8 most likely because of its protean symptoms (Table 1)9 and fluctuating clinical findings. Neurologic abnormalities—including cortical and motor symptoms—also can occur.1
Mortality risk. Delirium is an independent risk factor for mortality.1,4,5 It is a marker for serious and potentially life-threatening medical problems, such as organ failure or sepsis. When antipsychotics fail to control delirium, the 3 most common reasons are:
- delirium’s etiology has not been discovered or addressed
- delirium’s etiology is resistant to treatment or potentially irreversible
- antipsychotic dosage was inadequate.
3 subtypes. Delirium is classified as hyperactive, hypoactive, or mixed, depending on arousal disturbance and psychomotor behavior:
- the hyperactive subtype includes hallucinations, delusions, agitation, and disorientation.
- the hypoactive subtype includes confusion, sedation, and decreased alertness but rarely hallucinations or delusions.1
Up to 1 in 4 patients (14% to 24%) have delirium at hospital admission, and the annual incidence of delirium is 6% to 56% among hospital populations.4 Elderly inpatients who develop delirium have an estimated mortality rate of 22% to 76% during that hospitalization.1 At the end of life, the prevalence of delirium may be as high as 85%.5
Serotonergic, noradrenergic, opiatergic, glutamatergic, and histaminergic neurotransmitter systems may contribute to delirium as a syndrome. Evidence implicates underactivity of the cholinergic system as the final common pathway.6,7
The acetylcholine-dopamine hypothesis explains the efficacy of dopamine antagonists in treating delirium by regulating the imbalance between cholinergic and dopaminergic activity.5,6 Cytokines—including interleukin-1, interleukin-2, and interleukin-6—and chronic hypercortisolism may also contribute to delirium.4
Antipsychotics: Limited evidence
- identifying and eliminating contributing factors
- instituting nonpharmacologic interventions based on environmental strategies (Table 2)4
- providing pharmacologic interventions—primarily antipsychotics—as needed.
- delirium severity improved with haloperidol, chlorpromazine, olanzapine, risperidone, or quetiapine
- comparison trials did not identify any antipsychotic as more efficacious than another.
Michaud et al11 reviewed guidelines, systematic reviews, randomized controlled trials, and cohort studies on delirium management. They concluded that the experts agree on 3 points:
- prevention should be emphasized
- atypical antipsychotics are not first-choice drugs because of data on adverse events in the elderly
- pharmacologic treatment is recommended when the patient’s condition prevents adequate care or puts the patient or staff at risk.
Conventional antipsychotics
Haloperidol, the most-studied antipsychotic in delirium treatment, often is the drug of choice because of its high potency, low sedative effect, few anticholinergic side effects, minimal cardiovascular side effects, no active metabolites, and multiple administration routes.1
An IV route can facilitate rapid onset of medication effects. Compared with oral haloperidol, IV administration is associated with a lower risk of extrapyramidal symptoms (EPS), which allows use of higher doses.
Any IV use of injectable haloperidol is off-label, however. If you choose the IV route, monitor patients carefully for cardiac arrhythmias. Haloperidol’s prescribing information carries a new warning of sudden death, QT prolongation, and torsades de pointes in patients given IV haloperidol.
Chlorpromazine. In a double-blind, randomized comparison trial of 30 hospitalized AIDS patients, our group12 found oral and IM haloperidol (n=11) or chlorpromazine (n=13) highly effective in controlling delirium. Delirium symptoms improved significantly in both hypoactive and hyperactive subtypes with low doses of either antipsychotic (approximately 2 mg of haloperidol equivalent/day).
No patients developed dystonic or dyskinetic symptoms. Lorazepam, given to 6 patients, worsened delirium and cognitive impairment.
Table 1
Recognizing delirium: Diagnostic clinical features*
| Altered level of alertness and arousal |
| Rapidly fluctuating course |
| Attention disturbance |
| Increased or decreased psychomotor activity |
| Disturbance of sleep-wake cycle |
| Affective symptoms |
| Altered perceptions |
| Disorganized thinking and incoherent speech |
| Disorientation and memory impairment |
| * Not all symptoms are present in every case. |
| Source: Reference 9 |
Nonpharmacologic approaches to managing delirium
| Search for and correct all causes of delirium, including underlying disease or a medication effect |
| Create a calm, comfortable environment |
| Provide orienting objects such as calendars and clocks |
| Have family members present |
| Limit room and staff changes |
| Allow patients uninterrupted rest at night to improve the sleep-wake cycle |
| Consider 1-to-1 nursing observation, as necessary |
| Source: Reference 4 |
Atypicals in delirium: Trial data
Risperidone. Three open-label studies of risperidone in patients with delirium reported minimal risk of sedation and EPS.13-15
A 7-day, double-blind, flexible-dose trial of 24 patients with delirium16 found no significant difference between haloperidol (mean 1.71 mg/d) and risperidone (mean 1.02 mg/d) in clinical efficacy or response rate. The authors acknowledged, that they were unable to obtain identical-looking haloperidol and risperidone tablets for the trial.
Kim et al17 studied dopamine transporter gene polymorphism and use of haloperidol vs risperidone in 42 patients with delirium. Relatively low doses of both antipsychotics showed similar efficacy, and the authors concluded that dopamine transporter gene polymorphism did not influence delirium treatment.
Olanzapine. In an open trial of 79 inpatients with advanced cancer, olanzapine (mean 6.3 mg/d, range 2.5 to 20 mg/d) resolved delirium in 76% of patients, with no incidence of EPS.18 Age >70, history of dementia, hypoxia, cerebral metastasis, and hypoactive delirium were associated with poor response to olanzapine. This study is unique in assessing olanzapine’s efficacy in different delirium subtypes.
A prospective, randomized trial compared olanzapine (mean 4.5 mg/d, range 2.5 to 13.5 mg/d) with haloperidol (mean 6.5 mg/d, range 1 to 28 mg/d) in patients admitted with delirium to a critical care setting.19 Both treatment groups showed similar improvement over 5 days. No side effects were reported in the patients receiving olanzapine.
Ziprasidone. In the first case report in which ziprasidone was used to treat delirium,21 an HIV/AIDS patient was given 100 mg/d. Delirium symptoms improved, but treatment was discontinued because of side effects (hypokalemia, hypomagnesemia, premature ventricular contractions, and QT interval prolongation).
Aripiprazole. Straker et al22 reported 14 cases delirium treated with aripiprazole, which showed few side effects. Twelve patients had a ≥50% decrease in Delirium Rating Scale scores, and 13 showed improvement in Clinical Global Impression scale scores.
Clinical options
When choosing an antipsychotic to treat delirium, consider the individual patient’s risks of EPS, sedation, anticholinergic side effects, cardiac arrhythmias, and drug-drug interactions.
Haloperidol. When medication is necessary for delirium, American Psychiatric Association (APA) guidelines consider low-dose haloperidol as first-line treatment (see Related Resources). Recommended dosage is 1 to 2 mg (0.25 to 0.5 mg for the elderly) every 4 hours as needed.
Adding oral or IV lorazepam (0.5 to 1 mg every 1 to 2 hours) to haloperidol may help rapidly sedate the agitated delirious patient and minimize the risk of EPS associated with haloperidol.1 Avoid benzodiazepine monotherapy unless delirium is related to alcohol or benzodiazepine withdrawal.
Chlorpromazine. We have successfully used oral or IV chlorpromazine (12.5 to 50 mg every 4 to 12 hours) instead of haloperidol plus lorazepam when increased sedation was required, especially:
- in the ICU, where close blood pressure monitoring was feasible
- for severe agitation in terminally ill patients to decrease distress for the patient, family and staff.
Atypical antipsychotics also may be used to treat delirium, as supported by the literature. Recommended dosing, available routes administration routes, and clinical comments are summarized in Table 3.23
Table 3
Recommended antipsychotic dosing for delirium*
| Antipsychotic | Dosage | Route† | Comment | |
|---|---|---|---|---|
| Typical agents | ||||
| Haloperidol | Initial: 0.5 to 1 mg Range: 0.5 to 2 mg every 2 to 12 hours | Oral, IV, SC, IM | ‘First choice’ for delirium when antipsychotic treatment is needed (per APA guidelines) | |
| Chlorpromazine | Initial: 12.5 to 25 mg Range: 12.5 to 50 mg every 4 to 12 hours | Oral, IV, IM | Alternative to haloperidol plus lorazepam when increased sedation is needed | |
| Atypical agents | ||||
| Risperidone | Initial: 0.25 to 1 mg Range: 0.25 to 2 mg/d | Oral | Risk of sedation and orthostatic hypotension at higher doses | |
| Olanzapine | Initial: 2.5 to 5 mg nightly Range: 2.5 to 10 mg/d | Oral | Sedation (a potential limiting factor) may be beneficial for hyperactive delirium | |
| Quetiapine | Initial: 25 to 50 mg Range: 25 to 200 mg/d, usually divided into 2 daily doses | Oral | Sedation and orthostatic hypotension are dose-limiting factors | |
| Ziprasidone | Initial: 20 mg bid Range: 20 to 160 mg/d, usually divided into 2 daily doses | Oral | Limited data in delirium because of concerns about QT interval prolongation in medically ill patients | |
| Aripiprazole | Initial: 10 to 15 mg Range: 10 to 30 mg/d | Oral | ‘Dopamine stabilizing’ effect might be preferable in hypoactive delirium | |
| * For frail elderly patients, start with approximately one-half the suggested initial dose. | ||||
| † Risperidone and aripiprazole are available in liquid formulations. Risperidone, olanzapine, and aripiprazole are available in orally disintegrating tablets. | ||||
| APA: American Psychiatric Association; IM: intramuscular; IV: intravenous; SC: subcutaneous | ||||
| Source: Reference 23 | ||||
Managing adverse effects
Reassess patients frequently during a delirium episode to adjust the antipsychotic dose, search for underlying causes, and monitor for side effects (Table 4). In frail elderly patients, start with approximately one-half the recommended initial dose to reduce the side effect risk.
Antipsychotics may not be appropriate in certain populations with delirium, particularly in patients with:
- dementia of Lewy body type or Parkinson’s disease dementia
- stroke
- history of adverse reactions to antipsychotics.
Although the FDA advisory did not apply to typical antipsychotics, Wang et al3—in a retrospective cohort of nearly 23,000 patients age >65—found statistically significant higher mortality rates with typical vs atypical antipsychotics. The increased mortality risk with the typical agents was seen whether or not patients had dementia. The greatest increases in risk occurred early in therapy and with relatively high dosages.
The mortality risk associated with short-term antipsychotic treatment in medically ill elderly patients is unknown. Untreated delirium may impose a greater risk of morbidity and mortality than the risk associated with antipsychotics, however. Until more evidence becomes available, we recommend that you try to use low antipsychotic doses, especially for the elderly.
EPS are more common with conventional antipsychotics but also can be associated with the atypicals—particularly with risperidone at doses higher than 4 to 6 mg/d. To minimize EPS risk, monitor delirium patients daily during antipsychotic treatment and identify populations at risk.
QT interval prolongation. A prolonged QT interval increases the risk of ventricular arrhythmias—such as torsades de pointes and ventricular fibrillation—that can lead to syncope, cardiac arrest, or sudden cardiac death. Among the atypicals, ziprasidone has been associated with the highest rates of QT interval prolongation, followed by quetiapine, risperidone, and olanzapine.24 Thioridazine carries the greatest risk among the typical agents.25
When using antipsychotics for delirium, identify patients at risk for QT interval changes and monitor all patients during treatment. Risk factors include older age, female sex, preexisting heart disease, bradycardia, electrolyte abnormalities, and use of drugs that block potassium. APA guidelines recommend discontinuing antipsychotic therapy if QTc exceeds 450 msec or increases >25% from baseline.1 Consult with a cardiologist when antipsychotic treatment is necessary despite QT prolongation.
Metabolic syndrome. Long-term use of atypical antipsychotics—particularly olanzapine—has been associated with metabolic dysregulation and increased risk of obesity and diabetes. In the absence of data on the atypicals’ short-term effects on metabolism, we recommend careful monitoring for metabolic syndrome when using these agents, especially in patients with preexisting metabolic disturbances.26
Table 4
Monitoring for antipsychotic side effects during delirium treatment
| Side effects | How to monitor |
|---|---|
| EPS (parkinsonism, akathisia, dystonia) | Neurologic examination |
| Neuroleptic malignant syndrome | Neurologic examination, serum creatinine phosphokinase, serum prolactin |
| QT interval prolongation, torsades de pointes | ECG, serum potassium and magnesium, family history of QT prolongation |
| Metabolic syndrome (hyperglycemia, hyperlipidemia, weight gain) | Fasting blood glucose, lipid profile, weight, hemoglobin A1c |
| Anticholinergic symptoms (dry mouth, constipation) | History and physical examination |
| EPS: extrapyramidal symptoms | |
Discontinuing antipsychotics
No evidence-based or expert consensus guidelines have addressed when or how to discontinue antipsychotic treatment of delirium. Several studies—including a randomized, controlled trial by our group12—used protocols that reflect expert clinician practice.
Antipsychotic therapy is initiated to control delirium’s symptoms and is presumed to be needed until the causes have been identified or have resolved. Thus, antipsychotics are typically given in 3 phases:
Maintenance. Continue the antipsychotic 7 to 10 days—typically at two-thirds to one-half the initial-phase dosage—to allow delirium causes to be identified and resolve.
Tapering/discontinuation. If delirium symptoms resolve, taper and discontinue the antipsychotic relatively slowly over 3 to 5 days to allow for rapid control should delirium symptoms reemerge. Re-emergence suggests that new or unrecognized causes of delirium are present or identified causes have not resolved.
- American Psychiatric Association. Treating delirium: a quick reference guide. www.psych.org/psych_pract/treatg/quick_ref_guide/DeliriumQRG_4-15-05.pdf.
- American Psychiatric Association. Guideline watch: practice guideline for the treatment of patients with delirium. www.psych.org/psych_pract/treatg/pg/Delirium.watch.pdf.
- American Psychosocial Oncology Society. Multidisciplinary training in psycho-oncology: delirium. www.apos-society.org/professionals/meetings-ed/webcasts/webcasts-multidisciplinary.aspx.
- Aripiprazole • Abilify
- Chlorpromazine • various
- Haloperidol • various
- Lorazepam • Ativan
- Olanzapine • Zyprexa
- Quetiapine • Seroquel
- Risperidone • Risperdal
- Ziprasidone • Geodon
Dr. Breitbart is a consultant to Cephalon and a speaker for Cephalon, Janssen Pharmaceutica, Purdue Pharma, Eli Lilly and Company, and Bristol-Myers Squibb.
Dr. Alici-Evcimen reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. American Psychiatric Association. Practice guidelines for the treatment of patients with delirium. Am J Psychiatry 1999;156:S1-S20.
2. Schneider LS, Dagerman KS, Insel P. Risk of death with atypical antipsychotic drug treatment for dementia: meta-analysis of randomized placebo-controlled trials. JAMA. 2005;294(15):1934-43.
3. Wang PS, Schneeweiss S, Avorn J, et al. Risk of death in elderly users of conventional vs. atypical antipsychotic medications. N Engl J Med 2005;353(22):2335-41.
4. Inouye SK. Delirium in older persons. N Engl J Med 2006;354(11):1157-65.
5. Casarett DJ, Inouye SK. Diagnosis and management of delirium near the end of life. Ann Intern Med 2001;135(1):32-40.
6. Trzepacz PT. Update on the neuropathogenesis of delirium. Dement Geriatr Cogn Disord 1999;10(5):330-4.
7. Trzepacz PT. Is there a final common neural pathway in delirium? Focus on acetylcholine and dopamine. Semin Clin Neuropsychiatry 2000;5(2):132-48.
8. Breitbart W, Gibson C, Tremblay A. The delirium experience: delirium recall and delirium-related distress in hospitalized patients with cancer, their spouses/caregivers, and their nurses. Psychosomatics 2002;43(3):183-94.
9. Diagnostic and statistical manual of mental disorders, 4th ed, text revision. Washington, DC: American Psychiatric Association; 2000.
10. Seitz DP, Gill SS, van Zyl LT. Antipsychotics in the treatment of delirium: a systematic review. J Clin Psychiatry 2007;68(1):11-21.
11. Michaud L, Bula C, Berney A, et al. Delirium: guidelines for general hospitals. J Psychosom Res 2007;62(3):371-83.
12. Breitbart W, Marotta R, Platt MM, et al. A double-blind trial of haloperidol, chlorpromazine, and lorazepam in the treatment of delirium in hospitalized AIDS patients. Am J Psychiatry 1996;153(2):231-7.
13. Horikawa N, Yamazaki T, Miyamoto K. Treatment for delirium with risperidone: results of a prospective open trial with 10 patients. Gen Hosp Psychiatry 2003;25:289-92.
14. Mittal D, Jimerson N, Neely E. Risperidone in the treatment of delirium: results from a prospective open-label trial. J Clin Psychiatry 2004;65:662-7.
15. Parellada E, Baeza I, de Pablo J. Risperidone in the treatment of patients with delirium. J Clin Psychiatry 2004;65:348-53.16.
16. Han C, Kim Y. A double-blind trial of risperidone and haloperidol for the treatment of delirium. Psychosomatics 2004;45(4):297-301.
17. Kim JY, Jung IK, Han C, et al. Antipsychotics and dopamine transporter gene polymorphisms in delirium patients. Psychiatry Clin Neurosci. 2005;59(2):183-8.
18. Breitbart W, Tremblay A, Gibson C. An open trial of olanzapine for the treatment of delirium in hospitalized cancer patients. Psychosomatics 2002;43(3):175-82.
19. Skrobik Y, Bergeron N, Dumont M. Olanzapine vs haloperidol: treating delirium in a critical care setting. Intensive Care Med 2004;30:444-9.
20. Pae C, Lee S, Lee C. A pilot trial of quetiapine for the treatment of patients with delirium. Hum Psychopharmacol 2004;19:125-7.
21. Leso L, Schwartz T. Ziprasidone treatment of delirium. Psychosomatics 2002;43:61-2.
22. Straker DA, Shapiro PA, Muskin PR. Aripiprazole in the treatment of delirium. Psychosomatics. 2006;47(5):385-91.
23. Boettger S, Breitbart W. Atypical antipsychotics in the management of delirium: a review of the empirical literature. Palliat Support Care 2005;3(3):227-37.
24. Al-Khatib SM, LaPointe NM, Kramer JM, Califf RM. What clinicians should know about the QT interval. JAMA 2003;289(16):2120-7.
25. Glassman AH, Bigger JT, Jr. Antipsychotic drugs: prolonged QTc interval, torsade de pointes, and sudden death. Am J Psychiatry. 2001;158(11):1774-82.
26. Nasrallah HA, Newcomer JW. Atypical antipsychotics and metabolic dysregulation: evaluating the risk/benefit equation and improving the standard of care. J Clin Psychopharmacol 2004;24(5 suppl 1):S7-S14.
Delirium is a medical emergency that needs to be identified and treated vigorously. Antipsychotics—including haloperidol and atypical agents—effectively manage a wide spectrum of delirium symptoms and are an essential component in the standard multimodal approach.1 Even so, antipsychotics are not FDA-approved for treating delirium, and evidence on their safety in medically ill patients is limited—particularly in the elderly, in whom delirium occurs most often.
The FDA has warned of increased risk of death when atypical antipsychotics are used to treat behavioral disturbances in elderly patients with dementia.2 Similarly, a retrospective study of elderly patients taking antipsychotics found higher mortality rates associated with typical antipsychotics than with atypicals.3
This article discusses the risks and benefits of using antipsychotics to manage delirium. Based on the literature and clinical experience, we offer recommendations on choosing among the available agents and avoiding side effects.
A challenging diagnosis
Delirium is a neuropsychiatric syndrome precipitated by an underlying medical condition or a medication effect on the brain. Its characteristic symptoms—abrupt onset of disturbed consciousness, attention, cognition, and perception—tend to fluctuate during the day. Delirium most often occurs in elderly patients (Box)1,4-7—particularly with dementia—but also occurs in younger patients with serious illnesses such as cancer or HIV-AIDS.
Delirium is underdiagnosed and under-treated in medical settings,4,8 most likely because of its protean symptoms (Table 1)9 and fluctuating clinical findings. Neurologic abnormalities—including cortical and motor symptoms—also can occur.1
Mortality risk. Delirium is an independent risk factor for mortality.1,4,5 It is a marker for serious and potentially life-threatening medical problems, such as organ failure or sepsis. When antipsychotics fail to control delirium, the 3 most common reasons are:
- delirium’s etiology has not been discovered or addressed
- delirium’s etiology is resistant to treatment or potentially irreversible
- antipsychotic dosage was inadequate.
3 subtypes. Delirium is classified as hyperactive, hypoactive, or mixed, depending on arousal disturbance and psychomotor behavior:
- the hyperactive subtype includes hallucinations, delusions, agitation, and disorientation.
- the hypoactive subtype includes confusion, sedation, and decreased alertness but rarely hallucinations or delusions.1
Up to 1 in 4 patients (14% to 24%) have delirium at hospital admission, and the annual incidence of delirium is 6% to 56% among hospital populations.4 Elderly inpatients who develop delirium have an estimated mortality rate of 22% to 76% during that hospitalization.1 At the end of life, the prevalence of delirium may be as high as 85%.5
Serotonergic, noradrenergic, opiatergic, glutamatergic, and histaminergic neurotransmitter systems may contribute to delirium as a syndrome. Evidence implicates underactivity of the cholinergic system as the final common pathway.6,7
The acetylcholine-dopamine hypothesis explains the efficacy of dopamine antagonists in treating delirium by regulating the imbalance between cholinergic and dopaminergic activity.5,6 Cytokines—including interleukin-1, interleukin-2, and interleukin-6—and chronic hypercortisolism may also contribute to delirium.4
Antipsychotics: Limited evidence
- identifying and eliminating contributing factors
- instituting nonpharmacologic interventions based on environmental strategies (Table 2)4
- providing pharmacologic interventions—primarily antipsychotics—as needed.
- delirium severity improved with haloperidol, chlorpromazine, olanzapine, risperidone, or quetiapine
- comparison trials did not identify any antipsychotic as more efficacious than another.
Michaud et al11 reviewed guidelines, systematic reviews, randomized controlled trials, and cohort studies on delirium management. They concluded that the experts agree on 3 points:
- prevention should be emphasized
- atypical antipsychotics are not first-choice drugs because of data on adverse events in the elderly
- pharmacologic treatment is recommended when the patient’s condition prevents adequate care or puts the patient or staff at risk.
Conventional antipsychotics
Haloperidol, the most-studied antipsychotic in delirium treatment, often is the drug of choice because of its high potency, low sedative effect, few anticholinergic side effects, minimal cardiovascular side effects, no active metabolites, and multiple administration routes.1
An IV route can facilitate rapid onset of medication effects. Compared with oral haloperidol, IV administration is associated with a lower risk of extrapyramidal symptoms (EPS), which allows use of higher doses.
Any IV use of injectable haloperidol is off-label, however. If you choose the IV route, monitor patients carefully for cardiac arrhythmias. Haloperidol’s prescribing information carries a new warning of sudden death, QT prolongation, and torsades de pointes in patients given IV haloperidol.
Chlorpromazine. In a double-blind, randomized comparison trial of 30 hospitalized AIDS patients, our group12 found oral and IM haloperidol (n=11) or chlorpromazine (n=13) highly effective in controlling delirium. Delirium symptoms improved significantly in both hypoactive and hyperactive subtypes with low doses of either antipsychotic (approximately 2 mg of haloperidol equivalent/day).
No patients developed dystonic or dyskinetic symptoms. Lorazepam, given to 6 patients, worsened delirium and cognitive impairment.
Table 1
Recognizing delirium: Diagnostic clinical features*
| Altered level of alertness and arousal |
| Rapidly fluctuating course |
| Attention disturbance |
| Increased or decreased psychomotor activity |
| Disturbance of sleep-wake cycle |
| Affective symptoms |
| Altered perceptions |
| Disorganized thinking and incoherent speech |
| Disorientation and memory impairment |
| * Not all symptoms are present in every case. |
| Source: Reference 9 |
Nonpharmacologic approaches to managing delirium
| Search for and correct all causes of delirium, including underlying disease or a medication effect |
| Create a calm, comfortable environment |
| Provide orienting objects such as calendars and clocks |
| Have family members present |
| Limit room and staff changes |
| Allow patients uninterrupted rest at night to improve the sleep-wake cycle |
| Consider 1-to-1 nursing observation, as necessary |
| Source: Reference 4 |
Atypicals in delirium: Trial data
Risperidone. Three open-label studies of risperidone in patients with delirium reported minimal risk of sedation and EPS.13-15
A 7-day, double-blind, flexible-dose trial of 24 patients with delirium16 found no significant difference between haloperidol (mean 1.71 mg/d) and risperidone (mean 1.02 mg/d) in clinical efficacy or response rate. The authors acknowledged, that they were unable to obtain identical-looking haloperidol and risperidone tablets for the trial.
Kim et al17 studied dopamine transporter gene polymorphism and use of haloperidol vs risperidone in 42 patients with delirium. Relatively low doses of both antipsychotics showed similar efficacy, and the authors concluded that dopamine transporter gene polymorphism did not influence delirium treatment.
Olanzapine. In an open trial of 79 inpatients with advanced cancer, olanzapine (mean 6.3 mg/d, range 2.5 to 20 mg/d) resolved delirium in 76% of patients, with no incidence of EPS.18 Age >70, history of dementia, hypoxia, cerebral metastasis, and hypoactive delirium were associated with poor response to olanzapine. This study is unique in assessing olanzapine’s efficacy in different delirium subtypes.
A prospective, randomized trial compared olanzapine (mean 4.5 mg/d, range 2.5 to 13.5 mg/d) with haloperidol (mean 6.5 mg/d, range 1 to 28 mg/d) in patients admitted with delirium to a critical care setting.19 Both treatment groups showed similar improvement over 5 days. No side effects were reported in the patients receiving olanzapine.
Ziprasidone. In the first case report in which ziprasidone was used to treat delirium,21 an HIV/AIDS patient was given 100 mg/d. Delirium symptoms improved, but treatment was discontinued because of side effects (hypokalemia, hypomagnesemia, premature ventricular contractions, and QT interval prolongation).
Aripiprazole. Straker et al22 reported 14 cases delirium treated with aripiprazole, which showed few side effects. Twelve patients had a ≥50% decrease in Delirium Rating Scale scores, and 13 showed improvement in Clinical Global Impression scale scores.
Clinical options
When choosing an antipsychotic to treat delirium, consider the individual patient’s risks of EPS, sedation, anticholinergic side effects, cardiac arrhythmias, and drug-drug interactions.
Haloperidol. When medication is necessary for delirium, American Psychiatric Association (APA) guidelines consider low-dose haloperidol as first-line treatment (see Related Resources). Recommended dosage is 1 to 2 mg (0.25 to 0.5 mg for the elderly) every 4 hours as needed.
Adding oral or IV lorazepam (0.5 to 1 mg every 1 to 2 hours) to haloperidol may help rapidly sedate the agitated delirious patient and minimize the risk of EPS associated with haloperidol.1 Avoid benzodiazepine monotherapy unless delirium is related to alcohol or benzodiazepine withdrawal.
Chlorpromazine. We have successfully used oral or IV chlorpromazine (12.5 to 50 mg every 4 to 12 hours) instead of haloperidol plus lorazepam when increased sedation was required, especially:
- in the ICU, where close blood pressure monitoring was feasible
- for severe agitation in terminally ill patients to decrease distress for the patient, family and staff.
Atypical antipsychotics also may be used to treat delirium, as supported by the literature. Recommended dosing, available routes administration routes, and clinical comments are summarized in Table 3.23
Table 3
Recommended antipsychotic dosing for delirium*
| Antipsychotic | Dosage | Route† | Comment | |
|---|---|---|---|---|
| Typical agents | ||||
| Haloperidol | Initial: 0.5 to 1 mg Range: 0.5 to 2 mg every 2 to 12 hours | Oral, IV, SC, IM | ‘First choice’ for delirium when antipsychotic treatment is needed (per APA guidelines) | |
| Chlorpromazine | Initial: 12.5 to 25 mg Range: 12.5 to 50 mg every 4 to 12 hours | Oral, IV, IM | Alternative to haloperidol plus lorazepam when increased sedation is needed | |
| Atypical agents | ||||
| Risperidone | Initial: 0.25 to 1 mg Range: 0.25 to 2 mg/d | Oral | Risk of sedation and orthostatic hypotension at higher doses | |
| Olanzapine | Initial: 2.5 to 5 mg nightly Range: 2.5 to 10 mg/d | Oral | Sedation (a potential limiting factor) may be beneficial for hyperactive delirium | |
| Quetiapine | Initial: 25 to 50 mg Range: 25 to 200 mg/d, usually divided into 2 daily doses | Oral | Sedation and orthostatic hypotension are dose-limiting factors | |
| Ziprasidone | Initial: 20 mg bid Range: 20 to 160 mg/d, usually divided into 2 daily doses | Oral | Limited data in delirium because of concerns about QT interval prolongation in medically ill patients | |
| Aripiprazole | Initial: 10 to 15 mg Range: 10 to 30 mg/d | Oral | ‘Dopamine stabilizing’ effect might be preferable in hypoactive delirium | |
| * For frail elderly patients, start with approximately one-half the suggested initial dose. | ||||
| † Risperidone and aripiprazole are available in liquid formulations. Risperidone, olanzapine, and aripiprazole are available in orally disintegrating tablets. | ||||
| APA: American Psychiatric Association; IM: intramuscular; IV: intravenous; SC: subcutaneous | ||||
| Source: Reference 23 | ||||
Managing adverse effects
Reassess patients frequently during a delirium episode to adjust the antipsychotic dose, search for underlying causes, and monitor for side effects (Table 4). In frail elderly patients, start with approximately one-half the recommended initial dose to reduce the side effect risk.
Antipsychotics may not be appropriate in certain populations with delirium, particularly in patients with:
- dementia of Lewy body type or Parkinson’s disease dementia
- stroke
- history of adverse reactions to antipsychotics.
Although the FDA advisory did not apply to typical antipsychotics, Wang et al3—in a retrospective cohort of nearly 23,000 patients age >65—found statistically significant higher mortality rates with typical vs atypical antipsychotics. The increased mortality risk with the typical agents was seen whether or not patients had dementia. The greatest increases in risk occurred early in therapy and with relatively high dosages.
The mortality risk associated with short-term antipsychotic treatment in medically ill elderly patients is unknown. Untreated delirium may impose a greater risk of morbidity and mortality than the risk associated with antipsychotics, however. Until more evidence becomes available, we recommend that you try to use low antipsychotic doses, especially for the elderly.
EPS are more common with conventional antipsychotics but also can be associated with the atypicals—particularly with risperidone at doses higher than 4 to 6 mg/d. To minimize EPS risk, monitor delirium patients daily during antipsychotic treatment and identify populations at risk.
QT interval prolongation. A prolonged QT interval increases the risk of ventricular arrhythmias—such as torsades de pointes and ventricular fibrillation—that can lead to syncope, cardiac arrest, or sudden cardiac death. Among the atypicals, ziprasidone has been associated with the highest rates of QT interval prolongation, followed by quetiapine, risperidone, and olanzapine.24 Thioridazine carries the greatest risk among the typical agents.25
When using antipsychotics for delirium, identify patients at risk for QT interval changes and monitor all patients during treatment. Risk factors include older age, female sex, preexisting heart disease, bradycardia, electrolyte abnormalities, and use of drugs that block potassium. APA guidelines recommend discontinuing antipsychotic therapy if QTc exceeds 450 msec or increases >25% from baseline.1 Consult with a cardiologist when antipsychotic treatment is necessary despite QT prolongation.
Metabolic syndrome. Long-term use of atypical antipsychotics—particularly olanzapine—has been associated with metabolic dysregulation and increased risk of obesity and diabetes. In the absence of data on the atypicals’ short-term effects on metabolism, we recommend careful monitoring for metabolic syndrome when using these agents, especially in patients with preexisting metabolic disturbances.26
Table 4
Monitoring for antipsychotic side effects during delirium treatment
| Side effects | How to monitor |
|---|---|
| EPS (parkinsonism, akathisia, dystonia) | Neurologic examination |
| Neuroleptic malignant syndrome | Neurologic examination, serum creatinine phosphokinase, serum prolactin |
| QT interval prolongation, torsades de pointes | ECG, serum potassium and magnesium, family history of QT prolongation |
| Metabolic syndrome (hyperglycemia, hyperlipidemia, weight gain) | Fasting blood glucose, lipid profile, weight, hemoglobin A1c |
| Anticholinergic symptoms (dry mouth, constipation) | History and physical examination |
| EPS: extrapyramidal symptoms | |
Discontinuing antipsychotics
No evidence-based or expert consensus guidelines have addressed when or how to discontinue antipsychotic treatment of delirium. Several studies—including a randomized, controlled trial by our group12—used protocols that reflect expert clinician practice.
Antipsychotic therapy is initiated to control delirium’s symptoms and is presumed to be needed until the causes have been identified or have resolved. Thus, antipsychotics are typically given in 3 phases:
Maintenance. Continue the antipsychotic 7 to 10 days—typically at two-thirds to one-half the initial-phase dosage—to allow delirium causes to be identified and resolve.
Tapering/discontinuation. If delirium symptoms resolve, taper and discontinue the antipsychotic relatively slowly over 3 to 5 days to allow for rapid control should delirium symptoms reemerge. Re-emergence suggests that new or unrecognized causes of delirium are present or identified causes have not resolved.
- American Psychiatric Association. Treating delirium: a quick reference guide. www.psych.org/psych_pract/treatg/quick_ref_guide/DeliriumQRG_4-15-05.pdf.
- American Psychiatric Association. Guideline watch: practice guideline for the treatment of patients with delirium. www.psych.org/psych_pract/treatg/pg/Delirium.watch.pdf.
- American Psychosocial Oncology Society. Multidisciplinary training in psycho-oncology: delirium. www.apos-society.org/professionals/meetings-ed/webcasts/webcasts-multidisciplinary.aspx.
- Aripiprazole • Abilify
- Chlorpromazine • various
- Haloperidol • various
- Lorazepam • Ativan
- Olanzapine • Zyprexa
- Quetiapine • Seroquel
- Risperidone • Risperdal
- Ziprasidone • Geodon
Dr. Breitbart is a consultant to Cephalon and a speaker for Cephalon, Janssen Pharmaceutica, Purdue Pharma, Eli Lilly and Company, and Bristol-Myers Squibb.
Dr. Alici-Evcimen reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
Delirium is a medical emergency that needs to be identified and treated vigorously. Antipsychotics—including haloperidol and atypical agents—effectively manage a wide spectrum of delirium symptoms and are an essential component in the standard multimodal approach.1 Even so, antipsychotics are not FDA-approved for treating delirium, and evidence on their safety in medically ill patients is limited—particularly in the elderly, in whom delirium occurs most often.
The FDA has warned of increased risk of death when atypical antipsychotics are used to treat behavioral disturbances in elderly patients with dementia.2 Similarly, a retrospective study of elderly patients taking antipsychotics found higher mortality rates associated with typical antipsychotics than with atypicals.3
This article discusses the risks and benefits of using antipsychotics to manage delirium. Based on the literature and clinical experience, we offer recommendations on choosing among the available agents and avoiding side effects.
A challenging diagnosis
Delirium is a neuropsychiatric syndrome precipitated by an underlying medical condition or a medication effect on the brain. Its characteristic symptoms—abrupt onset of disturbed consciousness, attention, cognition, and perception—tend to fluctuate during the day. Delirium most often occurs in elderly patients (Box)1,4-7—particularly with dementia—but also occurs in younger patients with serious illnesses such as cancer or HIV-AIDS.
Delirium is underdiagnosed and under-treated in medical settings,4,8 most likely because of its protean symptoms (Table 1)9 and fluctuating clinical findings. Neurologic abnormalities—including cortical and motor symptoms—also can occur.1
Mortality risk. Delirium is an independent risk factor for mortality.1,4,5 It is a marker for serious and potentially life-threatening medical problems, such as organ failure or sepsis. When antipsychotics fail to control delirium, the 3 most common reasons are:
- delirium’s etiology has not been discovered or addressed
- delirium’s etiology is resistant to treatment or potentially irreversible
- antipsychotic dosage was inadequate.
3 subtypes. Delirium is classified as hyperactive, hypoactive, or mixed, depending on arousal disturbance and psychomotor behavior:
- the hyperactive subtype includes hallucinations, delusions, agitation, and disorientation.
- the hypoactive subtype includes confusion, sedation, and decreased alertness but rarely hallucinations or delusions.1
Up to 1 in 4 patients (14% to 24%) have delirium at hospital admission, and the annual incidence of delirium is 6% to 56% among hospital populations.4 Elderly inpatients who develop delirium have an estimated mortality rate of 22% to 76% during that hospitalization.1 At the end of life, the prevalence of delirium may be as high as 85%.5
Serotonergic, noradrenergic, opiatergic, glutamatergic, and histaminergic neurotransmitter systems may contribute to delirium as a syndrome. Evidence implicates underactivity of the cholinergic system as the final common pathway.6,7
The acetylcholine-dopamine hypothesis explains the efficacy of dopamine antagonists in treating delirium by regulating the imbalance between cholinergic and dopaminergic activity.5,6 Cytokines—including interleukin-1, interleukin-2, and interleukin-6—and chronic hypercortisolism may also contribute to delirium.4
Antipsychotics: Limited evidence
- identifying and eliminating contributing factors
- instituting nonpharmacologic interventions based on environmental strategies (Table 2)4
- providing pharmacologic interventions—primarily antipsychotics—as needed.
- delirium severity improved with haloperidol, chlorpromazine, olanzapine, risperidone, or quetiapine
- comparison trials did not identify any antipsychotic as more efficacious than another.
Michaud et al11 reviewed guidelines, systematic reviews, randomized controlled trials, and cohort studies on delirium management. They concluded that the experts agree on 3 points:
- prevention should be emphasized
- atypical antipsychotics are not first-choice drugs because of data on adverse events in the elderly
- pharmacologic treatment is recommended when the patient’s condition prevents adequate care or puts the patient or staff at risk.
Conventional antipsychotics
Haloperidol, the most-studied antipsychotic in delirium treatment, often is the drug of choice because of its high potency, low sedative effect, few anticholinergic side effects, minimal cardiovascular side effects, no active metabolites, and multiple administration routes.1
An IV route can facilitate rapid onset of medication effects. Compared with oral haloperidol, IV administration is associated with a lower risk of extrapyramidal symptoms (EPS), which allows use of higher doses.
Any IV use of injectable haloperidol is off-label, however. If you choose the IV route, monitor patients carefully for cardiac arrhythmias. Haloperidol’s prescribing information carries a new warning of sudden death, QT prolongation, and torsades de pointes in patients given IV haloperidol.
Chlorpromazine. In a double-blind, randomized comparison trial of 30 hospitalized AIDS patients, our group12 found oral and IM haloperidol (n=11) or chlorpromazine (n=13) highly effective in controlling delirium. Delirium symptoms improved significantly in both hypoactive and hyperactive subtypes with low doses of either antipsychotic (approximately 2 mg of haloperidol equivalent/day).
No patients developed dystonic or dyskinetic symptoms. Lorazepam, given to 6 patients, worsened delirium and cognitive impairment.
Table 1
Recognizing delirium: Diagnostic clinical features*
| Altered level of alertness and arousal |
| Rapidly fluctuating course |
| Attention disturbance |
| Increased or decreased psychomotor activity |
| Disturbance of sleep-wake cycle |
| Affective symptoms |
| Altered perceptions |
| Disorganized thinking and incoherent speech |
| Disorientation and memory impairment |
| * Not all symptoms are present in every case. |
| Source: Reference 9 |
Nonpharmacologic approaches to managing delirium
| Search for and correct all causes of delirium, including underlying disease or a medication effect |
| Create a calm, comfortable environment |
| Provide orienting objects such as calendars and clocks |
| Have family members present |
| Limit room and staff changes |
| Allow patients uninterrupted rest at night to improve the sleep-wake cycle |
| Consider 1-to-1 nursing observation, as necessary |
| Source: Reference 4 |
Atypicals in delirium: Trial data
Risperidone. Three open-label studies of risperidone in patients with delirium reported minimal risk of sedation and EPS.13-15
A 7-day, double-blind, flexible-dose trial of 24 patients with delirium16 found no significant difference between haloperidol (mean 1.71 mg/d) and risperidone (mean 1.02 mg/d) in clinical efficacy or response rate. The authors acknowledged, that they were unable to obtain identical-looking haloperidol and risperidone tablets for the trial.
Kim et al17 studied dopamine transporter gene polymorphism and use of haloperidol vs risperidone in 42 patients with delirium. Relatively low doses of both antipsychotics showed similar efficacy, and the authors concluded that dopamine transporter gene polymorphism did not influence delirium treatment.
Olanzapine. In an open trial of 79 inpatients with advanced cancer, olanzapine (mean 6.3 mg/d, range 2.5 to 20 mg/d) resolved delirium in 76% of patients, with no incidence of EPS.18 Age >70, history of dementia, hypoxia, cerebral metastasis, and hypoactive delirium were associated with poor response to olanzapine. This study is unique in assessing olanzapine’s efficacy in different delirium subtypes.
A prospective, randomized trial compared olanzapine (mean 4.5 mg/d, range 2.5 to 13.5 mg/d) with haloperidol (mean 6.5 mg/d, range 1 to 28 mg/d) in patients admitted with delirium to a critical care setting.19 Both treatment groups showed similar improvement over 5 days. No side effects were reported in the patients receiving olanzapine.
Ziprasidone. In the first case report in which ziprasidone was used to treat delirium,21 an HIV/AIDS patient was given 100 mg/d. Delirium symptoms improved, but treatment was discontinued because of side effects (hypokalemia, hypomagnesemia, premature ventricular contractions, and QT interval prolongation).
Aripiprazole. Straker et al22 reported 14 cases delirium treated with aripiprazole, which showed few side effects. Twelve patients had a ≥50% decrease in Delirium Rating Scale scores, and 13 showed improvement in Clinical Global Impression scale scores.
Clinical options
When choosing an antipsychotic to treat delirium, consider the individual patient’s risks of EPS, sedation, anticholinergic side effects, cardiac arrhythmias, and drug-drug interactions.
Haloperidol. When medication is necessary for delirium, American Psychiatric Association (APA) guidelines consider low-dose haloperidol as first-line treatment (see Related Resources). Recommended dosage is 1 to 2 mg (0.25 to 0.5 mg for the elderly) every 4 hours as needed.
Adding oral or IV lorazepam (0.5 to 1 mg every 1 to 2 hours) to haloperidol may help rapidly sedate the agitated delirious patient and minimize the risk of EPS associated with haloperidol.1 Avoid benzodiazepine monotherapy unless delirium is related to alcohol or benzodiazepine withdrawal.
Chlorpromazine. We have successfully used oral or IV chlorpromazine (12.5 to 50 mg every 4 to 12 hours) instead of haloperidol plus lorazepam when increased sedation was required, especially:
- in the ICU, where close blood pressure monitoring was feasible
- for severe agitation in terminally ill patients to decrease distress for the patient, family and staff.
Atypical antipsychotics also may be used to treat delirium, as supported by the literature. Recommended dosing, available routes administration routes, and clinical comments are summarized in Table 3.23
Table 3
Recommended antipsychotic dosing for delirium*
| Antipsychotic | Dosage | Route† | Comment | |
|---|---|---|---|---|
| Typical agents | ||||
| Haloperidol | Initial: 0.5 to 1 mg Range: 0.5 to 2 mg every 2 to 12 hours | Oral, IV, SC, IM | ‘First choice’ for delirium when antipsychotic treatment is needed (per APA guidelines) | |
| Chlorpromazine | Initial: 12.5 to 25 mg Range: 12.5 to 50 mg every 4 to 12 hours | Oral, IV, IM | Alternative to haloperidol plus lorazepam when increased sedation is needed | |
| Atypical agents | ||||
| Risperidone | Initial: 0.25 to 1 mg Range: 0.25 to 2 mg/d | Oral | Risk of sedation and orthostatic hypotension at higher doses | |
| Olanzapine | Initial: 2.5 to 5 mg nightly Range: 2.5 to 10 mg/d | Oral | Sedation (a potential limiting factor) may be beneficial for hyperactive delirium | |
| Quetiapine | Initial: 25 to 50 mg Range: 25 to 200 mg/d, usually divided into 2 daily doses | Oral | Sedation and orthostatic hypotension are dose-limiting factors | |
| Ziprasidone | Initial: 20 mg bid Range: 20 to 160 mg/d, usually divided into 2 daily doses | Oral | Limited data in delirium because of concerns about QT interval prolongation in medically ill patients | |
| Aripiprazole | Initial: 10 to 15 mg Range: 10 to 30 mg/d | Oral | ‘Dopamine stabilizing’ effect might be preferable in hypoactive delirium | |
| * For frail elderly patients, start with approximately one-half the suggested initial dose. | ||||
| † Risperidone and aripiprazole are available in liquid formulations. Risperidone, olanzapine, and aripiprazole are available in orally disintegrating tablets. | ||||
| APA: American Psychiatric Association; IM: intramuscular; IV: intravenous; SC: subcutaneous | ||||
| Source: Reference 23 | ||||
Managing adverse effects
Reassess patients frequently during a delirium episode to adjust the antipsychotic dose, search for underlying causes, and monitor for side effects (Table 4). In frail elderly patients, start with approximately one-half the recommended initial dose to reduce the side effect risk.
Antipsychotics may not be appropriate in certain populations with delirium, particularly in patients with:
- dementia of Lewy body type or Parkinson’s disease dementia
- stroke
- history of adverse reactions to antipsychotics.
Although the FDA advisory did not apply to typical antipsychotics, Wang et al3—in a retrospective cohort of nearly 23,000 patients age >65—found statistically significant higher mortality rates with typical vs atypical antipsychotics. The increased mortality risk with the typical agents was seen whether or not patients had dementia. The greatest increases in risk occurred early in therapy and with relatively high dosages.
The mortality risk associated with short-term antipsychotic treatment in medically ill elderly patients is unknown. Untreated delirium may impose a greater risk of morbidity and mortality than the risk associated with antipsychotics, however. Until more evidence becomes available, we recommend that you try to use low antipsychotic doses, especially for the elderly.
EPS are more common with conventional antipsychotics but also can be associated with the atypicals—particularly with risperidone at doses higher than 4 to 6 mg/d. To minimize EPS risk, monitor delirium patients daily during antipsychotic treatment and identify populations at risk.
QT interval prolongation. A prolonged QT interval increases the risk of ventricular arrhythmias—such as torsades de pointes and ventricular fibrillation—that can lead to syncope, cardiac arrest, or sudden cardiac death. Among the atypicals, ziprasidone has been associated with the highest rates of QT interval prolongation, followed by quetiapine, risperidone, and olanzapine.24 Thioridazine carries the greatest risk among the typical agents.25
When using antipsychotics for delirium, identify patients at risk for QT interval changes and monitor all patients during treatment. Risk factors include older age, female sex, preexisting heart disease, bradycardia, electrolyte abnormalities, and use of drugs that block potassium. APA guidelines recommend discontinuing antipsychotic therapy if QTc exceeds 450 msec or increases >25% from baseline.1 Consult with a cardiologist when antipsychotic treatment is necessary despite QT prolongation.
Metabolic syndrome. Long-term use of atypical antipsychotics—particularly olanzapine—has been associated with metabolic dysregulation and increased risk of obesity and diabetes. In the absence of data on the atypicals’ short-term effects on metabolism, we recommend careful monitoring for metabolic syndrome when using these agents, especially in patients with preexisting metabolic disturbances.26
Table 4
Monitoring for antipsychotic side effects during delirium treatment
| Side effects | How to monitor |
|---|---|
| EPS (parkinsonism, akathisia, dystonia) | Neurologic examination |
| Neuroleptic malignant syndrome | Neurologic examination, serum creatinine phosphokinase, serum prolactin |
| QT interval prolongation, torsades de pointes | ECG, serum potassium and magnesium, family history of QT prolongation |
| Metabolic syndrome (hyperglycemia, hyperlipidemia, weight gain) | Fasting blood glucose, lipid profile, weight, hemoglobin A1c |
| Anticholinergic symptoms (dry mouth, constipation) | History and physical examination |
| EPS: extrapyramidal symptoms | |
Discontinuing antipsychotics
No evidence-based or expert consensus guidelines have addressed when or how to discontinue antipsychotic treatment of delirium. Several studies—including a randomized, controlled trial by our group12—used protocols that reflect expert clinician practice.
Antipsychotic therapy is initiated to control delirium’s symptoms and is presumed to be needed until the causes have been identified or have resolved. Thus, antipsychotics are typically given in 3 phases:
Maintenance. Continue the antipsychotic 7 to 10 days—typically at two-thirds to one-half the initial-phase dosage—to allow delirium causes to be identified and resolve.
Tapering/discontinuation. If delirium symptoms resolve, taper and discontinue the antipsychotic relatively slowly over 3 to 5 days to allow for rapid control should delirium symptoms reemerge. Re-emergence suggests that new or unrecognized causes of delirium are present or identified causes have not resolved.
- American Psychiatric Association. Treating delirium: a quick reference guide. www.psych.org/psych_pract/treatg/quick_ref_guide/DeliriumQRG_4-15-05.pdf.
- American Psychiatric Association. Guideline watch: practice guideline for the treatment of patients with delirium. www.psych.org/psych_pract/treatg/pg/Delirium.watch.pdf.
- American Psychosocial Oncology Society. Multidisciplinary training in psycho-oncology: delirium. www.apos-society.org/professionals/meetings-ed/webcasts/webcasts-multidisciplinary.aspx.
- Aripiprazole • Abilify
- Chlorpromazine • various
- Haloperidol • various
- Lorazepam • Ativan
- Olanzapine • Zyprexa
- Quetiapine • Seroquel
- Risperidone • Risperdal
- Ziprasidone • Geodon
Dr. Breitbart is a consultant to Cephalon and a speaker for Cephalon, Janssen Pharmaceutica, Purdue Pharma, Eli Lilly and Company, and Bristol-Myers Squibb.
Dr. Alici-Evcimen reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. American Psychiatric Association. Practice guidelines for the treatment of patients with delirium. Am J Psychiatry 1999;156:S1-S20.
2. Schneider LS, Dagerman KS, Insel P. Risk of death with atypical antipsychotic drug treatment for dementia: meta-analysis of randomized placebo-controlled trials. JAMA. 2005;294(15):1934-43.
3. Wang PS, Schneeweiss S, Avorn J, et al. Risk of death in elderly users of conventional vs. atypical antipsychotic medications. N Engl J Med 2005;353(22):2335-41.
4. Inouye SK. Delirium in older persons. N Engl J Med 2006;354(11):1157-65.
5. Casarett DJ, Inouye SK. Diagnosis and management of delirium near the end of life. Ann Intern Med 2001;135(1):32-40.
6. Trzepacz PT. Update on the neuropathogenesis of delirium. Dement Geriatr Cogn Disord 1999;10(5):330-4.
7. Trzepacz PT. Is there a final common neural pathway in delirium? Focus on acetylcholine and dopamine. Semin Clin Neuropsychiatry 2000;5(2):132-48.
8. Breitbart W, Gibson C, Tremblay A. The delirium experience: delirium recall and delirium-related distress in hospitalized patients with cancer, their spouses/caregivers, and their nurses. Psychosomatics 2002;43(3):183-94.
9. Diagnostic and statistical manual of mental disorders, 4th ed, text revision. Washington, DC: American Psychiatric Association; 2000.
10. Seitz DP, Gill SS, van Zyl LT. Antipsychotics in the treatment of delirium: a systematic review. J Clin Psychiatry 2007;68(1):11-21.
11. Michaud L, Bula C, Berney A, et al. Delirium: guidelines for general hospitals. J Psychosom Res 2007;62(3):371-83.
12. Breitbart W, Marotta R, Platt MM, et al. A double-blind trial of haloperidol, chlorpromazine, and lorazepam in the treatment of delirium in hospitalized AIDS patients. Am J Psychiatry 1996;153(2):231-7.
13. Horikawa N, Yamazaki T, Miyamoto K. Treatment for delirium with risperidone: results of a prospective open trial with 10 patients. Gen Hosp Psychiatry 2003;25:289-92.
14. Mittal D, Jimerson N, Neely E. Risperidone in the treatment of delirium: results from a prospective open-label trial. J Clin Psychiatry 2004;65:662-7.
15. Parellada E, Baeza I, de Pablo J. Risperidone in the treatment of patients with delirium. J Clin Psychiatry 2004;65:348-53.16.
16. Han C, Kim Y. A double-blind trial of risperidone and haloperidol for the treatment of delirium. Psychosomatics 2004;45(4):297-301.
17. Kim JY, Jung IK, Han C, et al. Antipsychotics and dopamine transporter gene polymorphisms in delirium patients. Psychiatry Clin Neurosci. 2005;59(2):183-8.
18. Breitbart W, Tremblay A, Gibson C. An open trial of olanzapine for the treatment of delirium in hospitalized cancer patients. Psychosomatics 2002;43(3):175-82.
19. Skrobik Y, Bergeron N, Dumont M. Olanzapine vs haloperidol: treating delirium in a critical care setting. Intensive Care Med 2004;30:444-9.
20. Pae C, Lee S, Lee C. A pilot trial of quetiapine for the treatment of patients with delirium. Hum Psychopharmacol 2004;19:125-7.
21. Leso L, Schwartz T. Ziprasidone treatment of delirium. Psychosomatics 2002;43:61-2.
22. Straker DA, Shapiro PA, Muskin PR. Aripiprazole in the treatment of delirium. Psychosomatics. 2006;47(5):385-91.
23. Boettger S, Breitbart W. Atypical antipsychotics in the management of delirium: a review of the empirical literature. Palliat Support Care 2005;3(3):227-37.
24. Al-Khatib SM, LaPointe NM, Kramer JM, Califf RM. What clinicians should know about the QT interval. JAMA 2003;289(16):2120-7.
25. Glassman AH, Bigger JT, Jr. Antipsychotic drugs: prolonged QTc interval, torsade de pointes, and sudden death. Am J Psychiatry. 2001;158(11):1774-82.
26. Nasrallah HA, Newcomer JW. Atypical antipsychotics and metabolic dysregulation: evaluating the risk/benefit equation and improving the standard of care. J Clin Psychopharmacol 2004;24(5 suppl 1):S7-S14.
1. American Psychiatric Association. Practice guidelines for the treatment of patients with delirium. Am J Psychiatry 1999;156:S1-S20.
2. Schneider LS, Dagerman KS, Insel P. Risk of death with atypical antipsychotic drug treatment for dementia: meta-analysis of randomized placebo-controlled trials. JAMA. 2005;294(15):1934-43.
3. Wang PS, Schneeweiss S, Avorn J, et al. Risk of death in elderly users of conventional vs. atypical antipsychotic medications. N Engl J Med 2005;353(22):2335-41.
4. Inouye SK. Delirium in older persons. N Engl J Med 2006;354(11):1157-65.
5. Casarett DJ, Inouye SK. Diagnosis and management of delirium near the end of life. Ann Intern Med 2001;135(1):32-40.
6. Trzepacz PT. Update on the neuropathogenesis of delirium. Dement Geriatr Cogn Disord 1999;10(5):330-4.
7. Trzepacz PT. Is there a final common neural pathway in delirium? Focus on acetylcholine and dopamine. Semin Clin Neuropsychiatry 2000;5(2):132-48.
8. Breitbart W, Gibson C, Tremblay A. The delirium experience: delirium recall and delirium-related distress in hospitalized patients with cancer, their spouses/caregivers, and their nurses. Psychosomatics 2002;43(3):183-94.
9. Diagnostic and statistical manual of mental disorders, 4th ed, text revision. Washington, DC: American Psychiatric Association; 2000.
10. Seitz DP, Gill SS, van Zyl LT. Antipsychotics in the treatment of delirium: a systematic review. J Clin Psychiatry 2007;68(1):11-21.
11. Michaud L, Bula C, Berney A, et al. Delirium: guidelines for general hospitals. J Psychosom Res 2007;62(3):371-83.
12. Breitbart W, Marotta R, Platt MM, et al. A double-blind trial of haloperidol, chlorpromazine, and lorazepam in the treatment of delirium in hospitalized AIDS patients. Am J Psychiatry 1996;153(2):231-7.
13. Horikawa N, Yamazaki T, Miyamoto K. Treatment for delirium with risperidone: results of a prospective open trial with 10 patients. Gen Hosp Psychiatry 2003;25:289-92.
14. Mittal D, Jimerson N, Neely E. Risperidone in the treatment of delirium: results from a prospective open-label trial. J Clin Psychiatry 2004;65:662-7.
15. Parellada E, Baeza I, de Pablo J. Risperidone in the treatment of patients with delirium. J Clin Psychiatry 2004;65:348-53.16.
16. Han C, Kim Y. A double-blind trial of risperidone and haloperidol for the treatment of delirium. Psychosomatics 2004;45(4):297-301.
17. Kim JY, Jung IK, Han C, et al. Antipsychotics and dopamine transporter gene polymorphisms in delirium patients. Psychiatry Clin Neurosci. 2005;59(2):183-8.
18. Breitbart W, Tremblay A, Gibson C. An open trial of olanzapine for the treatment of delirium in hospitalized cancer patients. Psychosomatics 2002;43(3):175-82.
19. Skrobik Y, Bergeron N, Dumont M. Olanzapine vs haloperidol: treating delirium in a critical care setting. Intensive Care Med 2004;30:444-9.
20. Pae C, Lee S, Lee C. A pilot trial of quetiapine for the treatment of patients with delirium. Hum Psychopharmacol 2004;19:125-7.
21. Leso L, Schwartz T. Ziprasidone treatment of delirium. Psychosomatics 2002;43:61-2.
22. Straker DA, Shapiro PA, Muskin PR. Aripiprazole in the treatment of delirium. Psychosomatics. 2006;47(5):385-91.
23. Boettger S, Breitbart W. Atypical antipsychotics in the management of delirium: a review of the empirical literature. Palliat Support Care 2005;3(3):227-37.
24. Al-Khatib SM, LaPointe NM, Kramer JM, Califf RM. What clinicians should know about the QT interval. JAMA 2003;289(16):2120-7.
25. Glassman AH, Bigger JT, Jr. Antipsychotic drugs: prolonged QTc interval, torsade de pointes, and sudden death. Am J Psychiatry. 2001;158(11):1774-82.
26. Nasrallah HA, Newcomer JW. Atypical antipsychotics and metabolic dysregulation: evaluating the risk/benefit equation and improving the standard of care. J Clin Psychopharmacol 2004;24(5 suppl 1):S7-S14.
Antidepressants: The spectrum beyond depression
A molecule is a molecule is a molecule—until it becomes identified with a purpose. Consider, for example, (-)-trans-4R-(4’-fluorophenyl)-3S-[(3’,4’-methylenedioxyphenoxy) methyl] piperidine. You probably know this molecule as paroxetine—an antidepressant, of course, but it is more than that. If you examine paroxetine’s FDA-approved indications, it also has anti-panic, anti-social anxiety, anti-obsessive-compulsive disorder, anti-posttraumatic stress disorder, and anti-premenstrual dysphoric disorder effects.
“Antidepressants” have achieved fame as antidepressants; one could say these molecules’ search for meaning has been fulfilled. Yet even within psychiatry, their many other uses (Table) can create semantic misunderstandings. Beyond psychiatry, consider the nondepressed patient with neurocardiogenic syncope who wonders why he’s being treated with an antidepressant.
Rather than calling antidepressants “panaceas,” the better choice is to educate patients about the drugs’ wide spectrum of activity. Let’s look broadly across the so-called antidepressants and examine their varied uses in psychiatry and other medical specialties.
Table
FDA-approved psychiatric indications for serotonin uptake inhibitors*
| SSRIs | ||||||||
| Citalopram | X | |||||||
| Escitalopram | X | X | ||||||
| Fluoxetine | X | X | X | X | X | |||
| Fluvoxamine | X | |||||||
| Paroxetine | X | X | X | X | X | X | X | |
| Sertraline | X | X | X | X | X | X | ||
| SNRIs | ||||||||
| Duloxetine | X | X | ||||||
| Venlafaxine | X | X | X | X | ||||
| SSRIs: selective serotonin reuptake inhibitors; SNRIs: serotonin-norepinephrine reuptake inhibitors; MDD: major depressive disorder; PD: panic disorder; SAD: social anxiety disorder; PTSD: posttraumatic stress disorder; GAD: generalized anxiety disorder; OCD: obsessive-compulsive disorder; PMDD: premenstrual dysphoric disorder; BUL: bulimia | ||||||||
| * The absence of an X does not necessarily imply that a drug is ineffective for a given indication but, more likely, that definitive studies are lacking. | ||||||||
Pain syndromes
Peripheral neuropathy. The only antidepressant with an FDA-approved pain indication is duloxetine, a serotonin-norepinephrine reuptake inhibitor (SNRI). Its approval for diabetic peripheral neuropathic pain (DPNP) was based on two 12-week, randomized, double-blind, placebo-controlled studies using fixed doses of 60 mg once or twice daily.1,2 Another SNRI—venlafaxine XR, 150 to 225 mg/d, but not 75 mg/d—also was found to be more effective than placebo for this indication in a 6-week, double-blind study (Box 1).3
Using antidepressants to treat pain syndromes is neither new nor restricted to SNRIs, however. In combined double-blind, cross-over studies of patients with DPNP, Max et al4 found:
- moderate or greater pain relief in 74% and 61% of subjects, respectively, from the tricyclics amitriptyline, mean 105 mg/d, and desipramine, mean 111 mg/d—with pain reduced by equal amounts in depressed and nondepressed patients
- no statistically significant difference in pain relief between the selective serotonin reuptake inhibitor (SSRI) fluoxetine, 40 mg/d, and placebo.
- tricyclics: 1 in every 2 to 3 patients
- SNRIs: 1 in every 4 to 5 patients
- SSRIs: 1 in every 7 patients.
Chronic headache. A meta-analysis7 of randomized, placebo-controlled studies found antidepressants more effective than placebo for chronic migraine and tension headache prophylaxis. Although a subgroup meta-analysis found similar effects for tricyclics and SSRIs, the authors characterized the tricyclics’ results as well established and the SSRIs’ as “less certain.”
The results of this meta-analysis might not accurately reflect bona fide antidepressants, however. Some of the 38 studies (25 of migraine, 12 of tension headache, 1 of both) included treatment with serotonin antagonists—most commonly pizotifen, which is not available in the United States and does not appear to be an antidepressant.
Back pain. Patients with chronic low back pain (average 10 years) seem to benefit from antidepressants, according to a meta-analysis of 9 randomized, controlled trials by Salerno et al.8 The effect on pain in the total 504 patients was “small but significant,” and improvement in function was “small but nonsignificant.” Individual sample sizes also were small, however, and only 2 studies excluded depressed patients.
Fibromyalgia, with chronic generalized musculoskeletal pain and tenderness, has been a focus of antidepressant drug therapy. Goldenberg et al9 concluded from an ambitious literature review (505 articles) that evidence of efficacy was strong for amitripty-line and modest for SSRIs and SNRIs.
Overall, antidepressants are generally understood to have analgesic effects in the absence of depression. Benefits for patients with pain syndromes are well established for tricyclics (especially amitriptyline) and recently with SNRIs, whereas SSRIs are less effective.
Serotonin and norepinephrine are involved in pain modulation via descending inhibitory pathways in the brain and spinal cord. Serotonin-norepinephrine reuptake inhibitors (SNRIs) have been shown to reduce the severity of diabetic peripheral neuropathic pain (DPNP) in randomized controlled trials.
Duloxetine. In 2 double-blind studies,1,2 nondepressed patients with DPNP received duloxetine, 60 mg once daily; duloxetine, 60 mg bid; or placebo for 12 weeks. They rated the severity of neuropathic pain every 24 hours on an 11-point Likert scale, and weekly mean scores were the primary outcome measure. Average pain scores improved more in both duloxetine groups vs placebo. Duloxetine treatment did not interfere with diabetic control, and both dosages were well tolerated.
The FDA approved an added indication for duloxetine in the management of DPNP.
Venlafaxine. In a double-blind study,3 244 adult outpatients with moderately severe DPNP received venlafaxine ER, 75 or 150 to 225 mg/d, or placebo for 6 weeks. Daily scores on the Visual Analog Pain Intensity (VAS-PI) and Pain Relief (VAS-PR) scales were primary efficacy measures.
Patients receiving the higher venlafaxine dosage—but not 75 mg/d—showed statistically significant less-intensive pain vs placebo. VAS-PI scores were 27% lower than at enrollment with placebo, 32% lower with venlafaxine, 75 mg/d, and 50% lower with venlafaxine, 150 to 225 mg/d (P
Nausea and somnolence were the most common side effects; clinically important ECG changes occurred in 7 patients treated with venlafaxine, 150 to 225 mg/d.
Smoking cessation
Bupropion SR is FDA-approved to aid smoking cessation, and this effect is independent of the drug’s antidepressant activity. Bupropion may act as a nicotine receptor antagonist as well as a norepinephrine dopamine reuptake inhibitor.
Other antidepressants have been studied for smoking cessation, with nortriptyline showing benefit in 2 large placebo-controlled trials. Studies with doxepin, fluoxetine, and moclobemide found little or no benefit for this indication.
Cardiovascular uses
Angina. Monoamine oxidase inhibitor (MAOI) antidepressants were used to treat angina pectoris in the late 1950s and early 1960s. This practice stopped after evidence showed that whereas angina pain may have improved with MAOIs, stress-induced ischemia on ECG did not.
Antiarrhythmia. Tricyclics had a brief fling in cardiovascular therapeutics when their quinidine-like class I antiarrhythmic activity was recognized. Imipramine was one of several drugs included in the Cardiac Arrhythmia Pilot Study in the 1980s that involved 502 postmyocardial infarction patients with ventricular arrhythmias. Imipramine was the least effective of the 4 drugs studied and the least well tolerated.13
variety of medications. Options include the vasopressor midodrine, fludrocortisone, beta blockers, and SSRIs— none
FDA-approved for this indication. Paroxetine, 20 mg/d, was considerably more effective than placebo in preventing
recurrent syncope in 68 patients who had been unresponsive
to or intolerant of traditional medications. During a mean 25 months of treatment, 82% of patients remained syncope-free on paroxetine vs 47% on placebo.14
Selective serotonin reuptake inhibitors (SSRIs) often are used to treat irritable bowel syndrome (IBS), though evidence of their effectiveness is scarce. SSRIs can improve IBS patients’ quality of life, but effects on abdominal pain and bloating are less clear.
Paroxetine. In a randomized, double-blind trial,16 gastroenterologists tested a highfiber diet plus paroxetine in nondepressed patients with IBS. Ninety-eight patients ages 18 to 65 who experienced IBS symptoms on low- or average-fiber diets were first put on high-fiber diets and assessed for well-being and abdominal pain and bloating. Of these, 81 symptomatic patients continued highfiber diets with added paroxetine, 10 to 40 mg/d (n=38) or placebo (n=43).
With paroxetine, patients’ overall well-being improved more than with placebo, but abdominal pain and bloating and social functioning did not.
Fluoxetine. In a double-blind, randomized trial,17 44 patients with pain and constipation-predominant IBS received fluoxetine, 20 mg/d, or placebo for 12 weeks. These patients met Rome II criteria for IBS—abdominal discomfort/pain for ≥12 weeks in past year that met 2 of 3 criteria:
- relieved by defecation
- onset associated with change in stool frequency
- onset associated with change in stool appearance.
Patients receiving fluoxetine had less abdominal discomfort, less bloating, more frequent bowel movements, and decreased consistency of stool vs placebo 4 weeks after treatment stopped. Mean number of symptoms per patient decreased from 4.6 to 0.7 in the fluoxetine group vs 4.5 to 2.9 in controls (P
Citalopram. IBS symptom severity was the primary outcome in a crossover trial comparing citalopram (20 mg for 3 weeks and 40 mg for 3 weeks) with placebo in 23 nondepressed patients.18 Abdominal pain and bloating, impact of symptoms on daily life, and overall well-being improved significantly more with citalopram than with placebo after 3 and 6 weeks.
Symptom improvements were not related to changes in depression, anxiety, or colonic sensorimotor function.
Gastrointestinal
Peptic ulcer disease was shown in the 1980s to respond to tricyclic antidepressants. At the time, both anticholinergic and antihistaminic effects were thought to be responsible, but the later observation that trimipramine inhibited Campylobacter pylori in vitro suggested an additional explanation. Today, tricyclics are only of historic interest as treatments for peptic ulcer.
Irritable bowel syndrome (IBS) patients have responded favorably to antidepressants, although it is often difficult to know if the benefit is independent of improved coexisting anxiety or depression. A meta-analysis of 12 randomized, placebo-controlled trials—mostly with tricyclics—found an odds ratio for improvement of 4.2 and a number needed to treat of 3.2.15
More recently, a few placebo-controlled studies have shown SSRIs to be beneficial for IBS,16-18 although not all symptoms improved and some IBS subtypes might be more responsive than others (Box 2). In an editorial, Talley19 concluded that antidepressant therapy of IBS was “at best only a ‘band-aid’ approach to management.”
Genitourinary
Nocturnal enuresis. In the 1960s, imipramine was shown—in some but not all placebo-controlled studies—to be beneficial for nocturnal enuresis in children and adults. Although imipramine is not FDA-approved for this indication, it is thought to work by relaxing bladder muscle and contracting bladder neck smooth muscle. Imipramine appears to have a vasopressin-independent antidiuretic effect in enuretic patients with nocturnal polyuria.
Duloxetine is thought to improve stress urinary incontinence by increasing urethral sphincter tone and the force of sphincter contraction. This indication is not FDA- approved for duloxetine but is approved in the European Union.
Oncology
At one time antidepressants were suggested to promote tumors, based on observations that amitriptyline, fluoxetine, and several antihistamines promoted tumor growth in rodents.21 In 1995, a few case reports associated these 2 antidepressants with atypical cutaneous lymphoid infiltrates.22 A review by Sternback in 200323 concluded that a link between antidepressants and cancer was questionable but acknowledged the need for very long-term studies.
Recently, a nested case-control study found an association between high-dose SSRI use for ≤5 years and reduced risk of colorectal cancer, whereas no association was found with tricyclic use.24 A study of this design does not establish a causal relationship, how-ever, and one can only speculate whether SSRIs might have direct cytotoxic or anti-promoter effects.
At present, it seems reasonable to continue to treat depressed cancer patients with antidepressants without concern that cancer will worsen or hope that it will improve as a result.
Immunology
The pathogenesis of depression may be linked to pro-inflammatory cytokines—proteins such as tumor necrosis factor-alpha (TNF-α) and certain interleukins that mediate immune function. Bupropion markedly lowered pro-inflammatory cytokine levels in a mouse inflammation model, prompting the authors to suggest that this anti-inflammatory effect be explored in humans.25
Case reports have suggested benefit from bupropion in Crohn’s disease, recurrent aphthous ulcerations, psoriasis, atopic dermatitis, and Blau syndrome (a rare autosomal-dominant trait characterized by granulomatous arthritis, iritis, and skin rash). Whether this antidepressant has much anti-inflammatory potential remains to be determined, however.
Delayed ejaculation is among the sexual side effects commonly associated with antidepressant medication. In a 6-week trial,27 3 selective serotonin reuptake inhibitors (SSRIs)— paroxetine, fluoxetine, and sertraline— were shown to improve intravaginal ejaculatory latency time (IELT) in men with lifelong rapid ejaculation. Compared with baseline, the greatest delay in ejaculation was seen with paroxetine, 20 mg/d, followed by fluoxetine, 20 mg/d, and then sertraline, 50 mg/d, whereas delay with fluvoxamine, 100 mg/d, did not differ significantly from placebo.
Dapoxetine is a non-antidepressant SSRI under investigation for on-demand treatment of moderate-to-severe premature ejaculation.28 In two 12-week, randomized, double-blind, placebo-controlled trials, 870 men took placebo, 874 took 30-mg dapoxetine, and 870 took 60-mg dapoxetine 1 to 3 hours before sexual activity. Efficacy was determined by IELT measured at home by stopwatch.
Both dapoxetine doses improved IELT significantly more than placebo (P
Nausea, diarrhea, headache, and dizziness occurred in ≤20% of patients and were more common with the 60-mg than 30-mg dapoxetine dose.
Infectious disease
Pathogenic protozoa—such as Trypanosoma cruzi (Chagas disease), Leishmania donovani (Kala-azar), Leishmania major (Oriental sore), and Giardia lamblia (Giardiasis)—infect millions of humans worldwide. Clomipramine has been shown in vitro and in mice to inhibit or kill these protozoa, but these potential benefits have not been extended to humans.
Sertraline, on the other hand, might exert antifungal activity. Three patients with recurrent vulvovaginal candidiasis had no episodes while being treated with sertraline for premenstrual dysphoric disorder but relapsed when the drug was discontinued.26 Although sertraline demonstrated antifungal activity in vitro against several Candida species, this SSRI seems unlikely to gain prominence as an antifungal agent.
Sexual function
Premature ejaculation. SSRIs are well-known causes of delayed or absent orgasm, but a perceived liability can become an asset in treating premature ejaculation. By measuring intravaginal ejaculation latency time under double-blind, placebo-controlled conditions, Waldinger et al27 showed pronounced delay in ejaculation with sertraline, fluoxetine, and paroxetine in men with long-standing rapid ejaculation. Dapoxetine—a short-acting non-antidepressant SSRI—is being studied as a treatment for this condition (Box 3).28
Spermicidal effect. SSRIs—including fluoxetine— have demonstrated in vitro spermicidal and antitrichomonas activity29 but are unlikely to be developed as microbicidal contraceptives.
Related Resources
- Gorman JM, Kent JM. SSRIs and SMRIs: broad spectrum of efficacy beyond major depression. J Clin Psychiatry 1999;60(suppl 4):33-8.
- About.com: Mental Health. Antidepressants for more than depression. http://mentalhealth.about.com/cs/psychopharmacology/a/antimore.htm.
- Amitriptyline • Elavil, Endep
- Bupropion • Wellbutrin, Zyban
- Citalopram • Celexa
- Clomipramine • Anafranil
- Desipramine • Norpramin, Pertofrane
- Doxepin • Adapin, Sinequan
- Duloxetine • Cymbalta
- Escitalopram • Lexapro
- Fludrocortisone • Florinef
- Fluoxetine • Prozac
- Fluvoxamine • Luvox
- Imipramine • Tofranil
- Midodrine • ProAmitine
- Nortriptyline • Pamelor, Aventyl
- Paroxetine • Paxil
- Phenelzine • Nardil
- Sertraline • Zoloft
- Trimipramine • Surmontil
- Venlafaxine • Effexor
Dr. Jefferson receives research support from Bristol-Myers Squibb, Forest Pharmaceuticals, GlaxoSmithKline, Janssen Pharmaceutica, Eli Lilly and Company, Novartis, Pfizer, Roche, Solvay, UCB Pharma, and Wyeth. He is a consultant to GlaxoSmithKline, Schwarz Pharma, Shire, and Organon and a speaker for Abbott Laboratories, AstraZeneca, Bristol-Myers Squibb, Forest Pharmaceuticals, GlaxoSmithKline, Eli Lilly and Company, Pfizer, Schwarz Pharma, Shire, and Wyeth. He holds stock in Bristol-Myers Squibb, GlaxoSmithKline, and SciClone.
1. Wernicke JF, Pritchett YL, D’Souza DN, et al. A randomized controlled trial of duloxetine in diabetic peripheral neuropathic pain. Neurology 2006;67(8):1411-20.
2. Raskin J, Pritchett YL, Wang F, et al. A double-blind, randomized multicenter trial comparing duloxetine with placebo in the management of diabetic peripheral neuropathic pain. Pain Med 2005;6(5):346-56.
3. Rowbotham MC, Goli V, Kunz NR, Lei D. Venlafaxine extended release in the treatment of painful diabetic neuropathy: a double-blind, placebo-controlled study. Pain 2004;110:697-706.
4. Max MB, Lynch SA, Muir J, et al. Effects of desipramine, amitriptyline, and fluoxetine on pain in diabetic neuropathy. N Engl J Med 1992;326:1250-6.
5. Sindrup SH, Otto M, Finnerup NB, Jensen TS. Antidepressants in the treatment of neuropathic pain. Basic Clin Pharmacol Toxicol 2005;96:399-409.
6. Semenchuk MR, Sherman S, Davis B. Double-blind, randomized trial of bupropion SR for the treatment of neuropathic pain. Neurology 2001;57:1583-8.
7. Tomkins GE, Jackson JL, O’Malley PG, et al. Treatment of chronic headache with antidepressants: a meta-analysis. Am J Med 2001;111:54-63.
8. Salerno SM, Browning R, Jackson JL. The effect of antidepressant treatment on chronic back pain. Arch Intern Med 2002;162:19-24.
9. Goldenberg DL, Burckhardt C, Crofford L. Management of fibromyalgia syndrome. JAMA 2004;292:2388-95.
10. Littlejohn GO, Guymer EK. Fibromyalgia syndrome: which antidepressant drug should we choose. Curr Pharm Des 2006;12(1):3-9.
11. 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;50:2974-84.
12. Arnold LM, Rosen A, Pritchett YL, et al. A randomized, double-blind, placebo-controlled trial of duloxetine in the treatment of women with fibromyalgia with or without major depressive disorder. Pain 2005;119:5-15.
13. Effects of encainide, flecainide, imipramine and moricizine on ventricular arrhythmias during the year after acute myocardial infarction: The CAPS. The Cardiac Arrhythmia Pilot Study (CAPS) Investigators. Am J Cardiol 1988;61(8):501-9.
14. Di Girolamo E, Di Iorio C, Sabatini P, et al. Effects of paroxetine hydrochloride, a selective serotonin reuptake inhibitor, on refractory vasovagal syncope: a randomized, double-blind, placebo-controlled study. J Am Coll Cardiol 1999;33:1227-30.
15. Jackson JL, O’Malley PG, Tomkins G, et al. Treatment of functional gastrointestinal disorders with antidepressant medications: a meta-analysis. Am J Med 2000;108:65-72.
16. Tabas G, Beaves M, Wang J, et al. Paroxetine to treat irritable bowel syndrome not responding to high-fiber diet: a double-blind, placebo-controlled trial. Am J Gastroenterol 2004;99(5):914-20.
17. Vahedi H, Merat S, Rashidioon A, et al. The effect of fluoxetine in patients with pain and constipation-predominant irritable bowel syndrome: a double-blind randomized-controlled study. Aliment Pharmacol Ther 2005;22:381-5.
18. Tack J, Broekaert D, Fischler B, et al. A controlled crossover study of the selective serotonin reuptake inhibitor citalopram in irritable bowel syndrome. Gut 2006;55:1095-103.
19. Talley NJ. Antidepressants in IBS: are we deluding ourselves? [editorial]. Am J Gastroenterol 2004;99:921-3.
20. Mariappan P, Ballantyne Z, N’Dow JM, Alhasso AA. Serotonin and noradrenaline reuptake inhibitors (SNRI) for stress urinary incontinence in adults. Cochrane Database Syst Rev 2005;Jul 20;(3):CD004742.-
21. Brandes LJ, Arron RJ, Bogdanovic RP, et al. Stimulation of malignant growth in rodents by antidepressant drugs at clinically relevant doses. Cancer Res 1992;52:3796-800.
22. Crowson AN, Magro CM. Antidepressant therapy. Arch Dermatol 1995;131:925-9.
23. Sternbach H. Are antidepressants carcinogenic? A review of preclinical and clinical studies. J Clin Psychiatry 2003;64:1153-62.
24. Xu W, Tamim H, Shapiro S, et al. Use of antidepressants and risk of colorectal cancer: a nested case-control study. Lancet Oncol 2006;7:301-8.
25. Brustolim D, Ribeiro-dos-Santos R, Kast RE, et al. A new chapter opens in anti-inflammatory treatments: the antidepressant bupropion lowers production of tumor necrosis factor-alpha and interferon-gamma in mice. Int Immunopharmacol 2006;6:903-7.
26. Lass-Flörl C, Dierich MP, Fuchs D, et al. Antifungal activity against Candida species of the selective serotonin-reuptake inhibitor, sertraline. Clin Infect Dis 2001;33:e135-6.
27. Waldinger MD, Hengeveld MW, Zsinderman AH, Olivier B. Effect of SSRI antidepressants on ejaculation: a double-blind, randomized, placebo-controlled study with fluoxetine, fluvoxamine, paroxetine, and sertraline. J Clin Psychopharmacol 1998;18(4):274-81.
28. Pryor JL, Althof SE, Steidle C, et al. Efficacy and tolerability of dapoxetine in treatment of premature ejaculation: an integrated analysis of two double-blind, randomised controlled trials. Lancet 2006;368:929-37.
29. Kumar VS, Sharma VL, Tiwari P, et al. The spermicidal and antitrichomonas activities of SSRI antidepressants. Bioorg Med Chem Lett 2006;16:2509-12.
A molecule is a molecule is a molecule—until it becomes identified with a purpose. Consider, for example, (-)-trans-4R-(4’-fluorophenyl)-3S-[(3’,4’-methylenedioxyphenoxy) methyl] piperidine. You probably know this molecule as paroxetine—an antidepressant, of course, but it is more than that. If you examine paroxetine’s FDA-approved indications, it also has anti-panic, anti-social anxiety, anti-obsessive-compulsive disorder, anti-posttraumatic stress disorder, and anti-premenstrual dysphoric disorder effects.
“Antidepressants” have achieved fame as antidepressants; one could say these molecules’ search for meaning has been fulfilled. Yet even within psychiatry, their many other uses (Table) can create semantic misunderstandings. Beyond psychiatry, consider the nondepressed patient with neurocardiogenic syncope who wonders why he’s being treated with an antidepressant.
Rather than calling antidepressants “panaceas,” the better choice is to educate patients about the drugs’ wide spectrum of activity. Let’s look broadly across the so-called antidepressants and examine their varied uses in psychiatry and other medical specialties.
Table
FDA-approved psychiatric indications for serotonin uptake inhibitors*
| SSRIs | ||||||||
| Citalopram | X | |||||||
| Escitalopram | X | X | ||||||
| Fluoxetine | X | X | X | X | X | |||
| Fluvoxamine | X | |||||||
| Paroxetine | X | X | X | X | X | X | X | |
| Sertraline | X | X | X | X | X | X | ||
| SNRIs | ||||||||
| Duloxetine | X | X | ||||||
| Venlafaxine | X | X | X | X | ||||
| SSRIs: selective serotonin reuptake inhibitors; SNRIs: serotonin-norepinephrine reuptake inhibitors; MDD: major depressive disorder; PD: panic disorder; SAD: social anxiety disorder; PTSD: posttraumatic stress disorder; GAD: generalized anxiety disorder; OCD: obsessive-compulsive disorder; PMDD: premenstrual dysphoric disorder; BUL: bulimia | ||||||||
| * The absence of an X does not necessarily imply that a drug is ineffective for a given indication but, more likely, that definitive studies are lacking. | ||||||||
Pain syndromes
Peripheral neuropathy. The only antidepressant with an FDA-approved pain indication is duloxetine, a serotonin-norepinephrine reuptake inhibitor (SNRI). Its approval for diabetic peripheral neuropathic pain (DPNP) was based on two 12-week, randomized, double-blind, placebo-controlled studies using fixed doses of 60 mg once or twice daily.1,2 Another SNRI—venlafaxine XR, 150 to 225 mg/d, but not 75 mg/d—also was found to be more effective than placebo for this indication in a 6-week, double-blind study (Box 1).3
Using antidepressants to treat pain syndromes is neither new nor restricted to SNRIs, however. In combined double-blind, cross-over studies of patients with DPNP, Max et al4 found:
- moderate or greater pain relief in 74% and 61% of subjects, respectively, from the tricyclics amitriptyline, mean 105 mg/d, and desipramine, mean 111 mg/d—with pain reduced by equal amounts in depressed and nondepressed patients
- no statistically significant difference in pain relief between the selective serotonin reuptake inhibitor (SSRI) fluoxetine, 40 mg/d, and placebo.
- tricyclics: 1 in every 2 to 3 patients
- SNRIs: 1 in every 4 to 5 patients
- SSRIs: 1 in every 7 patients.
Chronic headache. A meta-analysis7 of randomized, placebo-controlled studies found antidepressants more effective than placebo for chronic migraine and tension headache prophylaxis. Although a subgroup meta-analysis found similar effects for tricyclics and SSRIs, the authors characterized the tricyclics’ results as well established and the SSRIs’ as “less certain.”
The results of this meta-analysis might not accurately reflect bona fide antidepressants, however. Some of the 38 studies (25 of migraine, 12 of tension headache, 1 of both) included treatment with serotonin antagonists—most commonly pizotifen, which is not available in the United States and does not appear to be an antidepressant.
Back pain. Patients with chronic low back pain (average 10 years) seem to benefit from antidepressants, according to a meta-analysis of 9 randomized, controlled trials by Salerno et al.8 The effect on pain in the total 504 patients was “small but significant,” and improvement in function was “small but nonsignificant.” Individual sample sizes also were small, however, and only 2 studies excluded depressed patients.
Fibromyalgia, with chronic generalized musculoskeletal pain and tenderness, has been a focus of antidepressant drug therapy. Goldenberg et al9 concluded from an ambitious literature review (505 articles) that evidence of efficacy was strong for amitripty-line and modest for SSRIs and SNRIs.
Overall, antidepressants are generally understood to have analgesic effects in the absence of depression. Benefits for patients with pain syndromes are well established for tricyclics (especially amitriptyline) and recently with SNRIs, whereas SSRIs are less effective.
Serotonin and norepinephrine are involved in pain modulation via descending inhibitory pathways in the brain and spinal cord. Serotonin-norepinephrine reuptake inhibitors (SNRIs) have been shown to reduce the severity of diabetic peripheral neuropathic pain (DPNP) in randomized controlled trials.
Duloxetine. In 2 double-blind studies,1,2 nondepressed patients with DPNP received duloxetine, 60 mg once daily; duloxetine, 60 mg bid; or placebo for 12 weeks. They rated the severity of neuropathic pain every 24 hours on an 11-point Likert scale, and weekly mean scores were the primary outcome measure. Average pain scores improved more in both duloxetine groups vs placebo. Duloxetine treatment did not interfere with diabetic control, and both dosages were well tolerated.
The FDA approved an added indication for duloxetine in the management of DPNP.
Venlafaxine. In a double-blind study,3 244 adult outpatients with moderately severe DPNP received venlafaxine ER, 75 or 150 to 225 mg/d, or placebo for 6 weeks. Daily scores on the Visual Analog Pain Intensity (VAS-PI) and Pain Relief (VAS-PR) scales were primary efficacy measures.
Patients receiving the higher venlafaxine dosage—but not 75 mg/d—showed statistically significant less-intensive pain vs placebo. VAS-PI scores were 27% lower than at enrollment with placebo, 32% lower with venlafaxine, 75 mg/d, and 50% lower with venlafaxine, 150 to 225 mg/d (P
Nausea and somnolence were the most common side effects; clinically important ECG changes occurred in 7 patients treated with venlafaxine, 150 to 225 mg/d.
Smoking cessation
Bupropion SR is FDA-approved to aid smoking cessation, and this effect is independent of the drug’s antidepressant activity. Bupropion may act as a nicotine receptor antagonist as well as a norepinephrine dopamine reuptake inhibitor.
Other antidepressants have been studied for smoking cessation, with nortriptyline showing benefit in 2 large placebo-controlled trials. Studies with doxepin, fluoxetine, and moclobemide found little or no benefit for this indication.
Cardiovascular uses
Angina. Monoamine oxidase inhibitor (MAOI) antidepressants were used to treat angina pectoris in the late 1950s and early 1960s. This practice stopped after evidence showed that whereas angina pain may have improved with MAOIs, stress-induced ischemia on ECG did not.
Antiarrhythmia. Tricyclics had a brief fling in cardiovascular therapeutics when their quinidine-like class I antiarrhythmic activity was recognized. Imipramine was one of several drugs included in the Cardiac Arrhythmia Pilot Study in the 1980s that involved 502 postmyocardial infarction patients with ventricular arrhythmias. Imipramine was the least effective of the 4 drugs studied and the least well tolerated.13
variety of medications. Options include the vasopressor midodrine, fludrocortisone, beta blockers, and SSRIs— none
FDA-approved for this indication. Paroxetine, 20 mg/d, was considerably more effective than placebo in preventing
recurrent syncope in 68 patients who had been unresponsive
to or intolerant of traditional medications. During a mean 25 months of treatment, 82% of patients remained syncope-free on paroxetine vs 47% on placebo.14
Selective serotonin reuptake inhibitors (SSRIs) often are used to treat irritable bowel syndrome (IBS), though evidence of their effectiveness is scarce. SSRIs can improve IBS patients’ quality of life, but effects on abdominal pain and bloating are less clear.
Paroxetine. In a randomized, double-blind trial,16 gastroenterologists tested a highfiber diet plus paroxetine in nondepressed patients with IBS. Ninety-eight patients ages 18 to 65 who experienced IBS symptoms on low- or average-fiber diets were first put on high-fiber diets and assessed for well-being and abdominal pain and bloating. Of these, 81 symptomatic patients continued highfiber diets with added paroxetine, 10 to 40 mg/d (n=38) or placebo (n=43).
With paroxetine, patients’ overall well-being improved more than with placebo, but abdominal pain and bloating and social functioning did not.
Fluoxetine. In a double-blind, randomized trial,17 44 patients with pain and constipation-predominant IBS received fluoxetine, 20 mg/d, or placebo for 12 weeks. These patients met Rome II criteria for IBS—abdominal discomfort/pain for ≥12 weeks in past year that met 2 of 3 criteria:
- relieved by defecation
- onset associated with change in stool frequency
- onset associated with change in stool appearance.
Patients receiving fluoxetine had less abdominal discomfort, less bloating, more frequent bowel movements, and decreased consistency of stool vs placebo 4 weeks after treatment stopped. Mean number of symptoms per patient decreased from 4.6 to 0.7 in the fluoxetine group vs 4.5 to 2.9 in controls (P
Citalopram. IBS symptom severity was the primary outcome in a crossover trial comparing citalopram (20 mg for 3 weeks and 40 mg for 3 weeks) with placebo in 23 nondepressed patients.18 Abdominal pain and bloating, impact of symptoms on daily life, and overall well-being improved significantly more with citalopram than with placebo after 3 and 6 weeks.
Symptom improvements were not related to changes in depression, anxiety, or colonic sensorimotor function.
Gastrointestinal
Peptic ulcer disease was shown in the 1980s to respond to tricyclic antidepressants. At the time, both anticholinergic and antihistaminic effects were thought to be responsible, but the later observation that trimipramine inhibited Campylobacter pylori in vitro suggested an additional explanation. Today, tricyclics are only of historic interest as treatments for peptic ulcer.
Irritable bowel syndrome (IBS) patients have responded favorably to antidepressants, although it is often difficult to know if the benefit is independent of improved coexisting anxiety or depression. A meta-analysis of 12 randomized, placebo-controlled trials—mostly with tricyclics—found an odds ratio for improvement of 4.2 and a number needed to treat of 3.2.15
More recently, a few placebo-controlled studies have shown SSRIs to be beneficial for IBS,16-18 although not all symptoms improved and some IBS subtypes might be more responsive than others (Box 2). In an editorial, Talley19 concluded that antidepressant therapy of IBS was “at best only a ‘band-aid’ approach to management.”
Genitourinary
Nocturnal enuresis. In the 1960s, imipramine was shown—in some but not all placebo-controlled studies—to be beneficial for nocturnal enuresis in children and adults. Although imipramine is not FDA-approved for this indication, it is thought to work by relaxing bladder muscle and contracting bladder neck smooth muscle. Imipramine appears to have a vasopressin-independent antidiuretic effect in enuretic patients with nocturnal polyuria.
Duloxetine is thought to improve stress urinary incontinence by increasing urethral sphincter tone and the force of sphincter contraction. This indication is not FDA- approved for duloxetine but is approved in the European Union.
Oncology
At one time antidepressants were suggested to promote tumors, based on observations that amitriptyline, fluoxetine, and several antihistamines promoted tumor growth in rodents.21 In 1995, a few case reports associated these 2 antidepressants with atypical cutaneous lymphoid infiltrates.22 A review by Sternback in 200323 concluded that a link between antidepressants and cancer was questionable but acknowledged the need for very long-term studies.
Recently, a nested case-control study found an association between high-dose SSRI use for ≤5 years and reduced risk of colorectal cancer, whereas no association was found with tricyclic use.24 A study of this design does not establish a causal relationship, how-ever, and one can only speculate whether SSRIs might have direct cytotoxic or anti-promoter effects.
At present, it seems reasonable to continue to treat depressed cancer patients with antidepressants without concern that cancer will worsen or hope that it will improve as a result.
Immunology
The pathogenesis of depression may be linked to pro-inflammatory cytokines—proteins such as tumor necrosis factor-alpha (TNF-α) and certain interleukins that mediate immune function. Bupropion markedly lowered pro-inflammatory cytokine levels in a mouse inflammation model, prompting the authors to suggest that this anti-inflammatory effect be explored in humans.25
Case reports have suggested benefit from bupropion in Crohn’s disease, recurrent aphthous ulcerations, psoriasis, atopic dermatitis, and Blau syndrome (a rare autosomal-dominant trait characterized by granulomatous arthritis, iritis, and skin rash). Whether this antidepressant has much anti-inflammatory potential remains to be determined, however.
Delayed ejaculation is among the sexual side effects commonly associated with antidepressant medication. In a 6-week trial,27 3 selective serotonin reuptake inhibitors (SSRIs)— paroxetine, fluoxetine, and sertraline— were shown to improve intravaginal ejaculatory latency time (IELT) in men with lifelong rapid ejaculation. Compared with baseline, the greatest delay in ejaculation was seen with paroxetine, 20 mg/d, followed by fluoxetine, 20 mg/d, and then sertraline, 50 mg/d, whereas delay with fluvoxamine, 100 mg/d, did not differ significantly from placebo.
Dapoxetine is a non-antidepressant SSRI under investigation for on-demand treatment of moderate-to-severe premature ejaculation.28 In two 12-week, randomized, double-blind, placebo-controlled trials, 870 men took placebo, 874 took 30-mg dapoxetine, and 870 took 60-mg dapoxetine 1 to 3 hours before sexual activity. Efficacy was determined by IELT measured at home by stopwatch.
Both dapoxetine doses improved IELT significantly more than placebo (P
Nausea, diarrhea, headache, and dizziness occurred in ≤20% of patients and were more common with the 60-mg than 30-mg dapoxetine dose.
Infectious disease
Pathogenic protozoa—such as Trypanosoma cruzi (Chagas disease), Leishmania donovani (Kala-azar), Leishmania major (Oriental sore), and Giardia lamblia (Giardiasis)—infect millions of humans worldwide. Clomipramine has been shown in vitro and in mice to inhibit or kill these protozoa, but these potential benefits have not been extended to humans.
Sertraline, on the other hand, might exert antifungal activity. Three patients with recurrent vulvovaginal candidiasis had no episodes while being treated with sertraline for premenstrual dysphoric disorder but relapsed when the drug was discontinued.26 Although sertraline demonstrated antifungal activity in vitro against several Candida species, this SSRI seems unlikely to gain prominence as an antifungal agent.
Sexual function
Premature ejaculation. SSRIs are well-known causes of delayed or absent orgasm, but a perceived liability can become an asset in treating premature ejaculation. By measuring intravaginal ejaculation latency time under double-blind, placebo-controlled conditions, Waldinger et al27 showed pronounced delay in ejaculation with sertraline, fluoxetine, and paroxetine in men with long-standing rapid ejaculation. Dapoxetine—a short-acting non-antidepressant SSRI—is being studied as a treatment for this condition (Box 3).28
Spermicidal effect. SSRIs—including fluoxetine— have demonstrated in vitro spermicidal and antitrichomonas activity29 but are unlikely to be developed as microbicidal contraceptives.
Related Resources
- Gorman JM, Kent JM. SSRIs and SMRIs: broad spectrum of efficacy beyond major depression. J Clin Psychiatry 1999;60(suppl 4):33-8.
- About.com: Mental Health. Antidepressants for more than depression. http://mentalhealth.about.com/cs/psychopharmacology/a/antimore.htm.
- Amitriptyline • Elavil, Endep
- Bupropion • Wellbutrin, Zyban
- Citalopram • Celexa
- Clomipramine • Anafranil
- Desipramine • Norpramin, Pertofrane
- Doxepin • Adapin, Sinequan
- Duloxetine • Cymbalta
- Escitalopram • Lexapro
- Fludrocortisone • Florinef
- Fluoxetine • Prozac
- Fluvoxamine • Luvox
- Imipramine • Tofranil
- Midodrine • ProAmitine
- Nortriptyline • Pamelor, Aventyl
- Paroxetine • Paxil
- Phenelzine • Nardil
- Sertraline • Zoloft
- Trimipramine • Surmontil
- Venlafaxine • Effexor
Dr. Jefferson receives research support from Bristol-Myers Squibb, Forest Pharmaceuticals, GlaxoSmithKline, Janssen Pharmaceutica, Eli Lilly and Company, Novartis, Pfizer, Roche, Solvay, UCB Pharma, and Wyeth. He is a consultant to GlaxoSmithKline, Schwarz Pharma, Shire, and Organon and a speaker for Abbott Laboratories, AstraZeneca, Bristol-Myers Squibb, Forest Pharmaceuticals, GlaxoSmithKline, Eli Lilly and Company, Pfizer, Schwarz Pharma, Shire, and Wyeth. He holds stock in Bristol-Myers Squibb, GlaxoSmithKline, and SciClone.
A molecule is a molecule is a molecule—until it becomes identified with a purpose. Consider, for example, (-)-trans-4R-(4’-fluorophenyl)-3S-[(3’,4’-methylenedioxyphenoxy) methyl] piperidine. You probably know this molecule as paroxetine—an antidepressant, of course, but it is more than that. If you examine paroxetine’s FDA-approved indications, it also has anti-panic, anti-social anxiety, anti-obsessive-compulsive disorder, anti-posttraumatic stress disorder, and anti-premenstrual dysphoric disorder effects.
“Antidepressants” have achieved fame as antidepressants; one could say these molecules’ search for meaning has been fulfilled. Yet even within psychiatry, their many other uses (Table) can create semantic misunderstandings. Beyond psychiatry, consider the nondepressed patient with neurocardiogenic syncope who wonders why he’s being treated with an antidepressant.
Rather than calling antidepressants “panaceas,” the better choice is to educate patients about the drugs’ wide spectrum of activity. Let’s look broadly across the so-called antidepressants and examine their varied uses in psychiatry and other medical specialties.
Table
FDA-approved psychiatric indications for serotonin uptake inhibitors*
| SSRIs | ||||||||
| Citalopram | X | |||||||
| Escitalopram | X | X | ||||||
| Fluoxetine | X | X | X | X | X | |||
| Fluvoxamine | X | |||||||
| Paroxetine | X | X | X | X | X | X | X | |
| Sertraline | X | X | X | X | X | X | ||
| SNRIs | ||||||||
| Duloxetine | X | X | ||||||
| Venlafaxine | X | X | X | X | ||||
| SSRIs: selective serotonin reuptake inhibitors; SNRIs: serotonin-norepinephrine reuptake inhibitors; MDD: major depressive disorder; PD: panic disorder; SAD: social anxiety disorder; PTSD: posttraumatic stress disorder; GAD: generalized anxiety disorder; OCD: obsessive-compulsive disorder; PMDD: premenstrual dysphoric disorder; BUL: bulimia | ||||||||
| * The absence of an X does not necessarily imply that a drug is ineffective for a given indication but, more likely, that definitive studies are lacking. | ||||||||
Pain syndromes
Peripheral neuropathy. The only antidepressant with an FDA-approved pain indication is duloxetine, a serotonin-norepinephrine reuptake inhibitor (SNRI). Its approval for diabetic peripheral neuropathic pain (DPNP) was based on two 12-week, randomized, double-blind, placebo-controlled studies using fixed doses of 60 mg once or twice daily.1,2 Another SNRI—venlafaxine XR, 150 to 225 mg/d, but not 75 mg/d—also was found to be more effective than placebo for this indication in a 6-week, double-blind study (Box 1).3
Using antidepressants to treat pain syndromes is neither new nor restricted to SNRIs, however. In combined double-blind, cross-over studies of patients with DPNP, Max et al4 found:
- moderate or greater pain relief in 74% and 61% of subjects, respectively, from the tricyclics amitriptyline, mean 105 mg/d, and desipramine, mean 111 mg/d—with pain reduced by equal amounts in depressed and nondepressed patients
- no statistically significant difference in pain relief between the selective serotonin reuptake inhibitor (SSRI) fluoxetine, 40 mg/d, and placebo.
- tricyclics: 1 in every 2 to 3 patients
- SNRIs: 1 in every 4 to 5 patients
- SSRIs: 1 in every 7 patients.
Chronic headache. A meta-analysis7 of randomized, placebo-controlled studies found antidepressants more effective than placebo for chronic migraine and tension headache prophylaxis. Although a subgroup meta-analysis found similar effects for tricyclics and SSRIs, the authors characterized the tricyclics’ results as well established and the SSRIs’ as “less certain.”
The results of this meta-analysis might not accurately reflect bona fide antidepressants, however. Some of the 38 studies (25 of migraine, 12 of tension headache, 1 of both) included treatment with serotonin antagonists—most commonly pizotifen, which is not available in the United States and does not appear to be an antidepressant.
Back pain. Patients with chronic low back pain (average 10 years) seem to benefit from antidepressants, according to a meta-analysis of 9 randomized, controlled trials by Salerno et al.8 The effect on pain in the total 504 patients was “small but significant,” and improvement in function was “small but nonsignificant.” Individual sample sizes also were small, however, and only 2 studies excluded depressed patients.
Fibromyalgia, with chronic generalized musculoskeletal pain and tenderness, has been a focus of antidepressant drug therapy. Goldenberg et al9 concluded from an ambitious literature review (505 articles) that evidence of efficacy was strong for amitripty-line and modest for SSRIs and SNRIs.
Overall, antidepressants are generally understood to have analgesic effects in the absence of depression. Benefits for patients with pain syndromes are well established for tricyclics (especially amitriptyline) and recently with SNRIs, whereas SSRIs are less effective.
Serotonin and norepinephrine are involved in pain modulation via descending inhibitory pathways in the brain and spinal cord. Serotonin-norepinephrine reuptake inhibitors (SNRIs) have been shown to reduce the severity of diabetic peripheral neuropathic pain (DPNP) in randomized controlled trials.
Duloxetine. In 2 double-blind studies,1,2 nondepressed patients with DPNP received duloxetine, 60 mg once daily; duloxetine, 60 mg bid; or placebo for 12 weeks. They rated the severity of neuropathic pain every 24 hours on an 11-point Likert scale, and weekly mean scores were the primary outcome measure. Average pain scores improved more in both duloxetine groups vs placebo. Duloxetine treatment did not interfere with diabetic control, and both dosages were well tolerated.
The FDA approved an added indication for duloxetine in the management of DPNP.
Venlafaxine. In a double-blind study,3 244 adult outpatients with moderately severe DPNP received venlafaxine ER, 75 or 150 to 225 mg/d, or placebo for 6 weeks. Daily scores on the Visual Analog Pain Intensity (VAS-PI) and Pain Relief (VAS-PR) scales were primary efficacy measures.
Patients receiving the higher venlafaxine dosage—but not 75 mg/d—showed statistically significant less-intensive pain vs placebo. VAS-PI scores were 27% lower than at enrollment with placebo, 32% lower with venlafaxine, 75 mg/d, and 50% lower with venlafaxine, 150 to 225 mg/d (P
Nausea and somnolence were the most common side effects; clinically important ECG changes occurred in 7 patients treated with venlafaxine, 150 to 225 mg/d.
Smoking cessation
Bupropion SR is FDA-approved to aid smoking cessation, and this effect is independent of the drug’s antidepressant activity. Bupropion may act as a nicotine receptor antagonist as well as a norepinephrine dopamine reuptake inhibitor.
Other antidepressants have been studied for smoking cessation, with nortriptyline showing benefit in 2 large placebo-controlled trials. Studies with doxepin, fluoxetine, and moclobemide found little or no benefit for this indication.
Cardiovascular uses
Angina. Monoamine oxidase inhibitor (MAOI) antidepressants were used to treat angina pectoris in the late 1950s and early 1960s. This practice stopped after evidence showed that whereas angina pain may have improved with MAOIs, stress-induced ischemia on ECG did not.
Antiarrhythmia. Tricyclics had a brief fling in cardiovascular therapeutics when their quinidine-like class I antiarrhythmic activity was recognized. Imipramine was one of several drugs included in the Cardiac Arrhythmia Pilot Study in the 1980s that involved 502 postmyocardial infarction patients with ventricular arrhythmias. Imipramine was the least effective of the 4 drugs studied and the least well tolerated.13
variety of medications. Options include the vasopressor midodrine, fludrocortisone, beta blockers, and SSRIs— none
FDA-approved for this indication. Paroxetine, 20 mg/d, was considerably more effective than placebo in preventing
recurrent syncope in 68 patients who had been unresponsive
to or intolerant of traditional medications. During a mean 25 months of treatment, 82% of patients remained syncope-free on paroxetine vs 47% on placebo.14
Selective serotonin reuptake inhibitors (SSRIs) often are used to treat irritable bowel syndrome (IBS), though evidence of their effectiveness is scarce. SSRIs can improve IBS patients’ quality of life, but effects on abdominal pain and bloating are less clear.
Paroxetine. In a randomized, double-blind trial,16 gastroenterologists tested a highfiber diet plus paroxetine in nondepressed patients with IBS. Ninety-eight patients ages 18 to 65 who experienced IBS symptoms on low- or average-fiber diets were first put on high-fiber diets and assessed for well-being and abdominal pain and bloating. Of these, 81 symptomatic patients continued highfiber diets with added paroxetine, 10 to 40 mg/d (n=38) or placebo (n=43).
With paroxetine, patients’ overall well-being improved more than with placebo, but abdominal pain and bloating and social functioning did not.
Fluoxetine. In a double-blind, randomized trial,17 44 patients with pain and constipation-predominant IBS received fluoxetine, 20 mg/d, or placebo for 12 weeks. These patients met Rome II criteria for IBS—abdominal discomfort/pain for ≥12 weeks in past year that met 2 of 3 criteria:
- relieved by defecation
- onset associated with change in stool frequency
- onset associated with change in stool appearance.
Patients receiving fluoxetine had less abdominal discomfort, less bloating, more frequent bowel movements, and decreased consistency of stool vs placebo 4 weeks after treatment stopped. Mean number of symptoms per patient decreased from 4.6 to 0.7 in the fluoxetine group vs 4.5 to 2.9 in controls (P
Citalopram. IBS symptom severity was the primary outcome in a crossover trial comparing citalopram (20 mg for 3 weeks and 40 mg for 3 weeks) with placebo in 23 nondepressed patients.18 Abdominal pain and bloating, impact of symptoms on daily life, and overall well-being improved significantly more with citalopram than with placebo after 3 and 6 weeks.
Symptom improvements were not related to changes in depression, anxiety, or colonic sensorimotor function.
Gastrointestinal
Peptic ulcer disease was shown in the 1980s to respond to tricyclic antidepressants. At the time, both anticholinergic and antihistaminic effects were thought to be responsible, but the later observation that trimipramine inhibited Campylobacter pylori in vitro suggested an additional explanation. Today, tricyclics are only of historic interest as treatments for peptic ulcer.
Irritable bowel syndrome (IBS) patients have responded favorably to antidepressants, although it is often difficult to know if the benefit is independent of improved coexisting anxiety or depression. A meta-analysis of 12 randomized, placebo-controlled trials—mostly with tricyclics—found an odds ratio for improvement of 4.2 and a number needed to treat of 3.2.15
More recently, a few placebo-controlled studies have shown SSRIs to be beneficial for IBS,16-18 although not all symptoms improved and some IBS subtypes might be more responsive than others (Box 2). In an editorial, Talley19 concluded that antidepressant therapy of IBS was “at best only a ‘band-aid’ approach to management.”
Genitourinary
Nocturnal enuresis. In the 1960s, imipramine was shown—in some but not all placebo-controlled studies—to be beneficial for nocturnal enuresis in children and adults. Although imipramine is not FDA-approved for this indication, it is thought to work by relaxing bladder muscle and contracting bladder neck smooth muscle. Imipramine appears to have a vasopressin-independent antidiuretic effect in enuretic patients with nocturnal polyuria.
Duloxetine is thought to improve stress urinary incontinence by increasing urethral sphincter tone and the force of sphincter contraction. This indication is not FDA- approved for duloxetine but is approved in the European Union.
Oncology
At one time antidepressants were suggested to promote tumors, based on observations that amitriptyline, fluoxetine, and several antihistamines promoted tumor growth in rodents.21 In 1995, a few case reports associated these 2 antidepressants with atypical cutaneous lymphoid infiltrates.22 A review by Sternback in 200323 concluded that a link between antidepressants and cancer was questionable but acknowledged the need for very long-term studies.
Recently, a nested case-control study found an association between high-dose SSRI use for ≤5 years and reduced risk of colorectal cancer, whereas no association was found with tricyclic use.24 A study of this design does not establish a causal relationship, how-ever, and one can only speculate whether SSRIs might have direct cytotoxic or anti-promoter effects.
At present, it seems reasonable to continue to treat depressed cancer patients with antidepressants without concern that cancer will worsen or hope that it will improve as a result.
Immunology
The pathogenesis of depression may be linked to pro-inflammatory cytokines—proteins such as tumor necrosis factor-alpha (TNF-α) and certain interleukins that mediate immune function. Bupropion markedly lowered pro-inflammatory cytokine levels in a mouse inflammation model, prompting the authors to suggest that this anti-inflammatory effect be explored in humans.25
Case reports have suggested benefit from bupropion in Crohn’s disease, recurrent aphthous ulcerations, psoriasis, atopic dermatitis, and Blau syndrome (a rare autosomal-dominant trait characterized by granulomatous arthritis, iritis, and skin rash). Whether this antidepressant has much anti-inflammatory potential remains to be determined, however.
Delayed ejaculation is among the sexual side effects commonly associated with antidepressant medication. In a 6-week trial,27 3 selective serotonin reuptake inhibitors (SSRIs)— paroxetine, fluoxetine, and sertraline— were shown to improve intravaginal ejaculatory latency time (IELT) in men with lifelong rapid ejaculation. Compared with baseline, the greatest delay in ejaculation was seen with paroxetine, 20 mg/d, followed by fluoxetine, 20 mg/d, and then sertraline, 50 mg/d, whereas delay with fluvoxamine, 100 mg/d, did not differ significantly from placebo.
Dapoxetine is a non-antidepressant SSRI under investigation for on-demand treatment of moderate-to-severe premature ejaculation.28 In two 12-week, randomized, double-blind, placebo-controlled trials, 870 men took placebo, 874 took 30-mg dapoxetine, and 870 took 60-mg dapoxetine 1 to 3 hours before sexual activity. Efficacy was determined by IELT measured at home by stopwatch.
Both dapoxetine doses improved IELT significantly more than placebo (P
Nausea, diarrhea, headache, and dizziness occurred in ≤20% of patients and were more common with the 60-mg than 30-mg dapoxetine dose.
Infectious disease
Pathogenic protozoa—such as Trypanosoma cruzi (Chagas disease), Leishmania donovani (Kala-azar), Leishmania major (Oriental sore), and Giardia lamblia (Giardiasis)—infect millions of humans worldwide. Clomipramine has been shown in vitro and in mice to inhibit or kill these protozoa, but these potential benefits have not been extended to humans.
Sertraline, on the other hand, might exert antifungal activity. Three patients with recurrent vulvovaginal candidiasis had no episodes while being treated with sertraline for premenstrual dysphoric disorder but relapsed when the drug was discontinued.26 Although sertraline demonstrated antifungal activity in vitro against several Candida species, this SSRI seems unlikely to gain prominence as an antifungal agent.
Sexual function
Premature ejaculation. SSRIs are well-known causes of delayed or absent orgasm, but a perceived liability can become an asset in treating premature ejaculation. By measuring intravaginal ejaculation latency time under double-blind, placebo-controlled conditions, Waldinger et al27 showed pronounced delay in ejaculation with sertraline, fluoxetine, and paroxetine in men with long-standing rapid ejaculation. Dapoxetine—a short-acting non-antidepressant SSRI—is being studied as a treatment for this condition (Box 3).28
Spermicidal effect. SSRIs—including fluoxetine— have demonstrated in vitro spermicidal and antitrichomonas activity29 but are unlikely to be developed as microbicidal contraceptives.
Related Resources
- Gorman JM, Kent JM. SSRIs and SMRIs: broad spectrum of efficacy beyond major depression. J Clin Psychiatry 1999;60(suppl 4):33-8.
- About.com: Mental Health. Antidepressants for more than depression. http://mentalhealth.about.com/cs/psychopharmacology/a/antimore.htm.
- Amitriptyline • Elavil, Endep
- Bupropion • Wellbutrin, Zyban
- Citalopram • Celexa
- Clomipramine • Anafranil
- Desipramine • Norpramin, Pertofrane
- Doxepin • Adapin, Sinequan
- Duloxetine • Cymbalta
- Escitalopram • Lexapro
- Fludrocortisone • Florinef
- Fluoxetine • Prozac
- Fluvoxamine • Luvox
- Imipramine • Tofranil
- Midodrine • ProAmitine
- Nortriptyline • Pamelor, Aventyl
- Paroxetine • Paxil
- Phenelzine • Nardil
- Sertraline • Zoloft
- Trimipramine • Surmontil
- Venlafaxine • Effexor
Dr. Jefferson receives research support from Bristol-Myers Squibb, Forest Pharmaceuticals, GlaxoSmithKline, Janssen Pharmaceutica, Eli Lilly and Company, Novartis, Pfizer, Roche, Solvay, UCB Pharma, and Wyeth. He is a consultant to GlaxoSmithKline, Schwarz Pharma, Shire, and Organon and a speaker for Abbott Laboratories, AstraZeneca, Bristol-Myers Squibb, Forest Pharmaceuticals, GlaxoSmithKline, Eli Lilly and Company, Pfizer, Schwarz Pharma, Shire, and Wyeth. He holds stock in Bristol-Myers Squibb, GlaxoSmithKline, and SciClone.
1. Wernicke JF, Pritchett YL, D’Souza DN, et al. A randomized controlled trial of duloxetine in diabetic peripheral neuropathic pain. Neurology 2006;67(8):1411-20.
2. Raskin J, Pritchett YL, Wang F, et al. A double-blind, randomized multicenter trial comparing duloxetine with placebo in the management of diabetic peripheral neuropathic pain. Pain Med 2005;6(5):346-56.
3. Rowbotham MC, Goli V, Kunz NR, Lei D. Venlafaxine extended release in the treatment of painful diabetic neuropathy: a double-blind, placebo-controlled study. Pain 2004;110:697-706.
4. Max MB, Lynch SA, Muir J, et al. Effects of desipramine, amitriptyline, and fluoxetine on pain in diabetic neuropathy. N Engl J Med 1992;326:1250-6.
5. Sindrup SH, Otto M, Finnerup NB, Jensen TS. Antidepressants in the treatment of neuropathic pain. Basic Clin Pharmacol Toxicol 2005;96:399-409.
6. Semenchuk MR, Sherman S, Davis B. Double-blind, randomized trial of bupropion SR for the treatment of neuropathic pain. Neurology 2001;57:1583-8.
7. Tomkins GE, Jackson JL, O’Malley PG, et al. Treatment of chronic headache with antidepressants: a meta-analysis. Am J Med 2001;111:54-63.
8. Salerno SM, Browning R, Jackson JL. The effect of antidepressant treatment on chronic back pain. Arch Intern Med 2002;162:19-24.
9. Goldenberg DL, Burckhardt C, Crofford L. Management of fibromyalgia syndrome. JAMA 2004;292:2388-95.
10. Littlejohn GO, Guymer EK. Fibromyalgia syndrome: which antidepressant drug should we choose. Curr Pharm Des 2006;12(1):3-9.
11. 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;50:2974-84.
12. Arnold LM, Rosen A, Pritchett YL, et al. A randomized, double-blind, placebo-controlled trial of duloxetine in the treatment of women with fibromyalgia with or without major depressive disorder. Pain 2005;119:5-15.
13. Effects of encainide, flecainide, imipramine and moricizine on ventricular arrhythmias during the year after acute myocardial infarction: The CAPS. The Cardiac Arrhythmia Pilot Study (CAPS) Investigators. Am J Cardiol 1988;61(8):501-9.
14. Di Girolamo E, Di Iorio C, Sabatini P, et al. Effects of paroxetine hydrochloride, a selective serotonin reuptake inhibitor, on refractory vasovagal syncope: a randomized, double-blind, placebo-controlled study. J Am Coll Cardiol 1999;33:1227-30.
15. Jackson JL, O’Malley PG, Tomkins G, et al. Treatment of functional gastrointestinal disorders with antidepressant medications: a meta-analysis. Am J Med 2000;108:65-72.
16. Tabas G, Beaves M, Wang J, et al. Paroxetine to treat irritable bowel syndrome not responding to high-fiber diet: a double-blind, placebo-controlled trial. Am J Gastroenterol 2004;99(5):914-20.
17. Vahedi H, Merat S, Rashidioon A, et al. The effect of fluoxetine in patients with pain and constipation-predominant irritable bowel syndrome: a double-blind randomized-controlled study. Aliment Pharmacol Ther 2005;22:381-5.
18. Tack J, Broekaert D, Fischler B, et al. A controlled crossover study of the selective serotonin reuptake inhibitor citalopram in irritable bowel syndrome. Gut 2006;55:1095-103.
19. Talley NJ. Antidepressants in IBS: are we deluding ourselves? [editorial]. Am J Gastroenterol 2004;99:921-3.
20. Mariappan P, Ballantyne Z, N’Dow JM, Alhasso AA. Serotonin and noradrenaline reuptake inhibitors (SNRI) for stress urinary incontinence in adults. Cochrane Database Syst Rev 2005;Jul 20;(3):CD004742.-
21. Brandes LJ, Arron RJ, Bogdanovic RP, et al. Stimulation of malignant growth in rodents by antidepressant drugs at clinically relevant doses. Cancer Res 1992;52:3796-800.
22. Crowson AN, Magro CM. Antidepressant therapy. Arch Dermatol 1995;131:925-9.
23. Sternbach H. Are antidepressants carcinogenic? A review of preclinical and clinical studies. J Clin Psychiatry 2003;64:1153-62.
24. Xu W, Tamim H, Shapiro S, et al. Use of antidepressants and risk of colorectal cancer: a nested case-control study. Lancet Oncol 2006;7:301-8.
25. Brustolim D, Ribeiro-dos-Santos R, Kast RE, et al. A new chapter opens in anti-inflammatory treatments: the antidepressant bupropion lowers production of tumor necrosis factor-alpha and interferon-gamma in mice. Int Immunopharmacol 2006;6:903-7.
26. Lass-Flörl C, Dierich MP, Fuchs D, et al. Antifungal activity against Candida species of the selective serotonin-reuptake inhibitor, sertraline. Clin Infect Dis 2001;33:e135-6.
27. Waldinger MD, Hengeveld MW, Zsinderman AH, Olivier B. Effect of SSRI antidepressants on ejaculation: a double-blind, randomized, placebo-controlled study with fluoxetine, fluvoxamine, paroxetine, and sertraline. J Clin Psychopharmacol 1998;18(4):274-81.
28. Pryor JL, Althof SE, Steidle C, et al. Efficacy and tolerability of dapoxetine in treatment of premature ejaculation: an integrated analysis of two double-blind, randomised controlled trials. Lancet 2006;368:929-37.
29. Kumar VS, Sharma VL, Tiwari P, et al. The spermicidal and antitrichomonas activities of SSRI antidepressants. Bioorg Med Chem Lett 2006;16:2509-12.
1. Wernicke JF, Pritchett YL, D’Souza DN, et al. A randomized controlled trial of duloxetine in diabetic peripheral neuropathic pain. Neurology 2006;67(8):1411-20.
2. Raskin J, Pritchett YL, Wang F, et al. A double-blind, randomized multicenter trial comparing duloxetine with placebo in the management of diabetic peripheral neuropathic pain. Pain Med 2005;6(5):346-56.
3. Rowbotham MC, Goli V, Kunz NR, Lei D. Venlafaxine extended release in the treatment of painful diabetic neuropathy: a double-blind, placebo-controlled study. Pain 2004;110:697-706.
4. Max MB, Lynch SA, Muir J, et al. Effects of desipramine, amitriptyline, and fluoxetine on pain in diabetic neuropathy. N Engl J Med 1992;326:1250-6.
5. Sindrup SH, Otto M, Finnerup NB, Jensen TS. Antidepressants in the treatment of neuropathic pain. Basic Clin Pharmacol Toxicol 2005;96:399-409.
6. Semenchuk MR, Sherman S, Davis B. Double-blind, randomized trial of bupropion SR for the treatment of neuropathic pain. Neurology 2001;57:1583-8.
7. Tomkins GE, Jackson JL, O’Malley PG, et al. Treatment of chronic headache with antidepressants: a meta-analysis. Am J Med 2001;111:54-63.
8. Salerno SM, Browning R, Jackson JL. The effect of antidepressant treatment on chronic back pain. Arch Intern Med 2002;162:19-24.
9. Goldenberg DL, Burckhardt C, Crofford L. Management of fibromyalgia syndrome. JAMA 2004;292:2388-95.
10. Littlejohn GO, Guymer EK. Fibromyalgia syndrome: which antidepressant drug should we choose. Curr Pharm Des 2006;12(1):3-9.
11. 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;50:2974-84.
12. Arnold LM, Rosen A, Pritchett YL, et al. A randomized, double-blind, placebo-controlled trial of duloxetine in the treatment of women with fibromyalgia with or without major depressive disorder. Pain 2005;119:5-15.
13. Effects of encainide, flecainide, imipramine and moricizine on ventricular arrhythmias during the year after acute myocardial infarction: The CAPS. The Cardiac Arrhythmia Pilot Study (CAPS) Investigators. Am J Cardiol 1988;61(8):501-9.
14. Di Girolamo E, Di Iorio C, Sabatini P, et al. Effects of paroxetine hydrochloride, a selective serotonin reuptake inhibitor, on refractory vasovagal syncope: a randomized, double-blind, placebo-controlled study. J Am Coll Cardiol 1999;33:1227-30.
15. Jackson JL, O’Malley PG, Tomkins G, et al. Treatment of functional gastrointestinal disorders with antidepressant medications: a meta-analysis. Am J Med 2000;108:65-72.
16. Tabas G, Beaves M, Wang J, et al. Paroxetine to treat irritable bowel syndrome not responding to high-fiber diet: a double-blind, placebo-controlled trial. Am J Gastroenterol 2004;99(5):914-20.
17. Vahedi H, Merat S, Rashidioon A, et al. The effect of fluoxetine in patients with pain and constipation-predominant irritable bowel syndrome: a double-blind randomized-controlled study. Aliment Pharmacol Ther 2005;22:381-5.
18. Tack J, Broekaert D, Fischler B, et al. A controlled crossover study of the selective serotonin reuptake inhibitor citalopram in irritable bowel syndrome. Gut 2006;55:1095-103.
19. Talley NJ. Antidepressants in IBS: are we deluding ourselves? [editorial]. Am J Gastroenterol 2004;99:921-3.
20. Mariappan P, Ballantyne Z, N’Dow JM, Alhasso AA. Serotonin and noradrenaline reuptake inhibitors (SNRI) for stress urinary incontinence in adults. Cochrane Database Syst Rev 2005;Jul 20;(3):CD004742.-
21. Brandes LJ, Arron RJ, Bogdanovic RP, et al. Stimulation of malignant growth in rodents by antidepressant drugs at clinically relevant doses. Cancer Res 1992;52:3796-800.
22. Crowson AN, Magro CM. Antidepressant therapy. Arch Dermatol 1995;131:925-9.
23. Sternbach H. Are antidepressants carcinogenic? A review of preclinical and clinical studies. J Clin Psychiatry 2003;64:1153-62.
24. Xu W, Tamim H, Shapiro S, et al. Use of antidepressants and risk of colorectal cancer: a nested case-control study. Lancet Oncol 2006;7:301-8.
25. Brustolim D, Ribeiro-dos-Santos R, Kast RE, et al. A new chapter opens in anti-inflammatory treatments: the antidepressant bupropion lowers production of tumor necrosis factor-alpha and interferon-gamma in mice. Int Immunopharmacol 2006;6:903-7.
26. Lass-Flörl C, Dierich MP, Fuchs D, et al. Antifungal activity against Candida species of the selective serotonin-reuptake inhibitor, sertraline. Clin Infect Dis 2001;33:e135-6.
27. Waldinger MD, Hengeveld MW, Zsinderman AH, Olivier B. Effect of SSRI antidepressants on ejaculation: a double-blind, randomized, placebo-controlled study with fluoxetine, fluvoxamine, paroxetine, and sertraline. J Clin Psychopharmacol 1998;18(4):274-81.
28. Pryor JL, Althof SE, Steidle C, et al. Efficacy and tolerability of dapoxetine in treatment of premature ejaculation: an integrated analysis of two double-blind, randomised controlled trials. Lancet 2006;368:929-37.
29. Kumar VS, Sharma VL, Tiwari P, et al. The spermicidal and antitrichomonas activities of SSRI antidepressants. Bioorg Med Chem Lett 2006;16:2509-12.
Is this child bipolar? What’s needed to improve diagnosis
When does bipolar disorder begin? That question confounds clinicians, worries parents, and is leading researchers such as Kiki D. Chang, MD, to look for answers in families with this highly heritable disorder.
“Parents with bipolar disorder know what’s happening if their children have early symptoms,” Dr. Chang says. “They tell me, ‘I don’t want my child to go through what I went through, and he’s having the same symptoms I did.’”
Dr. Chang believes early psychotherapy and medication might prevent prodromal bipolar disorder from fully developing. His team at Stanford University is among those seeking genetic and brain imaging biomarkers to make a pediatric bipolar diagnosis more reliable. Lack of age-specific criteria may be causing overdiagnosis, as suggested by a 40-fold increase in 10 years in the number of children and adolescents being treated for bipolar disorder.1
In this interview by Robert A. Kowatch, MD, PhD, Dr. Chang describes a child with probable early signs of bipolar disorder and discusses why early intervention is both complicated and promising.
Children at risk for bipolarity
DR. KOWATCH: You’re studying children considered at high risk for developing bipolar disorder; why are these studies important?
DR. CHANG: High-risk children represent a chance to understand risk factors for developing bipolar disorder and what the early symptoms are. By “high risk,” we mean children and adolescents who possess a genetic predisposition toward bipolar disorder.
Bipolar disorder develops over time; a boy such as “Brian” (Box 1) likely would have gone 3 to 5 years on the stimulant—not doing well—until he had a manic episode at age 14 or 15. The full mood episode usually does not develop until later, with the right—or you could say wrong— combination of environment and stressors acting on a genetic predisposition.
DR. KOWATCH: Do the parents of the children you’re studying have bipolar disorder?
DR. CHANG: Yes; we’re studying what we call “bipolar offspring”—children with biological parents with bipolar disorder (Box 2).2-4 One also could look at siblings; having a brother or sister with bipolar disorder increases risk as well. If you search back in these families, usually you’ll find many relatives with bipolar disorder who reflect the child’s genetic predisposition.
Mrs. M, age 35, had early-onset depression but was not diagnosed with bipolar disorder until age 22. She requests a consultation for her 10-year-old son, Brian, whom she suspects also may have bipolar disorder. “I know there’s something going on; he’s just like I was, but no one would listen to me,” she says.
The boy’s pediatrician prescribed methylphenidate for “a little inattention” but felt that Brian was doing okay in school and had some friends. The stimulant might be helping, says Mrs. M, but she is not sure.
You talk to Brian and learn he has some anxiety. He sometimes gets very excited and runs around, and sometimes he does not sleep well. If you consider all the symptoms, this child has anxiety, attention-deficit/hyperactivity disorder, short depressive periods that affect his functioning, and a parent with bipolar disorder.
You ask further, and Brian tells you about hearing conversations and voices of old friends, his parents, and unknown people in his head, usually neutral, and not commanding or commentating. No one has asked him about parapsychotic phenomena, and he’s never reported this to anyone.
In adults, the incidence of bipolar types I and II is approximately 4%.1 Because two-thirds of adults with bipolar disorder have onset during childhood or adolescence, the incidence of pediatric bipolar disorder may be 1% to 2%. It could be as high as 3% if you include children with prodromes or early forms of the disorder.
The risk of a child developing a bipolar disorder is probably 15% to 20% when 1 biological parent—or sibling—has a bipolar disorder.2 If both parents have bipolar disorder, some older studies suggest that the child’s risk of developing at least a mood disorder would be up to 75%,3 and depression in a child might develop into a bipolar disorder.
Therefore, the risk of bipolar disorder developing in a child whose parents both have bipolar disorder may be >50% and could approach 75%.
‘Kindling’ in bipolar disorder
DR. KOWATCH: What have you seen in children whose parents have bipolar disorder?
DR. CHANG: We’ve tracked more than 200 bipolar offspring for up to 10 years. In some families we’ve seen the natural progression toward full mania and bipolar disorder.
We’ve also seen children who start to show symptoms but don’t develop full bipolar disorder. These children have had clinical treatment, so we’re not sure if the intervention prevented full bipolar disorder or if they would not have developed it anyway. Some children have developed mood symptoms and other psychiatric problems that have resolved with early intervention.
DR. CHANG: Kindling, which originally referred to seizure disorders, also has been applied to affective disorders.5 Early stressors and triggers appear to add up over time and combine with genetic predisposition to create a full mood episode. After that break, it becomes easier and easier to have the next episode, and the disorder becomes chronic and more difficult to treat.
The goal of our work is to stop kindling in bipolar disorder—to prevent environmental or developmental “sparks” from interacting with genetic predisposition and igniting a chronic, spontaneous course of mood episodes.
Brain imaging biomarkers
DR. KOWATCH: Are researchers finding biomarkers for bipolar disorder?
DR. CHANG: The field is young but light-years ahead of where we were 10 years ago. Brain imaging has revealed consistently abnormal areas in children with bipolar disorder. These abnormalities are seen in adults with bipolar disorder as well, but chronic illness, substance abuse, and medication exposure affect the findings in adults. Children have had less exposure to these confounding variables.
We and other groups have identified areas of the prefrontal cortex, amygdala, cerebellum, and striatum that could represent biomarkers, although I wouldn’t say yet that there are any markers per se. A decrease in amygdala volume has been found consistently in children with bipolar disorder, for example, but it’s not specific to bipolar disorder. So we have a way to go before we find specific biomarkers.
In the future, clinicians will probably use a set of 10 to 20 biomarkers, and the more biomarkers a patient has, the greater the risk for bipolar disorder. Once a battery of biomarkers has been put together, the more certain a bipolar disorder diagnosis will become.
Genetic biomarkers
DR. KOWATCH: We’ve talked about high-risk families; are there genetic markers for bipolar disorder?
If you look at common polymorphisms in a set of genes, eventually you’ll be able to calculate the risk that a person will develop bipolar disorder. We’re also investigating whether genes control the age of onset.
DR. KOWATCH: How are you looking for genetic markers in the high-risk children you’re studying?
DR. CHANG: We start with the proband—the child of a bipolar parent—and then study as much of the family as we can. Approximately 50% of the probands’ first- or second-degree relatives have a mood disorder—so our samples are highly loaded.
We’re interested in the interaction between genes and brain function and structure: How do genetic predispositions lead to brain differences that create vulnerability for mood disorders—in this case, bipolar disorder?
To explore that question, we’re starting a 5-year study funded by the National Institutes of Health (NIH). We’re recruiting 50 sibling pairs in which 1 child has early bipolar symptoms and the other is healthy. We will compare these pairs’ genetic and brain imaging profiles with those of 30 healthy children with no genetic risk for bipolar disorder, as far as we can tell.
Something makes 1 child develop bipolar disorder and another child not. By matching siblings with shared environments, we’re trying to eliminate environmental factors and look at their genetic and brain function differences. We’ll use functional brain imaging to look at how children respond to mood-related tasks and standard tasks involving facial emotion exposure to activate brain areas bipolar disorder is thought to affect.
Preventing bipolar ‘kindling’
DR. KOWATCH: What interventions might interrupt kindling and help prevent bipolar I disorder from developing in high risk children?
DR. CHANG: Families affected by bipolar disorder are characterized by stress and high expressed emotion; they tend to fight a lot, and we’re trying to improve communication and their ability to work together. We think reduced stress could halt the progression of the disorder in at-risk children.
Our group is collaborating with Dr. David Miklowitz at the University of Colorado to develop a family psychotherapy program for children who have at least 1 parent or sibling with bipolar disorder and are showing early bipolar symptoms. In a 3-year, NIH-funded treatment development study, 40 children will be randomly assigned to receive 12 sessions of weekly family-focused therapy (FFT) or treatment as usual.
We also think some medications have potential for protecting the brain against the progression of bipolar disorder. In vitro evidence exists for lithium, valproate, and carbamazepine to some extent, other anticonvulsants such as lamotrigine, and atypical antipsychotics such as quetiapine and olanzapine. A few preliminary clinical trials have been conducted (Box 3)9-11 but no longitudinal studies.
A 12-week, open-label study of valproate8 showed symptom improvement in 18 of 23 (78%) children ages 6 to 18 with mood or behavioral symptoms and at least 1 parent with bipolar disorder. On the other hand, a double-blind, controlled trial found no difference in mood symptom changes in 56 children receiving valproate or placebo for up to 5 years. Children in this study were ages 5 to 17, met DSM-IV-TR criteria for cyclothymia or bipolar disorder not otherwise specified, and had at least 1 biological parent with bipolar disorder.9
A small, open-label, 12-week prospective study suggested that quetiapine may be useful for treating mood symptoms in adolescents with at least 1 first-degree relative with bipolar disorder. The 20 adolescents (ages 12 to 18) had mood disorder diagnoses but did not meet DSM-IV-TR criteria for bipolar I disorder.10
RECOMMENDATIONS
DR. KOWATCH: What do you recommend that psychiatrists do to help children at risk for bipolar disorder?
DR. CHANG: Ask your adult patients with bipolar disorder how their children are doing. If a child is not doing well, consider referral to a child and adolescent psychiatrist or take an interest yourself and assess the child for early signs of bipolar disorder.
DR. KOWATCH: What are the prodromal symptoms in children and adolescents?
DR. CHANG: In the past, the earliest reported symptoms were thought to include extreme hyperactivity, inappropriate sexuality, and severe depression at a very young age (preschool or school age children). Now data point to 2 major pathways toward bipolar disorder:
- early-onset depression, which elevates risk for later mania
- early attention-deficit/hyperactivity disorder (ADHD).
DR. KOWATCH: So you’ve got a group with depression and a group with severe ADHD that might develop into bipolar disorder?
DR. CHANG: The ADHD need not be severe. In these children, ADHD may reflect an underlying brain development trajectory toward mood dysregulation. We’ve also seen anxiety as an initial condition. A cross-sectional study found anxiety to be prevalent in bipolar offspring and a possible risk factor for later mania.12
Anxiety is very common in children, so it’s hard to tell if it’s a precursor for bipolar disorder in an individual child. But looking back, a lot of children who develop bipolar disorder had early anxiety, which may be a marker that they were not coping well with stress. What starts leaking out as anxiety eventually may leak out as a full mood episode.
DR. CHANG: Yes, although sometimes the risk comes not from the parents but from a second-degree or more distant relative. We have seen plenty of families in which (as far as we can tell) the parents don’t have any mood disorders, but a child has full bipolar disorder that began over time—as it usually does in bipolar offspring.
Children or adolescents who have first-break episodes after very little pre-morbid dysfunction comprise yet another subset. This group tends to present with episodic manic depression.
DR. KOWATCH: Do you think children with bipolar disorder have clear mood episodes?
DR. CHANG: Our research is trying to bypass that debate. We’re trying to understand whether biomarkers in the brain or blood can be used to distinguish different types of bipolar disorders, rather than relying on symptomatology.
Related resources
- Chang KD, Howe M, Gallelli, K, Miklowitz D. Prevention of pediatric bipolar disorder: integration of neurobiological and psychosocial processes. Ann NY Acad Sci 2006;1094:235–47.
- Chang KD, Gallelli KA. Bipolar disorders and genetics: clinical implications of high heritability. Medscape Psychiatry & Mental Health 2004;9(2). Available at: http://www.medscape.com/viewarticle/489331.
- Miklowitz D, Biuckians A, Richards JA. Early-onset bipolar disorder: a family treatment perspective. Dev Psychopathol 2006;18(4):1247–65.
- Carbamazepine • various
- Lamotrigine • Lamictal
- Lithium • Eskalith, Lithobid
- Methylphenidate • Ritalin
- Olanzapine • Zyprexa
- Quetiapine • Seroquel
- Valproate • Depakene, Depakote
Dr. Chang receives research support from AstraZeneca, Eli Lilly and Company, Otsuka, and the National Institute of Mental Health. He is a consultant to Abbott Laboratories, GlaxoSmithKline, and Shire, and a speaker for Abbott Laboratories and AstraZeneca.
Dr. Kowatch receives research support from Bristol-Myers Squibb, Stanley Research Foundation, National Institute of Mental Health, and National Institute of Child Health and Human Development. He is a speaker for Abbott Laboratories and AstraZeneca.
1. Moreno C, Laje G, Blanco C, et al. National trends in the outpatient diagnosis and treatment of bipolar disorder in youth. Arch Gen Psychiatry 2007;64(9):1032-9.
2. Kessler RC, Berglund P, Demler O, et al. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry 2005;62(6):593-602.
3. Chang KD, Adleman N, Dienes K, et al. Bipolar offspring: a window into bipolar disorder evolution. Biol Psychiatry 2003;53:941-5.
4. Gershon ES, Hamovit J, Guroff JJ, et al. A family study of schizoaffective, bipolar I, bipolar II, unipolar, and normal control probands. Arch Gen Psychiatry 1982;39(10):1157-67.
5. Post RM. Do the epilepsies, pain syndromes, and affective disorders share common kindling-like mechanisms? Epilepsy Res 2002;50:203-19.
6. Caspi A, Sugden K, Moffitt TE, et al. Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science 2003;18;301(5631):291-3.
7. Green EK, Raybould R, Macgregor S, et al. Genetic variation of brain-derived neurotrophic factor (BDNF) in bipolar disorder: case-control study of over 3000 individuals from the UK. Br J Psychiatry 2006;188:21-5.
8. Burdick KE, Funke B, Goldberg JF, et al. COMT genotype increases risk for bipolar I disorder and influences neurocognitive performance. Bipolar Disord 2007;9(4):370-6.
9. Chang KD, Dienes K, Blasey C, et al. Divalproex monotherapy in the treatment of bipolar offspring with mood and behavioral disorders and at least mild affective symptoms. J Clin Psychiatry 2003;64(8):936-42.
10. Findling RL, Frazier TW, Youngstrom EA, et al. Double-blind, placebo-controlled trial of divalproex monotherapy in the treatment of symptomatic youth at high risk for developing bipolar disorder. J Clin Psychiatry 2007;68(5):781-8.
11. DelBello MP, Adler CM, Whitsel RM, et al. A 12-week single-blind trial of quetiapine for the treatment of mood symptoms in adolescents at high risk for developing bipolar I disorder. J Clin Psychiatry 2007;68(5):789-95.
12. Henin A, Biederman J, Mick E, et al. Psychopathology in the offspring of parents with bipolar disorder: a controlled study. Biol Psychiatry 2005;58(7):554-61.
When does bipolar disorder begin? That question confounds clinicians, worries parents, and is leading researchers such as Kiki D. Chang, MD, to look for answers in families with this highly heritable disorder.
“Parents with bipolar disorder know what’s happening if their children have early symptoms,” Dr. Chang says. “They tell me, ‘I don’t want my child to go through what I went through, and he’s having the same symptoms I did.’”
Dr. Chang believes early psychotherapy and medication might prevent prodromal bipolar disorder from fully developing. His team at Stanford University is among those seeking genetic and brain imaging biomarkers to make a pediatric bipolar diagnosis more reliable. Lack of age-specific criteria may be causing overdiagnosis, as suggested by a 40-fold increase in 10 years in the number of children and adolescents being treated for bipolar disorder.1
In this interview by Robert A. Kowatch, MD, PhD, Dr. Chang describes a child with probable early signs of bipolar disorder and discusses why early intervention is both complicated and promising.
Children at risk for bipolarity
DR. KOWATCH: You’re studying children considered at high risk for developing bipolar disorder; why are these studies important?
DR. CHANG: High-risk children represent a chance to understand risk factors for developing bipolar disorder and what the early symptoms are. By “high risk,” we mean children and adolescents who possess a genetic predisposition toward bipolar disorder.
Bipolar disorder develops over time; a boy such as “Brian” (Box 1) likely would have gone 3 to 5 years on the stimulant—not doing well—until he had a manic episode at age 14 or 15. The full mood episode usually does not develop until later, with the right—or you could say wrong— combination of environment and stressors acting on a genetic predisposition.
DR. KOWATCH: Do the parents of the children you’re studying have bipolar disorder?
DR. CHANG: Yes; we’re studying what we call “bipolar offspring”—children with biological parents with bipolar disorder (Box 2).2-4 One also could look at siblings; having a brother or sister with bipolar disorder increases risk as well. If you search back in these families, usually you’ll find many relatives with bipolar disorder who reflect the child’s genetic predisposition.
Mrs. M, age 35, had early-onset depression but was not diagnosed with bipolar disorder until age 22. She requests a consultation for her 10-year-old son, Brian, whom she suspects also may have bipolar disorder. “I know there’s something going on; he’s just like I was, but no one would listen to me,” she says.
The boy’s pediatrician prescribed methylphenidate for “a little inattention” but felt that Brian was doing okay in school and had some friends. The stimulant might be helping, says Mrs. M, but she is not sure.
You talk to Brian and learn he has some anxiety. He sometimes gets very excited and runs around, and sometimes he does not sleep well. If you consider all the symptoms, this child has anxiety, attention-deficit/hyperactivity disorder, short depressive periods that affect his functioning, and a parent with bipolar disorder.
You ask further, and Brian tells you about hearing conversations and voices of old friends, his parents, and unknown people in his head, usually neutral, and not commanding or commentating. No one has asked him about parapsychotic phenomena, and he’s never reported this to anyone.
In adults, the incidence of bipolar types I and II is approximately 4%.1 Because two-thirds of adults with bipolar disorder have onset during childhood or adolescence, the incidence of pediatric bipolar disorder may be 1% to 2%. It could be as high as 3% if you include children with prodromes or early forms of the disorder.
The risk of a child developing a bipolar disorder is probably 15% to 20% when 1 biological parent—or sibling—has a bipolar disorder.2 If both parents have bipolar disorder, some older studies suggest that the child’s risk of developing at least a mood disorder would be up to 75%,3 and depression in a child might develop into a bipolar disorder.
Therefore, the risk of bipolar disorder developing in a child whose parents both have bipolar disorder may be >50% and could approach 75%.
‘Kindling’ in bipolar disorder
DR. KOWATCH: What have you seen in children whose parents have bipolar disorder?
DR. CHANG: We’ve tracked more than 200 bipolar offspring for up to 10 years. In some families we’ve seen the natural progression toward full mania and bipolar disorder.
We’ve also seen children who start to show symptoms but don’t develop full bipolar disorder. These children have had clinical treatment, so we’re not sure if the intervention prevented full bipolar disorder or if they would not have developed it anyway. Some children have developed mood symptoms and other psychiatric problems that have resolved with early intervention.
DR. CHANG: Kindling, which originally referred to seizure disorders, also has been applied to affective disorders.5 Early stressors and triggers appear to add up over time and combine with genetic predisposition to create a full mood episode. After that break, it becomes easier and easier to have the next episode, and the disorder becomes chronic and more difficult to treat.
The goal of our work is to stop kindling in bipolar disorder—to prevent environmental or developmental “sparks” from interacting with genetic predisposition and igniting a chronic, spontaneous course of mood episodes.
Brain imaging biomarkers
DR. KOWATCH: Are researchers finding biomarkers for bipolar disorder?
DR. CHANG: The field is young but light-years ahead of where we were 10 years ago. Brain imaging has revealed consistently abnormal areas in children with bipolar disorder. These abnormalities are seen in adults with bipolar disorder as well, but chronic illness, substance abuse, and medication exposure affect the findings in adults. Children have had less exposure to these confounding variables.
We and other groups have identified areas of the prefrontal cortex, amygdala, cerebellum, and striatum that could represent biomarkers, although I wouldn’t say yet that there are any markers per se. A decrease in amygdala volume has been found consistently in children with bipolar disorder, for example, but it’s not specific to bipolar disorder. So we have a way to go before we find specific biomarkers.
In the future, clinicians will probably use a set of 10 to 20 biomarkers, and the more biomarkers a patient has, the greater the risk for bipolar disorder. Once a battery of biomarkers has been put together, the more certain a bipolar disorder diagnosis will become.
Genetic biomarkers
DR. KOWATCH: We’ve talked about high-risk families; are there genetic markers for bipolar disorder?
If you look at common polymorphisms in a set of genes, eventually you’ll be able to calculate the risk that a person will develop bipolar disorder. We’re also investigating whether genes control the age of onset.
DR. KOWATCH: How are you looking for genetic markers in the high-risk children you’re studying?
DR. CHANG: We start with the proband—the child of a bipolar parent—and then study as much of the family as we can. Approximately 50% of the probands’ first- or second-degree relatives have a mood disorder—so our samples are highly loaded.
We’re interested in the interaction between genes and brain function and structure: How do genetic predispositions lead to brain differences that create vulnerability for mood disorders—in this case, bipolar disorder?
To explore that question, we’re starting a 5-year study funded by the National Institutes of Health (NIH). We’re recruiting 50 sibling pairs in which 1 child has early bipolar symptoms and the other is healthy. We will compare these pairs’ genetic and brain imaging profiles with those of 30 healthy children with no genetic risk for bipolar disorder, as far as we can tell.
Something makes 1 child develop bipolar disorder and another child not. By matching siblings with shared environments, we’re trying to eliminate environmental factors and look at their genetic and brain function differences. We’ll use functional brain imaging to look at how children respond to mood-related tasks and standard tasks involving facial emotion exposure to activate brain areas bipolar disorder is thought to affect.
Preventing bipolar ‘kindling’
DR. KOWATCH: What interventions might interrupt kindling and help prevent bipolar I disorder from developing in high risk children?
DR. CHANG: Families affected by bipolar disorder are characterized by stress and high expressed emotion; they tend to fight a lot, and we’re trying to improve communication and their ability to work together. We think reduced stress could halt the progression of the disorder in at-risk children.
Our group is collaborating with Dr. David Miklowitz at the University of Colorado to develop a family psychotherapy program for children who have at least 1 parent or sibling with bipolar disorder and are showing early bipolar symptoms. In a 3-year, NIH-funded treatment development study, 40 children will be randomly assigned to receive 12 sessions of weekly family-focused therapy (FFT) or treatment as usual.
We also think some medications have potential for protecting the brain against the progression of bipolar disorder. In vitro evidence exists for lithium, valproate, and carbamazepine to some extent, other anticonvulsants such as lamotrigine, and atypical antipsychotics such as quetiapine and olanzapine. A few preliminary clinical trials have been conducted (Box 3)9-11 but no longitudinal studies.
A 12-week, open-label study of valproate8 showed symptom improvement in 18 of 23 (78%) children ages 6 to 18 with mood or behavioral symptoms and at least 1 parent with bipolar disorder. On the other hand, a double-blind, controlled trial found no difference in mood symptom changes in 56 children receiving valproate or placebo for up to 5 years. Children in this study were ages 5 to 17, met DSM-IV-TR criteria for cyclothymia or bipolar disorder not otherwise specified, and had at least 1 biological parent with bipolar disorder.9
A small, open-label, 12-week prospective study suggested that quetiapine may be useful for treating mood symptoms in adolescents with at least 1 first-degree relative with bipolar disorder. The 20 adolescents (ages 12 to 18) had mood disorder diagnoses but did not meet DSM-IV-TR criteria for bipolar I disorder.10
RECOMMENDATIONS
DR. KOWATCH: What do you recommend that psychiatrists do to help children at risk for bipolar disorder?
DR. CHANG: Ask your adult patients with bipolar disorder how their children are doing. If a child is not doing well, consider referral to a child and adolescent psychiatrist or take an interest yourself and assess the child for early signs of bipolar disorder.
DR. KOWATCH: What are the prodromal symptoms in children and adolescents?
DR. CHANG: In the past, the earliest reported symptoms were thought to include extreme hyperactivity, inappropriate sexuality, and severe depression at a very young age (preschool or school age children). Now data point to 2 major pathways toward bipolar disorder:
- early-onset depression, which elevates risk for later mania
- early attention-deficit/hyperactivity disorder (ADHD).
DR. KOWATCH: So you’ve got a group with depression and a group with severe ADHD that might develop into bipolar disorder?
DR. CHANG: The ADHD need not be severe. In these children, ADHD may reflect an underlying brain development trajectory toward mood dysregulation. We’ve also seen anxiety as an initial condition. A cross-sectional study found anxiety to be prevalent in bipolar offspring and a possible risk factor for later mania.12
Anxiety is very common in children, so it’s hard to tell if it’s a precursor for bipolar disorder in an individual child. But looking back, a lot of children who develop bipolar disorder had early anxiety, which may be a marker that they were not coping well with stress. What starts leaking out as anxiety eventually may leak out as a full mood episode.
DR. CHANG: Yes, although sometimes the risk comes not from the parents but from a second-degree or more distant relative. We have seen plenty of families in which (as far as we can tell) the parents don’t have any mood disorders, but a child has full bipolar disorder that began over time—as it usually does in bipolar offspring.
Children or adolescents who have first-break episodes after very little pre-morbid dysfunction comprise yet another subset. This group tends to present with episodic manic depression.
DR. KOWATCH: Do you think children with bipolar disorder have clear mood episodes?
DR. CHANG: Our research is trying to bypass that debate. We’re trying to understand whether biomarkers in the brain or blood can be used to distinguish different types of bipolar disorders, rather than relying on symptomatology.
Related resources
- Chang KD, Howe M, Gallelli, K, Miklowitz D. Prevention of pediatric bipolar disorder: integration of neurobiological and psychosocial processes. Ann NY Acad Sci 2006;1094:235–47.
- Chang KD, Gallelli KA. Bipolar disorders and genetics: clinical implications of high heritability. Medscape Psychiatry & Mental Health 2004;9(2). Available at: http://www.medscape.com/viewarticle/489331.
- Miklowitz D, Biuckians A, Richards JA. Early-onset bipolar disorder: a family treatment perspective. Dev Psychopathol 2006;18(4):1247–65.
- Carbamazepine • various
- Lamotrigine • Lamictal
- Lithium • Eskalith, Lithobid
- Methylphenidate • Ritalin
- Olanzapine • Zyprexa
- Quetiapine • Seroquel
- Valproate • Depakene, Depakote
Dr. Chang receives research support from AstraZeneca, Eli Lilly and Company, Otsuka, and the National Institute of Mental Health. He is a consultant to Abbott Laboratories, GlaxoSmithKline, and Shire, and a speaker for Abbott Laboratories and AstraZeneca.
Dr. Kowatch receives research support from Bristol-Myers Squibb, Stanley Research Foundation, National Institute of Mental Health, and National Institute of Child Health and Human Development. He is a speaker for Abbott Laboratories and AstraZeneca.
When does bipolar disorder begin? That question confounds clinicians, worries parents, and is leading researchers such as Kiki D. Chang, MD, to look for answers in families with this highly heritable disorder.
“Parents with bipolar disorder know what’s happening if their children have early symptoms,” Dr. Chang says. “They tell me, ‘I don’t want my child to go through what I went through, and he’s having the same symptoms I did.’”
Dr. Chang believes early psychotherapy and medication might prevent prodromal bipolar disorder from fully developing. His team at Stanford University is among those seeking genetic and brain imaging biomarkers to make a pediatric bipolar diagnosis more reliable. Lack of age-specific criteria may be causing overdiagnosis, as suggested by a 40-fold increase in 10 years in the number of children and adolescents being treated for bipolar disorder.1
In this interview by Robert A. Kowatch, MD, PhD, Dr. Chang describes a child with probable early signs of bipolar disorder and discusses why early intervention is both complicated and promising.
Children at risk for bipolarity
DR. KOWATCH: You’re studying children considered at high risk for developing bipolar disorder; why are these studies important?
DR. CHANG: High-risk children represent a chance to understand risk factors for developing bipolar disorder and what the early symptoms are. By “high risk,” we mean children and adolescents who possess a genetic predisposition toward bipolar disorder.
Bipolar disorder develops over time; a boy such as “Brian” (Box 1) likely would have gone 3 to 5 years on the stimulant—not doing well—until he had a manic episode at age 14 or 15. The full mood episode usually does not develop until later, with the right—or you could say wrong— combination of environment and stressors acting on a genetic predisposition.
DR. KOWATCH: Do the parents of the children you’re studying have bipolar disorder?
DR. CHANG: Yes; we’re studying what we call “bipolar offspring”—children with biological parents with bipolar disorder (Box 2).2-4 One also could look at siblings; having a brother or sister with bipolar disorder increases risk as well. If you search back in these families, usually you’ll find many relatives with bipolar disorder who reflect the child’s genetic predisposition.
Mrs. M, age 35, had early-onset depression but was not diagnosed with bipolar disorder until age 22. She requests a consultation for her 10-year-old son, Brian, whom she suspects also may have bipolar disorder. “I know there’s something going on; he’s just like I was, but no one would listen to me,” she says.
The boy’s pediatrician prescribed methylphenidate for “a little inattention” but felt that Brian was doing okay in school and had some friends. The stimulant might be helping, says Mrs. M, but she is not sure.
You talk to Brian and learn he has some anxiety. He sometimes gets very excited and runs around, and sometimes he does not sleep well. If you consider all the symptoms, this child has anxiety, attention-deficit/hyperactivity disorder, short depressive periods that affect his functioning, and a parent with bipolar disorder.
You ask further, and Brian tells you about hearing conversations and voices of old friends, his parents, and unknown people in his head, usually neutral, and not commanding or commentating. No one has asked him about parapsychotic phenomena, and he’s never reported this to anyone.
In adults, the incidence of bipolar types I and II is approximately 4%.1 Because two-thirds of adults with bipolar disorder have onset during childhood or adolescence, the incidence of pediatric bipolar disorder may be 1% to 2%. It could be as high as 3% if you include children with prodromes or early forms of the disorder.
The risk of a child developing a bipolar disorder is probably 15% to 20% when 1 biological parent—or sibling—has a bipolar disorder.2 If both parents have bipolar disorder, some older studies suggest that the child’s risk of developing at least a mood disorder would be up to 75%,3 and depression in a child might develop into a bipolar disorder.
Therefore, the risk of bipolar disorder developing in a child whose parents both have bipolar disorder may be >50% and could approach 75%.
‘Kindling’ in bipolar disorder
DR. KOWATCH: What have you seen in children whose parents have bipolar disorder?
DR. CHANG: We’ve tracked more than 200 bipolar offspring for up to 10 years. In some families we’ve seen the natural progression toward full mania and bipolar disorder.
We’ve also seen children who start to show symptoms but don’t develop full bipolar disorder. These children have had clinical treatment, so we’re not sure if the intervention prevented full bipolar disorder or if they would not have developed it anyway. Some children have developed mood symptoms and other psychiatric problems that have resolved with early intervention.
DR. CHANG: Kindling, which originally referred to seizure disorders, also has been applied to affective disorders.5 Early stressors and triggers appear to add up over time and combine with genetic predisposition to create a full mood episode. After that break, it becomes easier and easier to have the next episode, and the disorder becomes chronic and more difficult to treat.
The goal of our work is to stop kindling in bipolar disorder—to prevent environmental or developmental “sparks” from interacting with genetic predisposition and igniting a chronic, spontaneous course of mood episodes.
Brain imaging biomarkers
DR. KOWATCH: Are researchers finding biomarkers for bipolar disorder?
DR. CHANG: The field is young but light-years ahead of where we were 10 years ago. Brain imaging has revealed consistently abnormal areas in children with bipolar disorder. These abnormalities are seen in adults with bipolar disorder as well, but chronic illness, substance abuse, and medication exposure affect the findings in adults. Children have had less exposure to these confounding variables.
We and other groups have identified areas of the prefrontal cortex, amygdala, cerebellum, and striatum that could represent biomarkers, although I wouldn’t say yet that there are any markers per se. A decrease in amygdala volume has been found consistently in children with bipolar disorder, for example, but it’s not specific to bipolar disorder. So we have a way to go before we find specific biomarkers.
In the future, clinicians will probably use a set of 10 to 20 biomarkers, and the more biomarkers a patient has, the greater the risk for bipolar disorder. Once a battery of biomarkers has been put together, the more certain a bipolar disorder diagnosis will become.
Genetic biomarkers
DR. KOWATCH: We’ve talked about high-risk families; are there genetic markers for bipolar disorder?
If you look at common polymorphisms in a set of genes, eventually you’ll be able to calculate the risk that a person will develop bipolar disorder. We’re also investigating whether genes control the age of onset.
DR. KOWATCH: How are you looking for genetic markers in the high-risk children you’re studying?
DR. CHANG: We start with the proband—the child of a bipolar parent—and then study as much of the family as we can. Approximately 50% of the probands’ first- or second-degree relatives have a mood disorder—so our samples are highly loaded.
We’re interested in the interaction between genes and brain function and structure: How do genetic predispositions lead to brain differences that create vulnerability for mood disorders—in this case, bipolar disorder?
To explore that question, we’re starting a 5-year study funded by the National Institutes of Health (NIH). We’re recruiting 50 sibling pairs in which 1 child has early bipolar symptoms and the other is healthy. We will compare these pairs’ genetic and brain imaging profiles with those of 30 healthy children with no genetic risk for bipolar disorder, as far as we can tell.
Something makes 1 child develop bipolar disorder and another child not. By matching siblings with shared environments, we’re trying to eliminate environmental factors and look at their genetic and brain function differences. We’ll use functional brain imaging to look at how children respond to mood-related tasks and standard tasks involving facial emotion exposure to activate brain areas bipolar disorder is thought to affect.
Preventing bipolar ‘kindling’
DR. KOWATCH: What interventions might interrupt kindling and help prevent bipolar I disorder from developing in high risk children?
DR. CHANG: Families affected by bipolar disorder are characterized by stress and high expressed emotion; they tend to fight a lot, and we’re trying to improve communication and their ability to work together. We think reduced stress could halt the progression of the disorder in at-risk children.
Our group is collaborating with Dr. David Miklowitz at the University of Colorado to develop a family psychotherapy program for children who have at least 1 parent or sibling with bipolar disorder and are showing early bipolar symptoms. In a 3-year, NIH-funded treatment development study, 40 children will be randomly assigned to receive 12 sessions of weekly family-focused therapy (FFT) or treatment as usual.
We also think some medications have potential for protecting the brain against the progression of bipolar disorder. In vitro evidence exists for lithium, valproate, and carbamazepine to some extent, other anticonvulsants such as lamotrigine, and atypical antipsychotics such as quetiapine and olanzapine. A few preliminary clinical trials have been conducted (Box 3)9-11 but no longitudinal studies.
A 12-week, open-label study of valproate8 showed symptom improvement in 18 of 23 (78%) children ages 6 to 18 with mood or behavioral symptoms and at least 1 parent with bipolar disorder. On the other hand, a double-blind, controlled trial found no difference in mood symptom changes in 56 children receiving valproate or placebo for up to 5 years. Children in this study were ages 5 to 17, met DSM-IV-TR criteria for cyclothymia or bipolar disorder not otherwise specified, and had at least 1 biological parent with bipolar disorder.9
A small, open-label, 12-week prospective study suggested that quetiapine may be useful for treating mood symptoms in adolescents with at least 1 first-degree relative with bipolar disorder. The 20 adolescents (ages 12 to 18) had mood disorder diagnoses but did not meet DSM-IV-TR criteria for bipolar I disorder.10
RECOMMENDATIONS
DR. KOWATCH: What do you recommend that psychiatrists do to help children at risk for bipolar disorder?
DR. CHANG: Ask your adult patients with bipolar disorder how their children are doing. If a child is not doing well, consider referral to a child and adolescent psychiatrist or take an interest yourself and assess the child for early signs of bipolar disorder.
DR. KOWATCH: What are the prodromal symptoms in children and adolescents?
DR. CHANG: In the past, the earliest reported symptoms were thought to include extreme hyperactivity, inappropriate sexuality, and severe depression at a very young age (preschool or school age children). Now data point to 2 major pathways toward bipolar disorder:
- early-onset depression, which elevates risk for later mania
- early attention-deficit/hyperactivity disorder (ADHD).
DR. KOWATCH: So you’ve got a group with depression and a group with severe ADHD that might develop into bipolar disorder?
DR. CHANG: The ADHD need not be severe. In these children, ADHD may reflect an underlying brain development trajectory toward mood dysregulation. We’ve also seen anxiety as an initial condition. A cross-sectional study found anxiety to be prevalent in bipolar offspring and a possible risk factor for later mania.12
Anxiety is very common in children, so it’s hard to tell if it’s a precursor for bipolar disorder in an individual child. But looking back, a lot of children who develop bipolar disorder had early anxiety, which may be a marker that they were not coping well with stress. What starts leaking out as anxiety eventually may leak out as a full mood episode.
DR. CHANG: Yes, although sometimes the risk comes not from the parents but from a second-degree or more distant relative. We have seen plenty of families in which (as far as we can tell) the parents don’t have any mood disorders, but a child has full bipolar disorder that began over time—as it usually does in bipolar offspring.
Children or adolescents who have first-break episodes after very little pre-morbid dysfunction comprise yet another subset. This group tends to present with episodic manic depression.
DR. KOWATCH: Do you think children with bipolar disorder have clear mood episodes?
DR. CHANG: Our research is trying to bypass that debate. We’re trying to understand whether biomarkers in the brain or blood can be used to distinguish different types of bipolar disorders, rather than relying on symptomatology.
Related resources
- Chang KD, Howe M, Gallelli, K, Miklowitz D. Prevention of pediatric bipolar disorder: integration of neurobiological and psychosocial processes. Ann NY Acad Sci 2006;1094:235–47.
- Chang KD, Gallelli KA. Bipolar disorders and genetics: clinical implications of high heritability. Medscape Psychiatry & Mental Health 2004;9(2). Available at: http://www.medscape.com/viewarticle/489331.
- Miklowitz D, Biuckians A, Richards JA. Early-onset bipolar disorder: a family treatment perspective. Dev Psychopathol 2006;18(4):1247–65.
- Carbamazepine • various
- Lamotrigine • Lamictal
- Lithium • Eskalith, Lithobid
- Methylphenidate • Ritalin
- Olanzapine • Zyprexa
- Quetiapine • Seroquel
- Valproate • Depakene, Depakote
Dr. Chang receives research support from AstraZeneca, Eli Lilly and Company, Otsuka, and the National Institute of Mental Health. He is a consultant to Abbott Laboratories, GlaxoSmithKline, and Shire, and a speaker for Abbott Laboratories and AstraZeneca.
Dr. Kowatch receives research support from Bristol-Myers Squibb, Stanley Research Foundation, National Institute of Mental Health, and National Institute of Child Health and Human Development. He is a speaker for Abbott Laboratories and AstraZeneca.
1. Moreno C, Laje G, Blanco C, et al. National trends in the outpatient diagnosis and treatment of bipolar disorder in youth. Arch Gen Psychiatry 2007;64(9):1032-9.
2. Kessler RC, Berglund P, Demler O, et al. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry 2005;62(6):593-602.
3. Chang KD, Adleman N, Dienes K, et al. Bipolar offspring: a window into bipolar disorder evolution. Biol Psychiatry 2003;53:941-5.
4. Gershon ES, Hamovit J, Guroff JJ, et al. A family study of schizoaffective, bipolar I, bipolar II, unipolar, and normal control probands. Arch Gen Psychiatry 1982;39(10):1157-67.
5. Post RM. Do the epilepsies, pain syndromes, and affective disorders share common kindling-like mechanisms? Epilepsy Res 2002;50:203-19.
6. Caspi A, Sugden K, Moffitt TE, et al. Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science 2003;18;301(5631):291-3.
7. Green EK, Raybould R, Macgregor S, et al. Genetic variation of brain-derived neurotrophic factor (BDNF) in bipolar disorder: case-control study of over 3000 individuals from the UK. Br J Psychiatry 2006;188:21-5.
8. Burdick KE, Funke B, Goldberg JF, et al. COMT genotype increases risk for bipolar I disorder and influences neurocognitive performance. Bipolar Disord 2007;9(4):370-6.
9. Chang KD, Dienes K, Blasey C, et al. Divalproex monotherapy in the treatment of bipolar offspring with mood and behavioral disorders and at least mild affective symptoms. J Clin Psychiatry 2003;64(8):936-42.
10. Findling RL, Frazier TW, Youngstrom EA, et al. Double-blind, placebo-controlled trial of divalproex monotherapy in the treatment of symptomatic youth at high risk for developing bipolar disorder. J Clin Psychiatry 2007;68(5):781-8.
11. DelBello MP, Adler CM, Whitsel RM, et al. A 12-week single-blind trial of quetiapine for the treatment of mood symptoms in adolescents at high risk for developing bipolar I disorder. J Clin Psychiatry 2007;68(5):789-95.
12. Henin A, Biederman J, Mick E, et al. Psychopathology in the offspring of parents with bipolar disorder: a controlled study. Biol Psychiatry 2005;58(7):554-61.
1. Moreno C, Laje G, Blanco C, et al. National trends in the outpatient diagnosis and treatment of bipolar disorder in youth. Arch Gen Psychiatry 2007;64(9):1032-9.
2. Kessler RC, Berglund P, Demler O, et al. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry 2005;62(6):593-602.
3. Chang KD, Adleman N, Dienes K, et al. Bipolar offspring: a window into bipolar disorder evolution. Biol Psychiatry 2003;53:941-5.
4. Gershon ES, Hamovit J, Guroff JJ, et al. A family study of schizoaffective, bipolar I, bipolar II, unipolar, and normal control probands. Arch Gen Psychiatry 1982;39(10):1157-67.
5. Post RM. Do the epilepsies, pain syndromes, and affective disorders share common kindling-like mechanisms? Epilepsy Res 2002;50:203-19.
6. Caspi A, Sugden K, Moffitt TE, et al. Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science 2003;18;301(5631):291-3.
7. Green EK, Raybould R, Macgregor S, et al. Genetic variation of brain-derived neurotrophic factor (BDNF) in bipolar disorder: case-control study of over 3000 individuals from the UK. Br J Psychiatry 2006;188:21-5.
8. Burdick KE, Funke B, Goldberg JF, et al. COMT genotype increases risk for bipolar I disorder and influences neurocognitive performance. Bipolar Disord 2007;9(4):370-6.
9. Chang KD, Dienes K, Blasey C, et al. Divalproex monotherapy in the treatment of bipolar offspring with mood and behavioral disorders and at least mild affective symptoms. J Clin Psychiatry 2003;64(8):936-42.
10. Findling RL, Frazier TW, Youngstrom EA, et al. Double-blind, placebo-controlled trial of divalproex monotherapy in the treatment of symptomatic youth at high risk for developing bipolar disorder. J Clin Psychiatry 2007;68(5):781-8.
11. DelBello MP, Adler CM, Whitsel RM, et al. A 12-week single-blind trial of quetiapine for the treatment of mood symptoms in adolescents at high risk for developing bipolar I disorder. J Clin Psychiatry 2007;68(5):789-95.
12. Henin A, Biederman J, Mick E, et al. Psychopathology in the offspring of parents with bipolar disorder: a controlled study. Biol Psychiatry 2005;58(7):554-61.
Misinformation on herbals?
Although it is always important to determine what other therapies a patient is taking, Dr. Joseph I. Sirven (“Dangerous duo: Antiepileptics plus herbals,” Pearls, Current Psychiatry, July 2007) betrays a lack of knowledge about botanical medicine. No herbalist prescribes water hemlock, which is not available at health food stores and has been known to be a deadly poison since the time of Socrates. Yohimbine is a drug, not a botanical, although it is derived from the yohimbe plant. St. John’s wort—which was not discussed in the article—induces the cytochrome system, especially the 3A enzymes and the multidrug resistance transporter Pglycoprotein, and should not be taken with antiepileptic drugs (AEDs).
Antispasmodic herbs such as black cohosh and kava are unlikely to induce seizures, although they might potentiate AEDs and could help to lower dosages of these medications. Ginkgo seeds are a popular food in China and Korea because cooking inactivates toxins in the seeds. Guarana acts like caffeine and may cause vasoconstriction. Ephedra, which is only available in low doses or from Chinese herbalists who use small quantities for short-term respiratory illness, has a similar effect and could possibly interact with AEDs.
I urge clinicians to obtain information about these remedies from professionals specializing in the medicinal use of herbs to improve the care of patients who use complementary and alternative medicine.
Karen S. Vaughan
Brooklyn, NY
Dr. Sirven Responds
The reader and I both believe in delivering accurate information about the use of herbals and botanicals. Although botanicals carry many promises for the treatment of neurologic and psychiatric disease, there are also a number of risks—which was the rationale for publishing a “Pearls” article on this topic.
My experience more than qualifies me to comment on herbal use. I am a practicing, epilepsy fellowship-trained, board-certified neurologist who teaches, publishes, and conducts research on antiepileptic drugs and surveys ideas on complementary and alternative medicine (CAM) use in patients with seizures and epilepsy. Apparently the reader’s passion for the topic led to several inaccuracies and mischaracterizations about the “Pearls” article. Contrary to the reader’s claims, there were no errors in the information presented.
My practice at a tertiary care epilepsy center places me in the unfortunate position of seeing serious and even fatal outcomes that result from poor clinical care with botanicals. I agree that the clinical use of botanicals should be supervised by professionals who are trained in oriental medicine and are registered herbalists, but in collaboration with neurologists, psychiatrists, and other practitioners who are charged with caring for patients with serious and refractory epilepsy and psychiatric conditions—patients likely to be taking antiepileptics.
Because the use of antiepileptic drugs and herbals has increased significantly, it is important for clinicians to understand all interactions between these 2 classes of agents. I am compelled to ensure that other practitioners are aware of potential drug interactions so that we can improve the care of patients likely to use CAM.
The last paragraph of my article notes that many patients do not tell their neurologists or other healthcare practitioners about their CAM use, and herein lies the problem. CAMs are readily available, and patients will take botanicals and herbals without professional guidance. Thus, a respectful team approach is necessary for medicine to achieve good health outcomes for all.
Joseph I. Sirven, MD
Associate professor of neurology
Mayo Clinic College of Medicine
Phoenix, AZ
To comment on articles in this issue or other topics, send letters in care of Erica Vonderheid, Current Psychiatry, 110 Summit Avenue, Montvale, NJ 07645, [email protected].
Although it is always important to determine what other therapies a patient is taking, Dr. Joseph I. Sirven (“Dangerous duo: Antiepileptics plus herbals,” Pearls, Current Psychiatry, July 2007) betrays a lack of knowledge about botanical medicine. No herbalist prescribes water hemlock, which is not available at health food stores and has been known to be a deadly poison since the time of Socrates. Yohimbine is a drug, not a botanical, although it is derived from the yohimbe plant. St. John’s wort—which was not discussed in the article—induces the cytochrome system, especially the 3A enzymes and the multidrug resistance transporter Pglycoprotein, and should not be taken with antiepileptic drugs (AEDs).
Antispasmodic herbs such as black cohosh and kava are unlikely to induce seizures, although they might potentiate AEDs and could help to lower dosages of these medications. Ginkgo seeds are a popular food in China and Korea because cooking inactivates toxins in the seeds. Guarana acts like caffeine and may cause vasoconstriction. Ephedra, which is only available in low doses or from Chinese herbalists who use small quantities for short-term respiratory illness, has a similar effect and could possibly interact with AEDs.
I urge clinicians to obtain information about these remedies from professionals specializing in the medicinal use of herbs to improve the care of patients who use complementary and alternative medicine.
Karen S. Vaughan
Brooklyn, NY
Dr. Sirven Responds
The reader and I both believe in delivering accurate information about the use of herbals and botanicals. Although botanicals carry many promises for the treatment of neurologic and psychiatric disease, there are also a number of risks—which was the rationale for publishing a “Pearls” article on this topic.
My experience more than qualifies me to comment on herbal use. I am a practicing, epilepsy fellowship-trained, board-certified neurologist who teaches, publishes, and conducts research on antiepileptic drugs and surveys ideas on complementary and alternative medicine (CAM) use in patients with seizures and epilepsy. Apparently the reader’s passion for the topic led to several inaccuracies and mischaracterizations about the “Pearls” article. Contrary to the reader’s claims, there were no errors in the information presented.
My practice at a tertiary care epilepsy center places me in the unfortunate position of seeing serious and even fatal outcomes that result from poor clinical care with botanicals. I agree that the clinical use of botanicals should be supervised by professionals who are trained in oriental medicine and are registered herbalists, but in collaboration with neurologists, psychiatrists, and other practitioners who are charged with caring for patients with serious and refractory epilepsy and psychiatric conditions—patients likely to be taking antiepileptics.
Because the use of antiepileptic drugs and herbals has increased significantly, it is important for clinicians to understand all interactions between these 2 classes of agents. I am compelled to ensure that other practitioners are aware of potential drug interactions so that we can improve the care of patients likely to use CAM.
The last paragraph of my article notes that many patients do not tell their neurologists or other healthcare practitioners about their CAM use, and herein lies the problem. CAMs are readily available, and patients will take botanicals and herbals without professional guidance. Thus, a respectful team approach is necessary for medicine to achieve good health outcomes for all.
Joseph I. Sirven, MD
Associate professor of neurology
Mayo Clinic College of Medicine
Phoenix, AZ
Although it is always important to determine what other therapies a patient is taking, Dr. Joseph I. Sirven (“Dangerous duo: Antiepileptics plus herbals,” Pearls, Current Psychiatry, July 2007) betrays a lack of knowledge about botanical medicine. No herbalist prescribes water hemlock, which is not available at health food stores and has been known to be a deadly poison since the time of Socrates. Yohimbine is a drug, not a botanical, although it is derived from the yohimbe plant. St. John’s wort—which was not discussed in the article—induces the cytochrome system, especially the 3A enzymes and the multidrug resistance transporter Pglycoprotein, and should not be taken with antiepileptic drugs (AEDs).
Antispasmodic herbs such as black cohosh and kava are unlikely to induce seizures, although they might potentiate AEDs and could help to lower dosages of these medications. Ginkgo seeds are a popular food in China and Korea because cooking inactivates toxins in the seeds. Guarana acts like caffeine and may cause vasoconstriction. Ephedra, which is only available in low doses or from Chinese herbalists who use small quantities for short-term respiratory illness, has a similar effect and could possibly interact with AEDs.
I urge clinicians to obtain information about these remedies from professionals specializing in the medicinal use of herbs to improve the care of patients who use complementary and alternative medicine.
Karen S. Vaughan
Brooklyn, NY
Dr. Sirven Responds
The reader and I both believe in delivering accurate information about the use of herbals and botanicals. Although botanicals carry many promises for the treatment of neurologic and psychiatric disease, there are also a number of risks—which was the rationale for publishing a “Pearls” article on this topic.
My experience more than qualifies me to comment on herbal use. I am a practicing, epilepsy fellowship-trained, board-certified neurologist who teaches, publishes, and conducts research on antiepileptic drugs and surveys ideas on complementary and alternative medicine (CAM) use in patients with seizures and epilepsy. Apparently the reader’s passion for the topic led to several inaccuracies and mischaracterizations about the “Pearls” article. Contrary to the reader’s claims, there were no errors in the information presented.
My practice at a tertiary care epilepsy center places me in the unfortunate position of seeing serious and even fatal outcomes that result from poor clinical care with botanicals. I agree that the clinical use of botanicals should be supervised by professionals who are trained in oriental medicine and are registered herbalists, but in collaboration with neurologists, psychiatrists, and other practitioners who are charged with caring for patients with serious and refractory epilepsy and psychiatric conditions—patients likely to be taking antiepileptics.
Because the use of antiepileptic drugs and herbals has increased significantly, it is important for clinicians to understand all interactions between these 2 classes of agents. I am compelled to ensure that other practitioners are aware of potential drug interactions so that we can improve the care of patients likely to use CAM.
The last paragraph of my article notes that many patients do not tell their neurologists or other healthcare practitioners about their CAM use, and herein lies the problem. CAMs are readily available, and patients will take botanicals and herbals without professional guidance. Thus, a respectful team approach is necessary for medicine to achieve good health outcomes for all.
Joseph I. Sirven, MD
Associate professor of neurology
Mayo Clinic College of Medicine
Phoenix, AZ
To comment on articles in this issue or other topics, send letters in care of Erica Vonderheid, Current Psychiatry, 110 Summit Avenue, Montvale, NJ 07645, [email protected].
To comment on articles in this issue or other topics, send letters in care of Erica Vonderheid, Current Psychiatry, 110 Summit Avenue, Montvale, NJ 07645, [email protected].
Sleep hygiene helps patients catch some ZZZs
Proper sleep hygiene can help your patients fall and stay asleep consistently. Patients with insomnia are at a higher risk of developing or experiencing a recurrence of a mood disorder, and poor sleep can worsen psychiatric symptoms such as depression or mania.1 Data about combining behavioral approaches and hypnotic medications to treat insomnia are inconclusive;2 however, using the 2 together may help patients who do not respond to a single approach.
First rule out other causes of insomnia, such as sleep apnea, other medical conditions, or medications. Patients may improve after these factors are addressed.
Teaching sleep hygiene principles (Box) does not mean patients will adopt these habits, but employing the following suggestions could improve adherence:
Obtain a detailed sleep history to identify specific behaviors to be changed. For example, a patient might only have to stop watching television in bed to get a good night’s sleep, although some may find a brief exposure to television or radio facilitates relaxation.
Explain the rationale for changing a behavior. For example, when telling patients to limit caffeine or alcohol at night, list these substances’ negative effects on sleep. Similarly, when instructing patients to avoid watching television in bed, tell them that using the bedroom only for sleep or sex will help condition them for sleep at bedtime.
- Establish a regular sleep-wake schedule
- Limit caffeine and alcohol consumption
- Avoid naps
- Eliminate noise and light from the sleep environment
- Use the bed only for sleep or sex
- Avoid looking at a clock when trying to sleep
Discuss sleep regularly. A patient might not disclose poor sleeping habits during the first session.
Give your patient handouts on sleep hygiene principles and highlight the most pertinent information. Ask the patient to place the handout where he or she will see it regularly.
Involve the family to help identify a patient’s poor sleep habits and find ways to implement sleep hygiene principles.
Encourage patients to keep a sleep diary. Ask the patient to note how many hours and at what time he or she slept for at least 2 weeks, then bring this information to the next appointment. This record allows you to examine patients’ sleep patterns and recommend appropriate changes.
Ask patients for creative ideas to improve their sleep. This dialogue will facilitate the therapeutic alliance and encourage positive changes in patients’ lives.
1. Peterson MJ, Benca RM. Sleep in mood disorders. Psychiatr Clin North Am 2006;29:1009-32.
2. Mendelson WB. Combining pharmacological and non-pharmacological therapies for insomnia. J Clin Psychiatry 2007;68(suppl 5):19-23.
Dr. Khawaja is staff psychiatrist, VA Medical Center, Minneapolis, MN; Dr. Hurwitz is a psychiatrist and sleep medicine physician, VA Medical Center, Minneapolis, MN; Dr. Ebrahim is an endocrinologist, Minnesota Center for Obesity, Metabolism, and Endocrinology, Eagan, MN.
Proper sleep hygiene can help your patients fall and stay asleep consistently. Patients with insomnia are at a higher risk of developing or experiencing a recurrence of a mood disorder, and poor sleep can worsen psychiatric symptoms such as depression or mania.1 Data about combining behavioral approaches and hypnotic medications to treat insomnia are inconclusive;2 however, using the 2 together may help patients who do not respond to a single approach.
First rule out other causes of insomnia, such as sleep apnea, other medical conditions, or medications. Patients may improve after these factors are addressed.
Teaching sleep hygiene principles (Box) does not mean patients will adopt these habits, but employing the following suggestions could improve adherence:
Obtain a detailed sleep history to identify specific behaviors to be changed. For example, a patient might only have to stop watching television in bed to get a good night’s sleep, although some may find a brief exposure to television or radio facilitates relaxation.
Explain the rationale for changing a behavior. For example, when telling patients to limit caffeine or alcohol at night, list these substances’ negative effects on sleep. Similarly, when instructing patients to avoid watching television in bed, tell them that using the bedroom only for sleep or sex will help condition them for sleep at bedtime.
- Establish a regular sleep-wake schedule
- Limit caffeine and alcohol consumption
- Avoid naps
- Eliminate noise and light from the sleep environment
- Use the bed only for sleep or sex
- Avoid looking at a clock when trying to sleep
Discuss sleep regularly. A patient might not disclose poor sleeping habits during the first session.
Give your patient handouts on sleep hygiene principles and highlight the most pertinent information. Ask the patient to place the handout where he or she will see it regularly.
Involve the family to help identify a patient’s poor sleep habits and find ways to implement sleep hygiene principles.
Encourage patients to keep a sleep diary. Ask the patient to note how many hours and at what time he or she slept for at least 2 weeks, then bring this information to the next appointment. This record allows you to examine patients’ sleep patterns and recommend appropriate changes.
Ask patients for creative ideas to improve their sleep. This dialogue will facilitate the therapeutic alliance and encourage positive changes in patients’ lives.
Proper sleep hygiene can help your patients fall and stay asleep consistently. Patients with insomnia are at a higher risk of developing or experiencing a recurrence of a mood disorder, and poor sleep can worsen psychiatric symptoms such as depression or mania.1 Data about combining behavioral approaches and hypnotic medications to treat insomnia are inconclusive;2 however, using the 2 together may help patients who do not respond to a single approach.
First rule out other causes of insomnia, such as sleep apnea, other medical conditions, or medications. Patients may improve after these factors are addressed.
Teaching sleep hygiene principles (Box) does not mean patients will adopt these habits, but employing the following suggestions could improve adherence:
Obtain a detailed sleep history to identify specific behaviors to be changed. For example, a patient might only have to stop watching television in bed to get a good night’s sleep, although some may find a brief exposure to television or radio facilitates relaxation.
Explain the rationale for changing a behavior. For example, when telling patients to limit caffeine or alcohol at night, list these substances’ negative effects on sleep. Similarly, when instructing patients to avoid watching television in bed, tell them that using the bedroom only for sleep or sex will help condition them for sleep at bedtime.
- Establish a regular sleep-wake schedule
- Limit caffeine and alcohol consumption
- Avoid naps
- Eliminate noise and light from the sleep environment
- Use the bed only for sleep or sex
- Avoid looking at a clock when trying to sleep
Discuss sleep regularly. A patient might not disclose poor sleeping habits during the first session.
Give your patient handouts on sleep hygiene principles and highlight the most pertinent information. Ask the patient to place the handout where he or she will see it regularly.
Involve the family to help identify a patient’s poor sleep habits and find ways to implement sleep hygiene principles.
Encourage patients to keep a sleep diary. Ask the patient to note how many hours and at what time he or she slept for at least 2 weeks, then bring this information to the next appointment. This record allows you to examine patients’ sleep patterns and recommend appropriate changes.
Ask patients for creative ideas to improve their sleep. This dialogue will facilitate the therapeutic alliance and encourage positive changes in patients’ lives.
1. Peterson MJ, Benca RM. Sleep in mood disorders. Psychiatr Clin North Am 2006;29:1009-32.
2. Mendelson WB. Combining pharmacological and non-pharmacological therapies for insomnia. J Clin Psychiatry 2007;68(suppl 5):19-23.
Dr. Khawaja is staff psychiatrist, VA Medical Center, Minneapolis, MN; Dr. Hurwitz is a psychiatrist and sleep medicine physician, VA Medical Center, Minneapolis, MN; Dr. Ebrahim is an endocrinologist, Minnesota Center for Obesity, Metabolism, and Endocrinology, Eagan, MN.
1. Peterson MJ, Benca RM. Sleep in mood disorders. Psychiatr Clin North Am 2006;29:1009-32.
2. Mendelson WB. Combining pharmacological and non-pharmacological therapies for insomnia. J Clin Psychiatry 2007;68(suppl 5):19-23.
Dr. Khawaja is staff psychiatrist, VA Medical Center, Minneapolis, MN; Dr. Hurwitz is a psychiatrist and sleep medicine physician, VA Medical Center, Minneapolis, MN; Dr. Ebrahim is an endocrinologist, Minnesota Center for Obesity, Metabolism, and Endocrinology, Eagan, MN.
Tools, techniques to assess organ transplant candidates
With nearly 30,000 organ transplants being performed in the United States each year (Box 1),1 demand is growing for psychiatrists to provide presurgical and ongoing care.
How you might collaborate with a transplant team depends on each medical center’s protocols and individual patients’ mental health needs. A transplant candidate with depressive or anxiety symptoms may be referred to you for presurgical stabilization, for example, particularly if the patient lives far from a highly specialized transplant center.
Transplant assessments differ from usual psychiatric evaluations. Your findings will be used to help the transplant team evaluate the patient’s demographics, disease severity, and resources to give the patient the best chance for medical recovery. Inform patients at the beginning of the pretransplant evaluation that the results:
- will be shared with the transplant team
- may be used to help make decisions about transplant
- will not be the only factor determining if a transplant center will place a patient on an organ wait list.2
Pretransplant evaluation
Presurgical assessment helps determine the patient’s understanding of the transplant process and ability to provide consent (Table 1).3 Patients do not need a high level of medical sophistication to discuss transplantation, but they must understand the basics of the procedure and be able to rationally discuss their options. If a patient has severe cognitive impairment, dementia, or hepatic encephalopathy and cannot participate in the consent process, a surrogate is necessary.
Explore the patient’s attitudes and beliefs about transplant. If other team members have educated the patient about the procedure, your assessment can help determine how much the patient understood and if the patient has the capacity to make treatment decisions. Some patients believe the operation will “cure” them, despite education about the rigorous posttransplant routine. Alert the transplant team to these views, and begin aligning the patient’s views with reality.
In 2006, U.S. surgeons performed 28,931 organ transplants, bringing the total number of transplants since 1988 to >400,000. Each year, more kidney transplants are performed (17,091 in 2006) than all other organ transplants combined, according to the nonprofit United Network of Organ Sharing.1
Other organs being transplanted include liver, pancreas, heart, lung, and intestine. Some patients receive multiple organs, such as kidney/pancreas or heart/lung. As this article went to press, >96,000 candidates were on wait lists for organ donations.
Survival after transplantation has improved because of better immunosuppressant therapies introduced in the early 1980s and evolving physician and institutional experience. One-year survival rates for single-organ transplants range from 85% for lung to 98% for living donor kidney. Five-year survival rates range from 47% for lung to 86% for living donor kidney.
Source: Reference 1
Table 1
Psychiatric assessment of the pretransplant patient
| Assess understanding of his or her illness |
| Assess understanding of transplant process and ability to provide informed consent |
| Assess history of compliance with medical and psychiatric treatments |
| Identify substance abuse and other psychiatric comorbidities |
| Assess mental status |
| Evaluate social support system and possible interventions to bolster supports |
| Provide transplant team with information about patient’s need for education and support |
| Recommend treatment plan to address substance abuse and other psychiatric comorbidities |
| Source: Adapted from reference 3 |
Assessing psychiatric comorbidity. Like other patients with life-threatening medical illnesses, many transplant patients present with major depression and anxiety. Screen for symptoms of mood and anxiety disorders and past episodes of depression or mania. Explore the patient’s response to psychiatric treatment, current therapies, and history of treatment adherence.
Depression. Patients listed for transplant are seriously ill and coping with the difficulties of the sick role. Organ failure symptoms and resultant disability—such as insomnia, anorexia, fatigue, and impaired concentration—overlap with depression’s neurovegetative signs. Suspect depression if a patient presents with anhedonia, tearfulness, apathy, or guilt.
Among heart, lung, and liver transplant candidates, the reported lifetime prevalence of depression averages approximately 20%.4-6
Anxiety disorders. An estimated 40% of transplant patients have anxiety disorders,7 which may be caused by:
- stress of chronic illness
- uncertainty of the transplant process
- medical conditions such as hypothyroidism or pulmonary embolism.
Chronic mental illness. Patients with major mental illnesses such as schizophrenia might be appropriate candidates for organ transplant if they have adequate social support and history of treatment compliance.
Pharmacotherapy. Because of the variety of medical problems seen in transplant candidates, carefully consider medication side effects and drug-drug interactions when prescribing psychotropics.
Antidepressants. Among the selective serotonin reuptake inhibitors (SSRIs), citalopram, escitalopram, and sertraline are least likely to affect hepatic metabolism of other medications (Table 2).8 If a patient presents with liver failure, reduce the dosages of medications with hepatic metabolism.
Benzodiazepines. Use caution when treating anxiety with benzodiazepines because of the risk of tolerance, withdrawal, and dependence. Avoid benzodiazepines when treating transplant candidates with a substance abuse history. Also, these drugs might worsen hepatic encephalopathy and increase confusion.
Patients awaiting lung transplantation, especially those with high levels of CO2 retention, require special care because benzodiazepines might decrease respiratory drive. Try other agents such as buspirone, gabapentin, SSRIs, or second-generation antipsychotics to treat their anxiety.
Psychotherapy. Supportive psychotherapy can help patients navigate the often-lengthy process of waiting for a donor organ. Support groups for organ transplant candidates may help ease patients’ depressive symptoms.
Table 2
Antidepressants’ half-life and effect on hepatic metabolism
| Hepatic enzyme alterations | Half-life (hours) | |
|---|---|---|
| SSRIs | ||
| Fluoxetine | 2D6, 2C9, 2C19, 3A4 inhibition | 72 |
| Citalopram | None | 35 |
| Escitalopram | 2D6 inhibition (weak) | 32 |
| Sertraline | 2D6 inhibition (weak) | 30 |
| Paroxetine | 2D6 inhibition (strong) | 18 |
| Fluvoxamine | 1A2, 2C19, 2C9, 3A4 inhibition | 18 |
| Others | ||
| Mirtazapine | None | 30 |
| Bupropion SR | 2D6 inhibition | 21 |
| Venlafaxine XR | 2D6 inhibition | 5 |
| Trazodone | None | 5 |
| SSRIs: selective serotonin reuptake inhibitors | ||
| Source: Reference 8 | ||
Assessing substance abuse
Up to 50% of liver transplant candidates have a history of alcohol and/or drug abuse,9 the highest rate among transplant populations. Alcohol-induced cirrhosis and hepatitis C contracted from IV drug use are common indications for liver transplant. Effective treatment of substance abuse is essential because 30% to 50% of these patients relapse after the procedure.10 Assess:
- each substance abused, including onset, peak, and current use
- family history of substance abuse disorders
- past efforts at rehabilitation
- tobacco use (smoking before and after transplant is related to an increased incidence of new cancer diagnoses).11
Some transplant centers require patients with substance
use disorders to participate in 12-step programs or
rehabilitation. Regardless of the institutions’
requirements, encourage patients to participate in
rehabilitation to prevent relapse and mitigate the
negative impact of substance abuse on physical
and mental well-being.
Mental status examination includes the usual elements such as appearance, behavior, speech, affect, and thought process. Assess for suicidal thinking or hopelessness, which have been linked to serious medical illness.12 Question patients about hallucinations and give special attention to visual aberrations, which may occur in medically ill patients.
Cognitive testing. Use tools such as the Mini-Mental State Examination, clock drawing test, and Trail Making A and B tests to assess cognitive ability. If patients show signs of cognitive impairment, arrange for follow-up examinations and refer for neuropsychological testing.
Some cognitive impairment—such as that caused by hepatic encephalopathy—will likely improve after transplant, but other types—such as that caused by vascular disease—will not. If confusion is caused by hepatic encephalopathy, treatment with lactulose might rapidly improve symptoms. Remember that patients with hepatic encephalopathy might not exhibit elevated ammonia levels. Underlying causes of worsening hepatic encephalopathy—such as infections or bleeding—might require treatment.
Assessing adherence. Medication adherence after transplant is essential to prevent organ rejection and other complications. Posttransplant regimens are complex, and the frequency of follow-up assessments can be intense—particularly in the first year after transplant.
Your pretransplant assessment can identify where patients have struggled with adherence in the past. Before the transplant, your team can work to correct barriers such as inability to pay for medications, child care problems, or transportation needs.
Personality disorders have been identified as predictors of posttransplant nonadherence, and 50% to 60% of transplant programs consider personality disorders a relative contraindication to organ transplant.13 Address other contributors to poor adherence—such as substance abuse or depression—with ongoing psychiatric care.
When assessing a patient’s social support, look for evidence of:
- stable living situations
- long-term relationships with spouses, parents, children, or close friends
- adequate financial resources, including health insurance.
These factors help the patient manage the posttransplant process and numerous follow-up physician visits. Religious organizations or other social institutions also appear to provide the emotional support patients need to cope with an organ transplant.
Social support is essential to help with the normal difficulties such as frequent clinic visits and initial physical disability patients face after successful transplant (Box 2). Ask about the candidate’s family, friends, spirituality, and finances during your pretransplant assessment. Poor social support is related to the development of posttransplant psychiatric disorders14 and adherence difficulties.15
Assessment instruments—such as the Psychosocial Assessment of Candidates for Transplantation and the Transplant Evaluation Rating Scale3—include social support items and can be useful in identifying weak areas.
Data collected by other team members can be invaluable. A nurse or social worker, for example, may observe that a patient is unwilling to take medications, contrary to the patient’s report. Other sources of information include the patient’s family and friends, a primary care physician, or other mental health providers such as a therapist or case manager.
Posttransplant psychiatric care
Depression. The incidence of depression is higher in the year following transplant than before transplant or in the immediate posttransplant period.5 Predictors of posttransplant depression include:
- history of depression
- poor social support
- passive coping strategies
- poor physical status after transplantation.16,17
Carefully monitor patients who present with these factors after transplant. Treat depression with supportive measures designed to improve the patient’s social network and coping skills and pharmacotherapy. Select antidepressant medications based on side effect profiles and impact on the patient’s transplanted organs.
Substance abuse. Patients with a pretransplant history of substance abuse often relapse. Among transplant recipients with a history of alcoholic liver disease, drinking rates of 30% to 40% have been reported 5 years after transplant. Most of these data represent occasional use, not heavy or regular drinking.18 Relapse can occur despite careful assessment and follow-up.
Some evidence suggests that transplant patients who resume drinking have worse outcomes than those who abstain. Alcoholism relapse has other negative consequences, such as relationship problems and employment difficulties.
Predictors of relapse include:
- pretransplant history of alcohol dependence
- family history of alcoholism
- rehabilitation history, which could indicate a severe substance abuse disorder.3
Medications for alcoholism treatment have not been studied systematically in transplant patients, but low doses of acamprosate, ≤2 g/d, and naltrexone, ≤200 mg/d, are options for patients interested in pharmacotherapy. Support from 12-step programs also helps treat substance-abusing patients.
Altered mental status. Immunosuppressive medications—including cyclosporine, tacrolimus, and prednisone—can have neuropsychiatric effects and could cause a change in mental status (Table 3).19 Check cyclosporine and tacrolimus serum levels against reference ranges when delirium is present. If levels are toxic the dosage often can be lowered, which might lead to clinical improvement.
Quality of life. In general, patients’ quality of life improves after their transplant. After the first year—which patients might find difficult because of changes in physical and social status—quality of life typically improves.5
Table 3
Neuropsychiatric side effects of medications
commonly used in transplant patients
| Medication | Side effects |
|---|---|
| Cyclosporine | Tremor, headache, seizures, hallucinations, delirium |
| Tacrolimus | Tremor, headache, vivid dreams, anxiety, anorexia, seizures, delirium |
| Prednisone | Depression, mania, psychosis, delirium |
| Source: Adapted from references 3,7 | |
Psychiatric disorders such as depression can worsen quality of life. However, quality of life can improve after depression is diagnosed and treated. Other predictors of improved quality of life include older age, marriage, and the absence of a personality disorder.4
Other posttransplant concerns of patients include changes in employment, finances, and relationships. Patients often have been away from work before transplant, and returning after a long absence can be stressful. Patients may find that they cannot work as well as before becoming ill, which may lead to frustration, depression, and/or anxiety symptoms. Transplant surgery requires a large financial investment, and money concerns usually persist long after the transplant.
The transplant recipient’s role within the family may shift after surgery. Families might expect the patient to “return to normal” and resume old activities. Alternatively, family members might continue to treat the patient as a person with chronic illness despite physical improvement. If patients are struggling with these changes, supportive psychotherapy is indicated.
Related resource
- United Network for Organ Sharing. www.unos.org.
- Transplant living. www.transplantliving.org.
- Trzepacz PT, DiMartini AF, eds. The transplant patient. Cambridge, UK: Cambridge University Press; 2000.
- Klapheke MM. The role of the psychiatrist in organ transplantation. Bull Menninger Clin 1999;63(1):13-39.
Drug brand names
- Acamprosate • Campral
- Buspirone • BuSpar
- Bupropion SR • Wellbutrin SR
- Citalopram • Celexa
- Cyclosporine • Sandimmune
- Escitalopram • Lexapro
- Fluoxetine • Prozac
- Fluvoxamine • Luvox
- Gabapentin • Neurontin
- Lactulose • Cephulac, Chronulac
- Mirtazapine • Remeron
- Naltrexone • ReVia
- Paroxetine • Paxil
- Prednisone • Deltasone
- Sertraline • Zoloft
- Tacrolimus • Prograf
- Trazodone • Desyrel
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. United Network for Organ Sharing. Transplants by donor type. U.S. transplants performed: January 1, 1988 – April 30, 2007. Organ Procurement and Transplantation Network. Available at: http://www.unos.org. Accessed July 26, 2007.
2. Crone CC, Wise TN. Psychiatric aspects of transplantation, I: evaluation and selection of candidates. Crit Care Nurs 1999;19:79-87.
3. DiMartini AF, Dew MA, Trzepacz PT. Organ transplantation. In: Levenson JL, ed. The American Psychiatric Publishing textbook of psychosomatic medicine. Washington DC: American Psychiatric Publishing, Inc, 2005:675-700.
4. Cupples S, Dew MA, Grady KL, et al. Report of the psychosocial outcomes workgroup of the nursing and social sciences council of the international society for heart and lung transplantation: present status of research on psychosocial outcomes in cardiothoracic transplantation: review and recommendations for the field. J Heart Lung Transplant 2006;25:716-25.
5. Dew MA, DiMartini AF. Psychological disorders and distress after adult cardiothoracic transplantation. J Cardiovasc Nurs 2005;20:S51-S66.
6. Barbour KA, Blumenthal JA, Palmer SM. Psychosocial issues in the assessment and management of patients undergoing lung transplantation. Chest 2006;129:1367-74.
7. Trzepacz PT, Levenson JL, Tringali RA. Psychopharmacology and neuropsychiatric syndromes in organ transplantation. Gen Hosp Psychiatry 1991;13:233-45.
8. Crone CC, Gabriel GM. Treatment of anxiety and depression in transplant patients. Clin Pharmacokinet 2004;43:361-94.
9. DiMartini A, Weinrieb R, Mireman M. Liver transplantation in patients with alcohol and other substance use disorders. Psychiatr Clin North Am 2002;25:195-209.
10. Weinrieb RM, Van Horn DHA, McLellan AT, et al. Alcoholism treatment after liver transplantation: lessons learned from a clinical trial that failed. Psychosomatics 2001;42:110-6.
11. Jimenez C, Manrique A, Marques E, et al. Incidence and risk factors for the development of lung tumors after liver transplantation. Transpl Int 2007;20:57-63.
12. Juurlink DN, Herrmann N, Szalai JP, et al. Medical illness and the risk of suicide in the elderly. Arch Intern Med 2004;14:1179-84.
13. Levenson JL, Olbrisch ME. Psychosocial screening and selection of candidates for organ transplantation. In: Trzepacz PT, DiMartini AF, eds. The transplant patient. Cambridge, UK: Cambridge University Press, 2000:21-41.
14. Dew MA, Kormos RL, DiMartini AF, et al. Prevalence and risk of depression and anxiety-related disorders during the first three years after heart transplantation. Psychosomatics 2001;42:300-13.
15. Dew MA, Roth LH, Thompson ME, et al. Medical compliance and its predictors in the first year after cardiac transplantation. J Heart Lung Transplant 1996;15:631-45.
16. Dew MA, Myaskovsky L, Switzer GE, et al. Profiles and predictors of the course of psychological distress across four years after heart transplantation. Psychol Med 2005;35:1215-27.
17. Goetzmann L, Klaghofer R, Wagner-Huber R, et al. Psychosocial vulnerability predicts psychosocial outcome after an organ transplant: results of a prospective study with lung, liver, and bone-marrow patients. J Psychosom Res 2007;62:93-100.
18. Lucey M. Liver transplantation for alcoholic liver disease: a progress report. Graft 1999;2:S73-9.
19. Beresford TP. Neuropsychiatric complications of liver and other solid organ transplantation. Liver Transpl 2001;7(11 suppl 1):S36-S45.
With nearly 30,000 organ transplants being performed in the United States each year (Box 1),1 demand is growing for psychiatrists to provide presurgical and ongoing care.
How you might collaborate with a transplant team depends on each medical center’s protocols and individual patients’ mental health needs. A transplant candidate with depressive or anxiety symptoms may be referred to you for presurgical stabilization, for example, particularly if the patient lives far from a highly specialized transplant center.
Transplant assessments differ from usual psychiatric evaluations. Your findings will be used to help the transplant team evaluate the patient’s demographics, disease severity, and resources to give the patient the best chance for medical recovery. Inform patients at the beginning of the pretransplant evaluation that the results:
- will be shared with the transplant team
- may be used to help make decisions about transplant
- will not be the only factor determining if a transplant center will place a patient on an organ wait list.2
Pretransplant evaluation
Presurgical assessment helps determine the patient’s understanding of the transplant process and ability to provide consent (Table 1).3 Patients do not need a high level of medical sophistication to discuss transplantation, but they must understand the basics of the procedure and be able to rationally discuss their options. If a patient has severe cognitive impairment, dementia, or hepatic encephalopathy and cannot participate in the consent process, a surrogate is necessary.
Explore the patient’s attitudes and beliefs about transplant. If other team members have educated the patient about the procedure, your assessment can help determine how much the patient understood and if the patient has the capacity to make treatment decisions. Some patients believe the operation will “cure” them, despite education about the rigorous posttransplant routine. Alert the transplant team to these views, and begin aligning the patient’s views with reality.
In 2006, U.S. surgeons performed 28,931 organ transplants, bringing the total number of transplants since 1988 to >400,000. Each year, more kidney transplants are performed (17,091 in 2006) than all other organ transplants combined, according to the nonprofit United Network of Organ Sharing.1
Other organs being transplanted include liver, pancreas, heart, lung, and intestine. Some patients receive multiple organs, such as kidney/pancreas or heart/lung. As this article went to press, >96,000 candidates were on wait lists for organ donations.
Survival after transplantation has improved because of better immunosuppressant therapies introduced in the early 1980s and evolving physician and institutional experience. One-year survival rates for single-organ transplants range from 85% for lung to 98% for living donor kidney. Five-year survival rates range from 47% for lung to 86% for living donor kidney.
Source: Reference 1
Table 1
Psychiatric assessment of the pretransplant patient
| Assess understanding of his or her illness |
| Assess understanding of transplant process and ability to provide informed consent |
| Assess history of compliance with medical and psychiatric treatments |
| Identify substance abuse and other psychiatric comorbidities |
| Assess mental status |
| Evaluate social support system and possible interventions to bolster supports |
| Provide transplant team with information about patient’s need for education and support |
| Recommend treatment plan to address substance abuse and other psychiatric comorbidities |
| Source: Adapted from reference 3 |
Assessing psychiatric comorbidity. Like other patients with life-threatening medical illnesses, many transplant patients present with major depression and anxiety. Screen for symptoms of mood and anxiety disorders and past episodes of depression or mania. Explore the patient’s response to psychiatric treatment, current therapies, and history of treatment adherence.
Depression. Patients listed for transplant are seriously ill and coping with the difficulties of the sick role. Organ failure symptoms and resultant disability—such as insomnia, anorexia, fatigue, and impaired concentration—overlap with depression’s neurovegetative signs. Suspect depression if a patient presents with anhedonia, tearfulness, apathy, or guilt.
Among heart, lung, and liver transplant candidates, the reported lifetime prevalence of depression averages approximately 20%.4-6
Anxiety disorders. An estimated 40% of transplant patients have anxiety disorders,7 which may be caused by:
- stress of chronic illness
- uncertainty of the transplant process
- medical conditions such as hypothyroidism or pulmonary embolism.
Chronic mental illness. Patients with major mental illnesses such as schizophrenia might be appropriate candidates for organ transplant if they have adequate social support and history of treatment compliance.
Pharmacotherapy. Because of the variety of medical problems seen in transplant candidates, carefully consider medication side effects and drug-drug interactions when prescribing psychotropics.
Antidepressants. Among the selective serotonin reuptake inhibitors (SSRIs), citalopram, escitalopram, and sertraline are least likely to affect hepatic metabolism of other medications (Table 2).8 If a patient presents with liver failure, reduce the dosages of medications with hepatic metabolism.
Benzodiazepines. Use caution when treating anxiety with benzodiazepines because of the risk of tolerance, withdrawal, and dependence. Avoid benzodiazepines when treating transplant candidates with a substance abuse history. Also, these drugs might worsen hepatic encephalopathy and increase confusion.
Patients awaiting lung transplantation, especially those with high levels of CO2 retention, require special care because benzodiazepines might decrease respiratory drive. Try other agents such as buspirone, gabapentin, SSRIs, or second-generation antipsychotics to treat their anxiety.
Psychotherapy. Supportive psychotherapy can help patients navigate the often-lengthy process of waiting for a donor organ. Support groups for organ transplant candidates may help ease patients’ depressive symptoms.
Table 2
Antidepressants’ half-life and effect on hepatic metabolism
| Hepatic enzyme alterations | Half-life (hours) | |
|---|---|---|
| SSRIs | ||
| Fluoxetine | 2D6, 2C9, 2C19, 3A4 inhibition | 72 |
| Citalopram | None | 35 |
| Escitalopram | 2D6 inhibition (weak) | 32 |
| Sertraline | 2D6 inhibition (weak) | 30 |
| Paroxetine | 2D6 inhibition (strong) | 18 |
| Fluvoxamine | 1A2, 2C19, 2C9, 3A4 inhibition | 18 |
| Others | ||
| Mirtazapine | None | 30 |
| Bupropion SR | 2D6 inhibition | 21 |
| Venlafaxine XR | 2D6 inhibition | 5 |
| Trazodone | None | 5 |
| SSRIs: selective serotonin reuptake inhibitors | ||
| Source: Reference 8 | ||
Assessing substance abuse
Up to 50% of liver transplant candidates have a history of alcohol and/or drug abuse,9 the highest rate among transplant populations. Alcohol-induced cirrhosis and hepatitis C contracted from IV drug use are common indications for liver transplant. Effective treatment of substance abuse is essential because 30% to 50% of these patients relapse after the procedure.10 Assess:
- each substance abused, including onset, peak, and current use
- family history of substance abuse disorders
- past efforts at rehabilitation
- tobacco use (smoking before and after transplant is related to an increased incidence of new cancer diagnoses).11
Some transplant centers require patients with substance
use disorders to participate in 12-step programs or
rehabilitation. Regardless of the institutions’
requirements, encourage patients to participate in
rehabilitation to prevent relapse and mitigate the
negative impact of substance abuse on physical
and mental well-being.
Mental status examination includes the usual elements such as appearance, behavior, speech, affect, and thought process. Assess for suicidal thinking or hopelessness, which have been linked to serious medical illness.12 Question patients about hallucinations and give special attention to visual aberrations, which may occur in medically ill patients.
Cognitive testing. Use tools such as the Mini-Mental State Examination, clock drawing test, and Trail Making A and B tests to assess cognitive ability. If patients show signs of cognitive impairment, arrange for follow-up examinations and refer for neuropsychological testing.
Some cognitive impairment—such as that caused by hepatic encephalopathy—will likely improve after transplant, but other types—such as that caused by vascular disease—will not. If confusion is caused by hepatic encephalopathy, treatment with lactulose might rapidly improve symptoms. Remember that patients with hepatic encephalopathy might not exhibit elevated ammonia levels. Underlying causes of worsening hepatic encephalopathy—such as infections or bleeding—might require treatment.
Assessing adherence. Medication adherence after transplant is essential to prevent organ rejection and other complications. Posttransplant regimens are complex, and the frequency of follow-up assessments can be intense—particularly in the first year after transplant.
Your pretransplant assessment can identify where patients have struggled with adherence in the past. Before the transplant, your team can work to correct barriers such as inability to pay for medications, child care problems, or transportation needs.
Personality disorders have been identified as predictors of posttransplant nonadherence, and 50% to 60% of transplant programs consider personality disorders a relative contraindication to organ transplant.13 Address other contributors to poor adherence—such as substance abuse or depression—with ongoing psychiatric care.
When assessing a patient’s social support, look for evidence of:
- stable living situations
- long-term relationships with spouses, parents, children, or close friends
- adequate financial resources, including health insurance.
These factors help the patient manage the posttransplant process and numerous follow-up physician visits. Religious organizations or other social institutions also appear to provide the emotional support patients need to cope with an organ transplant.
Social support is essential to help with the normal difficulties such as frequent clinic visits and initial physical disability patients face after successful transplant (Box 2). Ask about the candidate’s family, friends, spirituality, and finances during your pretransplant assessment. Poor social support is related to the development of posttransplant psychiatric disorders14 and adherence difficulties.15
Assessment instruments—such as the Psychosocial Assessment of Candidates for Transplantation and the Transplant Evaluation Rating Scale3—include social support items and can be useful in identifying weak areas.
Data collected by other team members can be invaluable. A nurse or social worker, for example, may observe that a patient is unwilling to take medications, contrary to the patient’s report. Other sources of information include the patient’s family and friends, a primary care physician, or other mental health providers such as a therapist or case manager.
Posttransplant psychiatric care
Depression. The incidence of depression is higher in the year following transplant than before transplant or in the immediate posttransplant period.5 Predictors of posttransplant depression include:
- history of depression
- poor social support
- passive coping strategies
- poor physical status after transplantation.16,17
Carefully monitor patients who present with these factors after transplant. Treat depression with supportive measures designed to improve the patient’s social network and coping skills and pharmacotherapy. Select antidepressant medications based on side effect profiles and impact on the patient’s transplanted organs.
Substance abuse. Patients with a pretransplant history of substance abuse often relapse. Among transplant recipients with a history of alcoholic liver disease, drinking rates of 30% to 40% have been reported 5 years after transplant. Most of these data represent occasional use, not heavy or regular drinking.18 Relapse can occur despite careful assessment and follow-up.
Some evidence suggests that transplant patients who resume drinking have worse outcomes than those who abstain. Alcoholism relapse has other negative consequences, such as relationship problems and employment difficulties.
Predictors of relapse include:
- pretransplant history of alcohol dependence
- family history of alcoholism
- rehabilitation history, which could indicate a severe substance abuse disorder.3
Medications for alcoholism treatment have not been studied systematically in transplant patients, but low doses of acamprosate, ≤2 g/d, and naltrexone, ≤200 mg/d, are options for patients interested in pharmacotherapy. Support from 12-step programs also helps treat substance-abusing patients.
Altered mental status. Immunosuppressive medications—including cyclosporine, tacrolimus, and prednisone—can have neuropsychiatric effects and could cause a change in mental status (Table 3).19 Check cyclosporine and tacrolimus serum levels against reference ranges when delirium is present. If levels are toxic the dosage often can be lowered, which might lead to clinical improvement.
Quality of life. In general, patients’ quality of life improves after their transplant. After the first year—which patients might find difficult because of changes in physical and social status—quality of life typically improves.5
Table 3
Neuropsychiatric side effects of medications
commonly used in transplant patients
| Medication | Side effects |
|---|---|
| Cyclosporine | Tremor, headache, seizures, hallucinations, delirium |
| Tacrolimus | Tremor, headache, vivid dreams, anxiety, anorexia, seizures, delirium |
| Prednisone | Depression, mania, psychosis, delirium |
| Source: Adapted from references 3,7 | |
Psychiatric disorders such as depression can worsen quality of life. However, quality of life can improve after depression is diagnosed and treated. Other predictors of improved quality of life include older age, marriage, and the absence of a personality disorder.4
Other posttransplant concerns of patients include changes in employment, finances, and relationships. Patients often have been away from work before transplant, and returning after a long absence can be stressful. Patients may find that they cannot work as well as before becoming ill, which may lead to frustration, depression, and/or anxiety symptoms. Transplant surgery requires a large financial investment, and money concerns usually persist long after the transplant.
The transplant recipient’s role within the family may shift after surgery. Families might expect the patient to “return to normal” and resume old activities. Alternatively, family members might continue to treat the patient as a person with chronic illness despite physical improvement. If patients are struggling with these changes, supportive psychotherapy is indicated.
Related resource
- United Network for Organ Sharing. www.unos.org.
- Transplant living. www.transplantliving.org.
- Trzepacz PT, DiMartini AF, eds. The transplant patient. Cambridge, UK: Cambridge University Press; 2000.
- Klapheke MM. The role of the psychiatrist in organ transplantation. Bull Menninger Clin 1999;63(1):13-39.
Drug brand names
- Acamprosate • Campral
- Buspirone • BuSpar
- Bupropion SR • Wellbutrin SR
- Citalopram • Celexa
- Cyclosporine • Sandimmune
- Escitalopram • Lexapro
- Fluoxetine • Prozac
- Fluvoxamine • Luvox
- Gabapentin • Neurontin
- Lactulose • Cephulac, Chronulac
- Mirtazapine • Remeron
- Naltrexone • ReVia
- Paroxetine • Paxil
- Prednisone • Deltasone
- Sertraline • Zoloft
- Tacrolimus • Prograf
- Trazodone • Desyrel
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
With nearly 30,000 organ transplants being performed in the United States each year (Box 1),1 demand is growing for psychiatrists to provide presurgical and ongoing care.
How you might collaborate with a transplant team depends on each medical center’s protocols and individual patients’ mental health needs. A transplant candidate with depressive or anxiety symptoms may be referred to you for presurgical stabilization, for example, particularly if the patient lives far from a highly specialized transplant center.
Transplant assessments differ from usual psychiatric evaluations. Your findings will be used to help the transplant team evaluate the patient’s demographics, disease severity, and resources to give the patient the best chance for medical recovery. Inform patients at the beginning of the pretransplant evaluation that the results:
- will be shared with the transplant team
- may be used to help make decisions about transplant
- will not be the only factor determining if a transplant center will place a patient on an organ wait list.2
Pretransplant evaluation
Presurgical assessment helps determine the patient’s understanding of the transplant process and ability to provide consent (Table 1).3 Patients do not need a high level of medical sophistication to discuss transplantation, but they must understand the basics of the procedure and be able to rationally discuss their options. If a patient has severe cognitive impairment, dementia, or hepatic encephalopathy and cannot participate in the consent process, a surrogate is necessary.
Explore the patient’s attitudes and beliefs about transplant. If other team members have educated the patient about the procedure, your assessment can help determine how much the patient understood and if the patient has the capacity to make treatment decisions. Some patients believe the operation will “cure” them, despite education about the rigorous posttransplant routine. Alert the transplant team to these views, and begin aligning the patient’s views with reality.
In 2006, U.S. surgeons performed 28,931 organ transplants, bringing the total number of transplants since 1988 to >400,000. Each year, more kidney transplants are performed (17,091 in 2006) than all other organ transplants combined, according to the nonprofit United Network of Organ Sharing.1
Other organs being transplanted include liver, pancreas, heart, lung, and intestine. Some patients receive multiple organs, such as kidney/pancreas or heart/lung. As this article went to press, >96,000 candidates were on wait lists for organ donations.
Survival after transplantation has improved because of better immunosuppressant therapies introduced in the early 1980s and evolving physician and institutional experience. One-year survival rates for single-organ transplants range from 85% for lung to 98% for living donor kidney. Five-year survival rates range from 47% for lung to 86% for living donor kidney.
Source: Reference 1
Table 1
Psychiatric assessment of the pretransplant patient
| Assess understanding of his or her illness |
| Assess understanding of transplant process and ability to provide informed consent |
| Assess history of compliance with medical and psychiatric treatments |
| Identify substance abuse and other psychiatric comorbidities |
| Assess mental status |
| Evaluate social support system and possible interventions to bolster supports |
| Provide transplant team with information about patient’s need for education and support |
| Recommend treatment plan to address substance abuse and other psychiatric comorbidities |
| Source: Adapted from reference 3 |
Assessing psychiatric comorbidity. Like other patients with life-threatening medical illnesses, many transplant patients present with major depression and anxiety. Screen for symptoms of mood and anxiety disorders and past episodes of depression or mania. Explore the patient’s response to psychiatric treatment, current therapies, and history of treatment adherence.
Depression. Patients listed for transplant are seriously ill and coping with the difficulties of the sick role. Organ failure symptoms and resultant disability—such as insomnia, anorexia, fatigue, and impaired concentration—overlap with depression’s neurovegetative signs. Suspect depression if a patient presents with anhedonia, tearfulness, apathy, or guilt.
Among heart, lung, and liver transplant candidates, the reported lifetime prevalence of depression averages approximately 20%.4-6
Anxiety disorders. An estimated 40% of transplant patients have anxiety disorders,7 which may be caused by:
- stress of chronic illness
- uncertainty of the transplant process
- medical conditions such as hypothyroidism or pulmonary embolism.
Chronic mental illness. Patients with major mental illnesses such as schizophrenia might be appropriate candidates for organ transplant if they have adequate social support and history of treatment compliance.
Pharmacotherapy. Because of the variety of medical problems seen in transplant candidates, carefully consider medication side effects and drug-drug interactions when prescribing psychotropics.
Antidepressants. Among the selective serotonin reuptake inhibitors (SSRIs), citalopram, escitalopram, and sertraline are least likely to affect hepatic metabolism of other medications (Table 2).8 If a patient presents with liver failure, reduce the dosages of medications with hepatic metabolism.
Benzodiazepines. Use caution when treating anxiety with benzodiazepines because of the risk of tolerance, withdrawal, and dependence. Avoid benzodiazepines when treating transplant candidates with a substance abuse history. Also, these drugs might worsen hepatic encephalopathy and increase confusion.
Patients awaiting lung transplantation, especially those with high levels of CO2 retention, require special care because benzodiazepines might decrease respiratory drive. Try other agents such as buspirone, gabapentin, SSRIs, or second-generation antipsychotics to treat their anxiety.
Psychotherapy. Supportive psychotherapy can help patients navigate the often-lengthy process of waiting for a donor organ. Support groups for organ transplant candidates may help ease patients’ depressive symptoms.
Table 2
Antidepressants’ half-life and effect on hepatic metabolism
| Hepatic enzyme alterations | Half-life (hours) | |
|---|---|---|
| SSRIs | ||
| Fluoxetine | 2D6, 2C9, 2C19, 3A4 inhibition | 72 |
| Citalopram | None | 35 |
| Escitalopram | 2D6 inhibition (weak) | 32 |
| Sertraline | 2D6 inhibition (weak) | 30 |
| Paroxetine | 2D6 inhibition (strong) | 18 |
| Fluvoxamine | 1A2, 2C19, 2C9, 3A4 inhibition | 18 |
| Others | ||
| Mirtazapine | None | 30 |
| Bupropion SR | 2D6 inhibition | 21 |
| Venlafaxine XR | 2D6 inhibition | 5 |
| Trazodone | None | 5 |
| SSRIs: selective serotonin reuptake inhibitors | ||
| Source: Reference 8 | ||
Assessing substance abuse
Up to 50% of liver transplant candidates have a history of alcohol and/or drug abuse,9 the highest rate among transplant populations. Alcohol-induced cirrhosis and hepatitis C contracted from IV drug use are common indications for liver transplant. Effective treatment of substance abuse is essential because 30% to 50% of these patients relapse after the procedure.10 Assess:
- each substance abused, including onset, peak, and current use
- family history of substance abuse disorders
- past efforts at rehabilitation
- tobacco use (smoking before and after transplant is related to an increased incidence of new cancer diagnoses).11
Some transplant centers require patients with substance
use disorders to participate in 12-step programs or
rehabilitation. Regardless of the institutions’
requirements, encourage patients to participate in
rehabilitation to prevent relapse and mitigate the
negative impact of substance abuse on physical
and mental well-being.
Mental status examination includes the usual elements such as appearance, behavior, speech, affect, and thought process. Assess for suicidal thinking or hopelessness, which have been linked to serious medical illness.12 Question patients about hallucinations and give special attention to visual aberrations, which may occur in medically ill patients.
Cognitive testing. Use tools such as the Mini-Mental State Examination, clock drawing test, and Trail Making A and B tests to assess cognitive ability. If patients show signs of cognitive impairment, arrange for follow-up examinations and refer for neuropsychological testing.
Some cognitive impairment—such as that caused by hepatic encephalopathy—will likely improve after transplant, but other types—such as that caused by vascular disease—will not. If confusion is caused by hepatic encephalopathy, treatment with lactulose might rapidly improve symptoms. Remember that patients with hepatic encephalopathy might not exhibit elevated ammonia levels. Underlying causes of worsening hepatic encephalopathy—such as infections or bleeding—might require treatment.
Assessing adherence. Medication adherence after transplant is essential to prevent organ rejection and other complications. Posttransplant regimens are complex, and the frequency of follow-up assessments can be intense—particularly in the first year after transplant.
Your pretransplant assessment can identify where patients have struggled with adherence in the past. Before the transplant, your team can work to correct barriers such as inability to pay for medications, child care problems, or transportation needs.
Personality disorders have been identified as predictors of posttransplant nonadherence, and 50% to 60% of transplant programs consider personality disorders a relative contraindication to organ transplant.13 Address other contributors to poor adherence—such as substance abuse or depression—with ongoing psychiatric care.
When assessing a patient’s social support, look for evidence of:
- stable living situations
- long-term relationships with spouses, parents, children, or close friends
- adequate financial resources, including health insurance.
These factors help the patient manage the posttransplant process and numerous follow-up physician visits. Religious organizations or other social institutions also appear to provide the emotional support patients need to cope with an organ transplant.
Social support is essential to help with the normal difficulties such as frequent clinic visits and initial physical disability patients face after successful transplant (Box 2). Ask about the candidate’s family, friends, spirituality, and finances during your pretransplant assessment. Poor social support is related to the development of posttransplant psychiatric disorders14 and adherence difficulties.15
Assessment instruments—such as the Psychosocial Assessment of Candidates for Transplantation and the Transplant Evaluation Rating Scale3—include social support items and can be useful in identifying weak areas.
Data collected by other team members can be invaluable. A nurse or social worker, for example, may observe that a patient is unwilling to take medications, contrary to the patient’s report. Other sources of information include the patient’s family and friends, a primary care physician, or other mental health providers such as a therapist or case manager.
Posttransplant psychiatric care
Depression. The incidence of depression is higher in the year following transplant than before transplant or in the immediate posttransplant period.5 Predictors of posttransplant depression include:
- history of depression
- poor social support
- passive coping strategies
- poor physical status after transplantation.16,17
Carefully monitor patients who present with these factors after transplant. Treat depression with supportive measures designed to improve the patient’s social network and coping skills and pharmacotherapy. Select antidepressant medications based on side effect profiles and impact on the patient’s transplanted organs.
Substance abuse. Patients with a pretransplant history of substance abuse often relapse. Among transplant recipients with a history of alcoholic liver disease, drinking rates of 30% to 40% have been reported 5 years after transplant. Most of these data represent occasional use, not heavy or regular drinking.18 Relapse can occur despite careful assessment and follow-up.
Some evidence suggests that transplant patients who resume drinking have worse outcomes than those who abstain. Alcoholism relapse has other negative consequences, such as relationship problems and employment difficulties.
Predictors of relapse include:
- pretransplant history of alcohol dependence
- family history of alcoholism
- rehabilitation history, which could indicate a severe substance abuse disorder.3
Medications for alcoholism treatment have not been studied systematically in transplant patients, but low doses of acamprosate, ≤2 g/d, and naltrexone, ≤200 mg/d, are options for patients interested in pharmacotherapy. Support from 12-step programs also helps treat substance-abusing patients.
Altered mental status. Immunosuppressive medications—including cyclosporine, tacrolimus, and prednisone—can have neuropsychiatric effects and could cause a change in mental status (Table 3).19 Check cyclosporine and tacrolimus serum levels against reference ranges when delirium is present. If levels are toxic the dosage often can be lowered, which might lead to clinical improvement.
Quality of life. In general, patients’ quality of life improves after their transplant. After the first year—which patients might find difficult because of changes in physical and social status—quality of life typically improves.5
Table 3
Neuropsychiatric side effects of medications
commonly used in transplant patients
| Medication | Side effects |
|---|---|
| Cyclosporine | Tremor, headache, seizures, hallucinations, delirium |
| Tacrolimus | Tremor, headache, vivid dreams, anxiety, anorexia, seizures, delirium |
| Prednisone | Depression, mania, psychosis, delirium |
| Source: Adapted from references 3,7 | |
Psychiatric disorders such as depression can worsen quality of life. However, quality of life can improve after depression is diagnosed and treated. Other predictors of improved quality of life include older age, marriage, and the absence of a personality disorder.4
Other posttransplant concerns of patients include changes in employment, finances, and relationships. Patients often have been away from work before transplant, and returning after a long absence can be stressful. Patients may find that they cannot work as well as before becoming ill, which may lead to frustration, depression, and/or anxiety symptoms. Transplant surgery requires a large financial investment, and money concerns usually persist long after the transplant.
The transplant recipient’s role within the family may shift after surgery. Families might expect the patient to “return to normal” and resume old activities. Alternatively, family members might continue to treat the patient as a person with chronic illness despite physical improvement. If patients are struggling with these changes, supportive psychotherapy is indicated.
Related resource
- United Network for Organ Sharing. www.unos.org.
- Transplant living. www.transplantliving.org.
- Trzepacz PT, DiMartini AF, eds. The transplant patient. Cambridge, UK: Cambridge University Press; 2000.
- Klapheke MM. The role of the psychiatrist in organ transplantation. Bull Menninger Clin 1999;63(1):13-39.
Drug brand names
- Acamprosate • Campral
- Buspirone • BuSpar
- Bupropion SR • Wellbutrin SR
- Citalopram • Celexa
- Cyclosporine • Sandimmune
- Escitalopram • Lexapro
- Fluoxetine • Prozac
- Fluvoxamine • Luvox
- Gabapentin • Neurontin
- Lactulose • Cephulac, Chronulac
- Mirtazapine • Remeron
- Naltrexone • ReVia
- Paroxetine • Paxil
- Prednisone • Deltasone
- Sertraline • Zoloft
- Tacrolimus • Prograf
- Trazodone • Desyrel
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. United Network for Organ Sharing. Transplants by donor type. U.S. transplants performed: January 1, 1988 – April 30, 2007. Organ Procurement and Transplantation Network. Available at: http://www.unos.org. Accessed July 26, 2007.
2. Crone CC, Wise TN. Psychiatric aspects of transplantation, I: evaluation and selection of candidates. Crit Care Nurs 1999;19:79-87.
3. DiMartini AF, Dew MA, Trzepacz PT. Organ transplantation. In: Levenson JL, ed. The American Psychiatric Publishing textbook of psychosomatic medicine. Washington DC: American Psychiatric Publishing, Inc, 2005:675-700.
4. Cupples S, Dew MA, Grady KL, et al. Report of the psychosocial outcomes workgroup of the nursing and social sciences council of the international society for heart and lung transplantation: present status of research on psychosocial outcomes in cardiothoracic transplantation: review and recommendations for the field. J Heart Lung Transplant 2006;25:716-25.
5. Dew MA, DiMartini AF. Psychological disorders and distress after adult cardiothoracic transplantation. J Cardiovasc Nurs 2005;20:S51-S66.
6. Barbour KA, Blumenthal JA, Palmer SM. Psychosocial issues in the assessment and management of patients undergoing lung transplantation. Chest 2006;129:1367-74.
7. Trzepacz PT, Levenson JL, Tringali RA. Psychopharmacology and neuropsychiatric syndromes in organ transplantation. Gen Hosp Psychiatry 1991;13:233-45.
8. Crone CC, Gabriel GM. Treatment of anxiety and depression in transplant patients. Clin Pharmacokinet 2004;43:361-94.
9. DiMartini A, Weinrieb R, Mireman M. Liver transplantation in patients with alcohol and other substance use disorders. Psychiatr Clin North Am 2002;25:195-209.
10. Weinrieb RM, Van Horn DHA, McLellan AT, et al. Alcoholism treatment after liver transplantation: lessons learned from a clinical trial that failed. Psychosomatics 2001;42:110-6.
11. Jimenez C, Manrique A, Marques E, et al. Incidence and risk factors for the development of lung tumors after liver transplantation. Transpl Int 2007;20:57-63.
12. Juurlink DN, Herrmann N, Szalai JP, et al. Medical illness and the risk of suicide in the elderly. Arch Intern Med 2004;14:1179-84.
13. Levenson JL, Olbrisch ME. Psychosocial screening and selection of candidates for organ transplantation. In: Trzepacz PT, DiMartini AF, eds. The transplant patient. Cambridge, UK: Cambridge University Press, 2000:21-41.
14. Dew MA, Kormos RL, DiMartini AF, et al. Prevalence and risk of depression and anxiety-related disorders during the first three years after heart transplantation. Psychosomatics 2001;42:300-13.
15. Dew MA, Roth LH, Thompson ME, et al. Medical compliance and its predictors in the first year after cardiac transplantation. J Heart Lung Transplant 1996;15:631-45.
16. Dew MA, Myaskovsky L, Switzer GE, et al. Profiles and predictors of the course of psychological distress across four years after heart transplantation. Psychol Med 2005;35:1215-27.
17. Goetzmann L, Klaghofer R, Wagner-Huber R, et al. Psychosocial vulnerability predicts psychosocial outcome after an organ transplant: results of a prospective study with lung, liver, and bone-marrow patients. J Psychosom Res 2007;62:93-100.
18. Lucey M. Liver transplantation for alcoholic liver disease: a progress report. Graft 1999;2:S73-9.
19. Beresford TP. Neuropsychiatric complications of liver and other solid organ transplantation. Liver Transpl 2001;7(11 suppl 1):S36-S45.
1. United Network for Organ Sharing. Transplants by donor type. U.S. transplants performed: January 1, 1988 – April 30, 2007. Organ Procurement and Transplantation Network. Available at: http://www.unos.org. Accessed July 26, 2007.
2. Crone CC, Wise TN. Psychiatric aspects of transplantation, I: evaluation and selection of candidates. Crit Care Nurs 1999;19:79-87.
3. DiMartini AF, Dew MA, Trzepacz PT. Organ transplantation. In: Levenson JL, ed. The American Psychiatric Publishing textbook of psychosomatic medicine. Washington DC: American Psychiatric Publishing, Inc, 2005:675-700.
4. Cupples S, Dew MA, Grady KL, et al. Report of the psychosocial outcomes workgroup of the nursing and social sciences council of the international society for heart and lung transplantation: present status of research on psychosocial outcomes in cardiothoracic transplantation: review and recommendations for the field. J Heart Lung Transplant 2006;25:716-25.
5. Dew MA, DiMartini AF. Psychological disorders and distress after adult cardiothoracic transplantation. J Cardiovasc Nurs 2005;20:S51-S66.
6. Barbour KA, Blumenthal JA, Palmer SM. Psychosocial issues in the assessment and management of patients undergoing lung transplantation. Chest 2006;129:1367-74.
7. Trzepacz PT, Levenson JL, Tringali RA. Psychopharmacology and neuropsychiatric syndromes in organ transplantation. Gen Hosp Psychiatry 1991;13:233-45.
8. Crone CC, Gabriel GM. Treatment of anxiety and depression in transplant patients. Clin Pharmacokinet 2004;43:361-94.
9. DiMartini A, Weinrieb R, Mireman M. Liver transplantation in patients with alcohol and other substance use disorders. Psychiatr Clin North Am 2002;25:195-209.
10. Weinrieb RM, Van Horn DHA, McLellan AT, et al. Alcoholism treatment after liver transplantation: lessons learned from a clinical trial that failed. Psychosomatics 2001;42:110-6.
11. Jimenez C, Manrique A, Marques E, et al. Incidence and risk factors for the development of lung tumors after liver transplantation. Transpl Int 2007;20:57-63.
12. Juurlink DN, Herrmann N, Szalai JP, et al. Medical illness and the risk of suicide in the elderly. Arch Intern Med 2004;14:1179-84.
13. Levenson JL, Olbrisch ME. Psychosocial screening and selection of candidates for organ transplantation. In: Trzepacz PT, DiMartini AF, eds. The transplant patient. Cambridge, UK: Cambridge University Press, 2000:21-41.
14. Dew MA, Kormos RL, DiMartini AF, et al. Prevalence and risk of depression and anxiety-related disorders during the first three years after heart transplantation. Psychosomatics 2001;42:300-13.
15. Dew MA, Roth LH, Thompson ME, et al. Medical compliance and its predictors in the first year after cardiac transplantation. J Heart Lung Transplant 1996;15:631-45.
16. Dew MA, Myaskovsky L, Switzer GE, et al. Profiles and predictors of the course of psychological distress across four years after heart transplantation. Psychol Med 2005;35:1215-27.
17. Goetzmann L, Klaghofer R, Wagner-Huber R, et al. Psychosocial vulnerability predicts psychosocial outcome after an organ transplant: results of a prospective study with lung, liver, and bone-marrow patients. J Psychosom Res 2007;62:93-100.
18. Lucey M. Liver transplantation for alcoholic liver disease: a progress report. Graft 1999;2:S73-9.
19. Beresford TP. Neuropsychiatric complications of liver and other solid organ transplantation. Liver Transpl 2001;7(11 suppl 1):S36-S45.
Correction
A font problem resulted in several symbols being omitted from Figures 1 and 2 in the article, “Can you interpret confidence intervals? It’s not that difficult” (Current Psychiatry, August 2007).
In Figure 1, the y-axis should have included a positive infinity symbol at the top and a negative infinity symbol at the bottom. In Figure 2, an infinity symbol should have been included at the center of the y-axis.
A font problem resulted in several symbols being omitted from Figures 1 and 2 in the article, “Can you interpret confidence intervals? It’s not that difficult” (Current Psychiatry, August 2007).
In Figure 1, the y-axis should have included a positive infinity symbol at the top and a negative infinity symbol at the bottom. In Figure 2, an infinity symbol should have been included at the center of the y-axis.
A font problem resulted in several symbols being omitted from Figures 1 and 2 in the article, “Can you interpret confidence intervals? It’s not that difficult” (Current Psychiatry, August 2007).
In Figure 1, the y-axis should have included a positive infinity symbol at the top and a negative infinity symbol at the bottom. In Figure 2, an infinity symbol should have been included at the center of the y-axis.
How should dementia with psychosis be treated?
Many—if not most—psychiatrists treating dementia-related psychosis in geriatric patients use second-generation antipsychotics (SGAs), a practice not approved by the FDA. Consider the following 2 emergency consultation cases:
- Mrs. A, age 76, has exhibited serious memory difficulties for >1 year. She can no longer find her way home or take care of personal needs. This morning her husband brings her to the ER after she struck him on the head with a frying pan and threatened to kill a 72-year-old widowed neighbor with whom she accused him of having an affair.
- Mr. J, age 82, was diagnosed with Alzheimer’s disease 3 years ago and resides in a nursing home. You receive a call from staff that Mr. J has become very agitated, accused his roommate of stealing his belongings, and screamed at the roommate and staff to give him his “stuff” back.
These 2 vignettes describe classic cases of psychotic symptoms occurring in the context of dementia. Both patients clearly need an antipsychotic to control their delusions and prevent them from endangering themselves and others.
SGAs vs FGAs vs placebo. Late-life dementia is associated with a 50% incidence of psychosis. These patients are extremely susceptible to neurologic movement disorders (acute parkinsonism and subsequent tardive dyskinesia [TD]) when given first-generation antipsychotics (FGAs) and far less so with SGAs. After 9 months’ exposure, geriatric patients with psychotic symptoms have been shown to be 10 times more likely to develop TD with FGAs (28%) than with SGAs (2.5%).1
Even so, in 2005 the FDA imposed a “black-box” warning on the use of SGAs for psychosis related to dementia because the mortality rate in 17 pooled placebo-controlled dementia studies was approximately 1.7 times higher with SGAs (4.5%) compared with placebo (2.5%).2 Causes of death were mostly heart-related or pneumonia.
When treating psychosis in the elderly, however, we don’t choose between an SGA and placebo but between SGAs and FGAs. Thus, the relative mortality risk of these 2 drug classes is what really matters to clinicians who prefer to use SGAs but feel inhibited by the black-box warning.
New evidence. Many practitioners might not be aware that 4 studies published since 2004 of elderly patients receiving antipsychotics have addressed the relative risk of mortality with SGAs vs FGAs. In patients age ≥65 with dementia, these studies found:
- In a 2-year U.S. retrospective review, the mortality rate was 21% among those receiving haloperidol vs approximately 5% among those receiving risperidone or olanzapine.3
- In a retrospective U.S. study of 2,890 patients who started FGAs or SGAs between 1994 and 2003, FGAs were at least as likely as SGAs to increase the risk of death. The greatest increases in risk were seen soon after patients started antipsychotic therapy and with higher dosages of FGAs.4
- In a 2-year prospective study of 254 very frail patients (mean age 86) in Finnish hospitals or nursing homes, neither the use of FGAs nor SGAs increased the risk of death or hospital admission. The use of restraints, however, doubled the risk of death.5
- In a population-based, retrospective Canadian study of 27,259 matched pairs, a statistically significant increase in mortality was seen with SGAs compared with no antipsychotic use, whether patients lived at home or in long-term-care facilities. This difference was seen 30 days after treatment started and seemed to persist to 180 days. By comparison, the mortality risk appeared to be higher with FGAs than with SGAs at all measured time points.6
The black-box warning on SGAs does not guide clinicians in the use of antipsychotics; it simply compares a class of drugs with placebo, and sugar pills are not an option for managing psychosis. The 4 published studies represent a more useful guide about the relative mortality risk of FGAs and SGAs. They also provide evidence that supports clinical practice in managing patients with psychosis in late-life dementia.
1. Jeste DV, Lacro JP, Bailey A, et al. Lower incidence of tardive dyskinesia with risperidone compared with haloperidol in older patients. J Am Geriatr Soc 1999;47(6):716-9.
2. U.S. Food and Drug Administration. Deaths with antipsychotics in elderly patients with behavioral disturbances. Available at: http://www.fda.gov/cder/drug/advisory/antipsychotics.htm. Accessed August 7, 2007.
3. Nasrallah HA, White T, Nasrallah AT. Lower mortality in geriatric patients receiving risperidone and olanzapine versus haloperidol: preliminary analysis of retrospective data. Am J Geriatr Psychiatry 2004;12:475-9.
4. Wang PS, Schneeweiss S, Avorn J, et al. Risk of death in elderly users of conventional vs. atypical antipsychotic medications. N Engl J Med 2005;353:2335-41.
5. Gill SS, Bronskill SE, Normand SL, et al. Antipsychotic drug use and mortality in older adults with dementia. Ann Intern Med 2007;146:775-86.
6. Raivio MM, Laurila JV, Strandberg TE, et al. Neither atypical nor conventional antipsychotics increase mortality or hospital admissions among elderly patients with dementia: a two-year prospective study. Am J Geriatr Psychiatry 2007;15:416-24.
Many—if not most—psychiatrists treating dementia-related psychosis in geriatric patients use second-generation antipsychotics (SGAs), a practice not approved by the FDA. Consider the following 2 emergency consultation cases:
- Mrs. A, age 76, has exhibited serious memory difficulties for >1 year. She can no longer find her way home or take care of personal needs. This morning her husband brings her to the ER after she struck him on the head with a frying pan and threatened to kill a 72-year-old widowed neighbor with whom she accused him of having an affair.
- Mr. J, age 82, was diagnosed with Alzheimer’s disease 3 years ago and resides in a nursing home. You receive a call from staff that Mr. J has become very agitated, accused his roommate of stealing his belongings, and screamed at the roommate and staff to give him his “stuff” back.
These 2 vignettes describe classic cases of psychotic symptoms occurring in the context of dementia. Both patients clearly need an antipsychotic to control their delusions and prevent them from endangering themselves and others.
SGAs vs FGAs vs placebo. Late-life dementia is associated with a 50% incidence of psychosis. These patients are extremely susceptible to neurologic movement disorders (acute parkinsonism and subsequent tardive dyskinesia [TD]) when given first-generation antipsychotics (FGAs) and far less so with SGAs. After 9 months’ exposure, geriatric patients with psychotic symptoms have been shown to be 10 times more likely to develop TD with FGAs (28%) than with SGAs (2.5%).1
Even so, in 2005 the FDA imposed a “black-box” warning on the use of SGAs for psychosis related to dementia because the mortality rate in 17 pooled placebo-controlled dementia studies was approximately 1.7 times higher with SGAs (4.5%) compared with placebo (2.5%).2 Causes of death were mostly heart-related or pneumonia.
When treating psychosis in the elderly, however, we don’t choose between an SGA and placebo but between SGAs and FGAs. Thus, the relative mortality risk of these 2 drug classes is what really matters to clinicians who prefer to use SGAs but feel inhibited by the black-box warning.
New evidence. Many practitioners might not be aware that 4 studies published since 2004 of elderly patients receiving antipsychotics have addressed the relative risk of mortality with SGAs vs FGAs. In patients age ≥65 with dementia, these studies found:
- In a 2-year U.S. retrospective review, the mortality rate was 21% among those receiving haloperidol vs approximately 5% among those receiving risperidone or olanzapine.3
- In a retrospective U.S. study of 2,890 patients who started FGAs or SGAs between 1994 and 2003, FGAs were at least as likely as SGAs to increase the risk of death. The greatest increases in risk were seen soon after patients started antipsychotic therapy and with higher dosages of FGAs.4
- In a 2-year prospective study of 254 very frail patients (mean age 86) in Finnish hospitals or nursing homes, neither the use of FGAs nor SGAs increased the risk of death or hospital admission. The use of restraints, however, doubled the risk of death.5
- In a population-based, retrospective Canadian study of 27,259 matched pairs, a statistically significant increase in mortality was seen with SGAs compared with no antipsychotic use, whether patients lived at home or in long-term-care facilities. This difference was seen 30 days after treatment started and seemed to persist to 180 days. By comparison, the mortality risk appeared to be higher with FGAs than with SGAs at all measured time points.6
The black-box warning on SGAs does not guide clinicians in the use of antipsychotics; it simply compares a class of drugs with placebo, and sugar pills are not an option for managing psychosis. The 4 published studies represent a more useful guide about the relative mortality risk of FGAs and SGAs. They also provide evidence that supports clinical practice in managing patients with psychosis in late-life dementia.
Many—if not most—psychiatrists treating dementia-related psychosis in geriatric patients use second-generation antipsychotics (SGAs), a practice not approved by the FDA. Consider the following 2 emergency consultation cases:
- Mrs. A, age 76, has exhibited serious memory difficulties for >1 year. She can no longer find her way home or take care of personal needs. This morning her husband brings her to the ER after she struck him on the head with a frying pan and threatened to kill a 72-year-old widowed neighbor with whom she accused him of having an affair.
- Mr. J, age 82, was diagnosed with Alzheimer’s disease 3 years ago and resides in a nursing home. You receive a call from staff that Mr. J has become very agitated, accused his roommate of stealing his belongings, and screamed at the roommate and staff to give him his “stuff” back.
These 2 vignettes describe classic cases of psychotic symptoms occurring in the context of dementia. Both patients clearly need an antipsychotic to control their delusions and prevent them from endangering themselves and others.
SGAs vs FGAs vs placebo. Late-life dementia is associated with a 50% incidence of psychosis. These patients are extremely susceptible to neurologic movement disorders (acute parkinsonism and subsequent tardive dyskinesia [TD]) when given first-generation antipsychotics (FGAs) and far less so with SGAs. After 9 months’ exposure, geriatric patients with psychotic symptoms have been shown to be 10 times more likely to develop TD with FGAs (28%) than with SGAs (2.5%).1
Even so, in 2005 the FDA imposed a “black-box” warning on the use of SGAs for psychosis related to dementia because the mortality rate in 17 pooled placebo-controlled dementia studies was approximately 1.7 times higher with SGAs (4.5%) compared with placebo (2.5%).2 Causes of death were mostly heart-related or pneumonia.
When treating psychosis in the elderly, however, we don’t choose between an SGA and placebo but between SGAs and FGAs. Thus, the relative mortality risk of these 2 drug classes is what really matters to clinicians who prefer to use SGAs but feel inhibited by the black-box warning.
New evidence. Many practitioners might not be aware that 4 studies published since 2004 of elderly patients receiving antipsychotics have addressed the relative risk of mortality with SGAs vs FGAs. In patients age ≥65 with dementia, these studies found:
- In a 2-year U.S. retrospective review, the mortality rate was 21% among those receiving haloperidol vs approximately 5% among those receiving risperidone or olanzapine.3
- In a retrospective U.S. study of 2,890 patients who started FGAs or SGAs between 1994 and 2003, FGAs were at least as likely as SGAs to increase the risk of death. The greatest increases in risk were seen soon after patients started antipsychotic therapy and with higher dosages of FGAs.4
- In a 2-year prospective study of 254 very frail patients (mean age 86) in Finnish hospitals or nursing homes, neither the use of FGAs nor SGAs increased the risk of death or hospital admission. The use of restraints, however, doubled the risk of death.5
- In a population-based, retrospective Canadian study of 27,259 matched pairs, a statistically significant increase in mortality was seen with SGAs compared with no antipsychotic use, whether patients lived at home or in long-term-care facilities. This difference was seen 30 days after treatment started and seemed to persist to 180 days. By comparison, the mortality risk appeared to be higher with FGAs than with SGAs at all measured time points.6
The black-box warning on SGAs does not guide clinicians in the use of antipsychotics; it simply compares a class of drugs with placebo, and sugar pills are not an option for managing psychosis. The 4 published studies represent a more useful guide about the relative mortality risk of FGAs and SGAs. They also provide evidence that supports clinical practice in managing patients with psychosis in late-life dementia.
1. Jeste DV, Lacro JP, Bailey A, et al. Lower incidence of tardive dyskinesia with risperidone compared with haloperidol in older patients. J Am Geriatr Soc 1999;47(6):716-9.
2. U.S. Food and Drug Administration. Deaths with antipsychotics in elderly patients with behavioral disturbances. Available at: http://www.fda.gov/cder/drug/advisory/antipsychotics.htm. Accessed August 7, 2007.
3. Nasrallah HA, White T, Nasrallah AT. Lower mortality in geriatric patients receiving risperidone and olanzapine versus haloperidol: preliminary analysis of retrospective data. Am J Geriatr Psychiatry 2004;12:475-9.
4. Wang PS, Schneeweiss S, Avorn J, et al. Risk of death in elderly users of conventional vs. atypical antipsychotic medications. N Engl J Med 2005;353:2335-41.
5. Gill SS, Bronskill SE, Normand SL, et al. Antipsychotic drug use and mortality in older adults with dementia. Ann Intern Med 2007;146:775-86.
6. Raivio MM, Laurila JV, Strandberg TE, et al. Neither atypical nor conventional antipsychotics increase mortality or hospital admissions among elderly patients with dementia: a two-year prospective study. Am J Geriatr Psychiatry 2007;15:416-24.
1. Jeste DV, Lacro JP, Bailey A, et al. Lower incidence of tardive dyskinesia with risperidone compared with haloperidol in older patients. J Am Geriatr Soc 1999;47(6):716-9.
2. U.S. Food and Drug Administration. Deaths with antipsychotics in elderly patients with behavioral disturbances. Available at: http://www.fda.gov/cder/drug/advisory/antipsychotics.htm. Accessed August 7, 2007.
3. Nasrallah HA, White T, Nasrallah AT. Lower mortality in geriatric patients receiving risperidone and olanzapine versus haloperidol: preliminary analysis of retrospective data. Am J Geriatr Psychiatry 2004;12:475-9.
4. Wang PS, Schneeweiss S, Avorn J, et al. Risk of death in elderly users of conventional vs. atypical antipsychotic medications. N Engl J Med 2005;353:2335-41.
5. Gill SS, Bronskill SE, Normand SL, et al. Antipsychotic drug use and mortality in older adults with dementia. Ann Intern Med 2007;146:775-86.
6. Raivio MM, Laurila JV, Strandberg TE, et al. Neither atypical nor conventional antipsychotics increase mortality or hospital admissions among elderly patients with dementia: a two-year prospective study. Am J Geriatr Psychiatry 2007;15:416-24.
A young man’s affair of the heart
Case: You’re a ‘freak’
A local mental health agency refers Mr. Z, age 23, to our inpatient psychiatry service because of increasing suicidality and psychosis. He began receiving care from the mental health agency 3 years ago, after a psychiatrist diagnosed paranoid schizophrenia.
At presentation, Mr. Z is delusionally preoccupied with a brief relationship he had with a young woman at college 2 years ago. He feels embarrassed about his conduct toward her during a psychotic episode and her subsequent response. He believes strangers are ridiculing him, and he hears voices calling him a “freak” and making crude references to the encounter. He is also contemplating suicide and endorses a suicide plan.
Mr. Z was hospitalized for 1 month last year with schizophrenia symptoms. He is medically healthy and does not abuse alcohol or drugs.
We admit Mr. Z because of his suicidality. Four weeks later, he remains suicidal and hears voices telling him to “rape” and “kill.” Successive 2-week trials of risperidone, 1 mg/d titrated to 5 mg/d, and quetiapine, 200 mg/d titrated to 700 mg/d, cause intolerable akathisia. We try adding propranolol, 20 mg every 8 hours, to alleviate akathisia, but to no avail. Previous trials of olanzapine, 30 mg/d, and haloperidol, dosage unknown, were unsuccessful or caused akathisia.
The authors’ observations
Substantial evidence supports clozapine’s efficacy in treatment-resistant schizophrenia, and this second-generation antipsychotic (SGA) also might reduce suicidality.1,2 Clinicians often combine antipsychotics, switch to an antidepressant, or add a mood stabilizer for treatment-resistant schizophrenia,3 but little evidence supports these options.
Mr. Z had failed at least 4 antipsychotic trials. We consider clozapine for patients with severe psychosis who have failed 2 or 3 antipsychotic trials or cannot tolerate these medications. Severity of psychosis and presence of suicidality warrant use of clozapine in treatment-resistant cases.
If Mr. Z had tolerated risperidone or quetiapine, we would have waited as long as 8 weeks before switching to clozapine. In inpatients, improvement should be seen 2 to 4 weeks after starting an antipsychotic.
Perform blood tests weekly during the first 6 months of clozapine therapy and bi-weekly thereafter to check for abnormally low white blood cell counts that might suggest agranulocytosis.
Myocarditis is a potentially fatal inflammation of the myocardium that can result from a viral infection, toxins, medications, or hypersensitive immune reactions.
Data on myocarditis prevalence are scarce because no relatively noninvasive assessment tools exist. Among 2,200 patients with unexplained heart failure occurring over 5
An FDA-mandated “black box” in clozapine’s package insert describes an “increased risk of fatal myocarditis, especially during—but not limited to—the first month of therapy.”6 Proposed explanations of how clozapine causes myocarditis include:
- direct toxic effect on cardiac myocytes related to impaired clozapine metabolism in some patients7,8
- myocardial damage mediated by clozapine blockade of a muscarinic M2 receptor subtype9
- selenium deficiency or presence of reactive clozapine nitrenium metabolites contributing to myocardial toxicity.10,11
The common presence of peripheral eosinophilia on autopsy—including diffuse eosinophilic infiltrates in myocardial and perivascular areas—might suggest a hypereosinophilic syndrome or a type II hypersensitive immune reaction mediated by clozapine.7,12 Similar immune-mediated conditions of acute, progressive myocarditis have been noted after exposure to other medications such as penicillin or sulfonamides.13
Noting that clozapine increases inflammatory cytokines, some authors believe TNF-alpha and other inflammatory cytokines contribute to myocarditis.14
TREATMENT: New regimen
After discussing clozapine’s risks and benefits with Mr. Z and his parents, we start the medication at 25 mg/d to gauge tolerability, then titrate to 300 mg/d over 10 days. Mr. Z tolerates clozapine well, with some sedation and sialorrhea. A blood test taken 7 days after we start clozapine shows a normal white blood cell count.
After 10 days on clozapine, Mr. Z’s delusions and hallucinations are considerably less intense. He is no longer suicidal and visits his former college with his parents without thinking about his past acquaintance. We discharge him on clozapine, 300 mg/d, and refer him to the local mental health agency.
Two days later, Mr. Z’s parents report that since discharge their son has had extreme fatigue, shortness of breath, leg edema, and chest pain. We advise them to immediately take their son to the ER for cardiac workup.
The authors’ observations
Mr. Z’s sudden-onset physical symptoms suggest myocarditis, a rare but potentially fatal side effect of clozapine whose specific cause is unclear (Box 1).5-14 Myocarditis has been reported in 0.02% to 0.18% of patients exposed to clozapine,15-18 with incidence as high as 1.3% per 235 patients.19
Affected patients typically have been taking clozapine at therapeutic dosages (100 to 450 mg/d).7 Clozapine use is most prevalent among men ages 20 to 40, who tend to have more severe schizophrenia and lower cardiac risk than other populations. Correspondingly, clozapine-induced myocarditis is most prevalent in younger men,20 although what specifically causes this susceptibility is unknown.
Nonspecific symptoms such as dyspnea, tachycardia, chest pain, or fever can signal myocarditis (Table)7,21 and can surface within 4 to 8 weeks of starting clozapine.22 Haas et al20 reported other symptoms—such as leukocytosis—in young (median age 30), predominantly male patients with clozapine-induced myocarditis. Symptoms that typically occur during clozapine titration—such as fever and tachycardia—can mask “subclinical” myocarditis.22
Mr. Z’s nonspecific symptoms could signal clozapine-induced agranulocytosis or a viral syndrome, or could be delusional. The patient’s acute, sudden symptom onset strongly suggests a cardiac cause. Also, his delusions subsided, and normal blood readings helped us rule out agranulocytosis.
Coulter et al23 associated myocarditis and cardiomyopathy, a noninflammatory heart muscle disease, with several antipsychotics—including clozapine, chlorpromazine, fluphenazine, haloperidol, and risperidone—as well as lithium. More research is needed to confirm this association.
Order a cardiology consult and workup including:
- serum electrolytes
- complete blood count
- ECG21
- tests for myocardial damage including creatine kinase with MB fractionation (CK-MB) and testing for serum troponin I,25 lactic dehydrogenase, and aspartate transaminase (SGOT)21
- assessment for immune activation and peripheral eosinophilia.25
Table
Symptoms that could signal myocarditis in patients taking clozapine
|
| Source: Reference 7 |
TESTING: ‘Is this necessary?’
We contact the ER physician to request the above-mentioned tests, but he questions the need for such extensive and costly testing in a psychiatric patient with nonspecific symptoms.
After several phone conversations to review our recommendations, the emergency physician suggests sending Mr. Z home on a watch-and-wait protocol. We politely but firmly emphasize that Mr. Z needs a full cardiac workup, after which the physician consents to the tests (Box 2).
FINDINGS: suspicious readings
Mr. Z’s cardiac imaging results suggest a cardiopathy:
- echocardiogram shows mild ventricular enlargement with a decreased ejection fraction of 45% (normal reading, 55% to 60%)
- ECG shows normal sinus rhythm with low-voltage diffuse T-wave flattening throughout all leads without ST elevation
- creatine phosphokinase (CPK) and CKMB are within normal ranges
- troponin I is 0.33 ng/mL, a high-normal reading.
Based on these readings, the cardiology service admits Mr. Z with a presumptive diagnosis of clozapine-induced cardiomyopathy. The attending cardiologist stops clozapine and starts the angiotensin-converting enzyme inhibitor enalapril, 2.5 mg bid, for ventricular remodeling. Medical workup includes cytologic testing to rule out immunologic or viral disease.
Five days later, Mr. Z’s cardiac symptoms have resolved. The cardiology unit discharges him on enalapril, 2.5 mg bid, and schedules a cardiac ultrasound for 2 weeks after discharge to confirm progress.
The authors’ observations
Maintain high clinical suspicion while using clozapine. Similar to other patients with a clozapine-induced cardiopathy,16 Mr. Z showed rapid symptomatic changes after a benign initial course and experienced fairly vague symptoms that raised limited clinical concern at first.
Before starting clozapine therapy, screen all patients for pre-existing cardiac disease, which contraindicates this medication. Alert patients and caregivers to the risks and symptoms that require close monitoring early in treatment.
Many researchers suggest monitoring for myocarditis during the first month of therapy and ordering ECG at baseline and 2 and 4 weeks after starting clozapine.21,22 Berk et al26 suggest more aggressive monitoring, including:
- baseline ECG
- transthoracic echocardiogram
- baseline troponin/CK-MB
- ECG and troponin/CK-MB at 7 and 14 days
- echocardiogram at 6 and 12 months and then annually.
RELAPSE: Return of the ‘freak’
Immediately after Mr. Z’s discharge from the cardiology unit, we readmit him to inpatient psychiatry. His parents and case manager say he is again becoming preoccupied with his brief college relationship. He has been off clozapine for 5 days.
The authors’ observations
The American Psychiatric Association27 (see http://www.psych.org/psych_pract/treatg/pg/SchizPG-Complete-Feb04.pdf) recommends maximizing 1 medication for at least 2 to 4 weeks to assess schizophrenia symptom response and urges clinicians to consider adverse effects, medical comorbidities, and patient preference before continuing the medication.
These recommendations highlight the challenges of treating medication-resistant schizophrenia. Relapse is common after a serious reaction to clozapine, and combining 2 or more other antipsychotics could lead to significantly greater side effects. A time-limited trial with an antipsychotic and an adjunctive agent might be attempted while carefully weighing the combination’s risks and benefits.27
Clozapine reduced Mr. Z’s psychosis, but rechallenge would likely cause his potentially fatal cardiomyopathy to re-emerge. His sensitivity to adverse antipsychotic effects discourages polypharmacy and further complicates our decision.
How to convince other specialists
Many physicians are reluctant to pursue additional tests or procedures—and risk a confrontation with a consultant, insurer, or ER physician—especially when the risk of abnormality is extremely low. Advocating for cardiac workup in patients with vague symptoms is challenging, particularly if the suspected side effect is rare.
Taking the path of least resistance can increase the risk of a serious—albeit rare—adverse event. Failure to test could prolong a potentially harmful treatment, and the test results—even if negative—could be critical to planning care.
Calmly but firmly spell out the risks of missing a suspected cardiac problem (death, proceeding with potentially harmful treatment). Tell the ER manager or consultant, “I realize this is a very rare side effect, but not catching it could be life-threatening.”
Be circumspect when pleading your case—an overaggressive approach might cause the ER doctor to “dig in his heels” and reject your request. Use a medically focused response such as, “This is a known complication of this medicine with this common time course and presentation.”
TREATMENT: Another trial
We start olanzapine, 5 mg/d, and titrate to 20 mg/d over 1 week. We add sustained-release bupropion, 200 mg bid, for associated dysphoria.
Mr. Z’s symptoms and paranoia gradually decline, and he tolerates off-unit passes with friends and family before discharge. Staff works closely with him to develop cognitive-behavioral strategies to manage residual paranoia and hallucinations, such as assessing evidence for his delusional beliefs and developing tools to distract him from remaining “voices.” He reports no cardiac symptoms and continues taking enalapril, 2.5 mg bid.
We discharge Mr. Z after 1 week, at which point he shows no suicidal or homicidal thoughts. Follow-up echocardiogram 2 weeks later shows ejection fraction has improved to 60%, suggesting absence of cardiomyopathy.
Related resource
- Clozapine safety information.
www.clozaril.com/.
- Bupropion • Wellbutrin
- Chlorpromazine • Thorazine
- Clozapine • Clozaril
- Enalapril • Vasotec
- Fluphenazine • Prolixin, Permitil
- Haloperidol • Haldol
- Lithium • Eskalith, others
- Olanzapine • Zyprexa
- Propranolol • Inderal
- Quetiapine • Seroquel
- Risperidone • Risperdal
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Kane J, Honigfeld G, Singer J, Meltzer H. Clozapine for the treatment-resistant schizophrenic: a double-blind comparison with chlorpromazine. Arch Gen Psychiatry 1988;45:789-96.
2. Meltzer HY, Alphs L, Green AI, et al. International Suicide Prevention Trial Study Group. Clozapine treatment for suicidality in schizophrenia: International Suicide Prevention Trial (InterSePT). Arch Gen Psychiatry 2003;60:82-91.
3. Stahl SM. Antipsychotic polypharmacy, part 1: therapeutic option or dirty little secret? J Clin Psychiatry 1999;60:425-6.
4. Clozaril monograph. Novartis Phamaceuticals Corp.; April 12, 2006. Available at http://www.novartis.ca/downloads/en/products/clozaril_scrip_e.pdf. Accessed August 13, 2007.
5. Mason JW, O’Connell JB, Herskowitz A, et al. A clinical trial of immunosuppressive therapy for myocarditis. N Engl J Med 1995;333:269.-
6. Physicians’ desk reference. 61st ed. Montvale, NJ: Thomson PDR; 2007.
7. Merrill DB, Dec GW, Goff DC. Adverse cardiac effects associated with clozapine. J Clin Psychopharmacol 2005;25:32-41.
8. Jenie LE. Cardiovascular toxicity with clozapine therapy. Riverview Hospital Pharmacy Newsletter 2002;22:1-3.
9. Devarajan S, Kutcher SP, Dursun SM. Clozapine and sudden death. Lancet 2000;355:841.-
10. Vaddadi KS, Soosai E, Vaddadi G. Low blood selenium concentrations in schizophrenic patients on clozapine. Br J Clin Pharmacol 2003;55:307-9.
11. Williams DP, O’Donnell CJ, Maggs JL, et al. Bioactivation of clozapine by murine cardiac tissue in vivo and in vitro. Chem Res Toxicol 2003;16:1359-64.
12. Fineschi V, Neri M, Riezzo I, Turillazzi E. Sudden cardiac death due to hypersensitivity myocarditis during clozapine treatment. Int J Legal Med 2004;118:307-9.
13. Kendell KR, Day JD, Hruban RH, et al. Intimate association of eosinophils to collagen bundles in eosinophilic myocarditis and ranitidine induced hypersensitivity myocarditis. Arch Pathol Lab Med 1995;119:1154-60.
14. Pollmacher T, Schuld A, Kraus T, et al. On the clinical relevance of clozapine-triggered release of cytokines and soluble cytokine-receptors [in German]. Fortschr Neurol Psychiatr 2001;69(suppl 2):S65-S74.
15. Killian JG, Kerr K, Lawrence C, Celermajer DS. Myocarditis and cardiomyopathy associated with clozapine. Lancet 1999;354:1841-5.
16. Committee on Safety of Medicines Myocarditis with antipsychotics: recent cases with clozapine (Clozaril). Curr Probl Pharmacovigilance 1993;19:9.-
17. Degner D, Bleich S, Grohmann R, et al. Myocarditis associated with clozapine treatment. Aust NZ J Psychiatry 2000;34:880.-
18. La Grenade L, Graham D, Trontell A. Myocarditis and cardiomyopathy associated with clozapine use in the United States (letter). N Engl J Med 2001;345:224-5.
19. Reinders J, Parsonage W, Lange D, et al. Clozapinerelated myocarditis and cardiomyopathy in an Australian metropolitan psychiatric service. Aust NZ J Psychiatry 2004;38:915-22.
20. Haas SJ, Hill R, Krum H, et al. Clozapine-associated myocarditis: a review of 116 cases of suspected myocarditis associated with the use of clozapine in Australia during 1993-2003. Drug Saf 2007;30:47-57.
21. Wehmeier PM, Heiser P, Remschmidt H. Myocarditis, pericarditis and cardiomyopathy in patients treated with clozapine. J Clin Pharm Ther 2005;30:91-6.
22. Merrill DB, Ahmari SE, Bradford JM, Lieberman JA. Myocarditis during clozapine treatment. Am J Psychiatry 2006;163:204-8. Erratum in Am J Psychiatry 2006;163:556.-
23. Coulter DM, Bate A, Meyboom RH, et al. Antipsychotic drugs and heart muscle disorder in international pharmacovigilance: data mining study. BMJ 2001;322:1207-9.
24. Wooltorton E. Antipsychotic clozapine (Clozaril): myocarditis and cardiovascular toxicity. CMAJ 2002;166:1185-6.
25. Kay SE, Doery J, Sholl D. Clozapine associated pericarditis and elevated troponin I. Aust NZ J Psychiatry 2002;36:143-4.
26. Berk M, Fitzsimons J, Lambert T, et al. Monitoring the safe use of clozapine: a consensus view from Victoria, Australia. CNS Drugs 2007;21:117-27.
27. American Psychiatric Association Work Group on Schizophrenia, Lehman AF, chair. Practice guideline for the treatment of patients with schizophrenia, 2nd ed, 2004. Available at: http://www.psych.org/psych_pract/treatg/pg/SchizPG-Complete-Feb04.pdf. Accessed August 15, 2007.
Case: You’re a ‘freak’
A local mental health agency refers Mr. Z, age 23, to our inpatient psychiatry service because of increasing suicidality and psychosis. He began receiving care from the mental health agency 3 years ago, after a psychiatrist diagnosed paranoid schizophrenia.
At presentation, Mr. Z is delusionally preoccupied with a brief relationship he had with a young woman at college 2 years ago. He feels embarrassed about his conduct toward her during a psychotic episode and her subsequent response. He believes strangers are ridiculing him, and he hears voices calling him a “freak” and making crude references to the encounter. He is also contemplating suicide and endorses a suicide plan.
Mr. Z was hospitalized for 1 month last year with schizophrenia symptoms. He is medically healthy and does not abuse alcohol or drugs.
We admit Mr. Z because of his suicidality. Four weeks later, he remains suicidal and hears voices telling him to “rape” and “kill.” Successive 2-week trials of risperidone, 1 mg/d titrated to 5 mg/d, and quetiapine, 200 mg/d titrated to 700 mg/d, cause intolerable akathisia. We try adding propranolol, 20 mg every 8 hours, to alleviate akathisia, but to no avail. Previous trials of olanzapine, 30 mg/d, and haloperidol, dosage unknown, were unsuccessful or caused akathisia.
The authors’ observations
Substantial evidence supports clozapine’s efficacy in treatment-resistant schizophrenia, and this second-generation antipsychotic (SGA) also might reduce suicidality.1,2 Clinicians often combine antipsychotics, switch to an antidepressant, or add a mood stabilizer for treatment-resistant schizophrenia,3 but little evidence supports these options.
Mr. Z had failed at least 4 antipsychotic trials. We consider clozapine for patients with severe psychosis who have failed 2 or 3 antipsychotic trials or cannot tolerate these medications. Severity of psychosis and presence of suicidality warrant use of clozapine in treatment-resistant cases.
If Mr. Z had tolerated risperidone or quetiapine, we would have waited as long as 8 weeks before switching to clozapine. In inpatients, improvement should be seen 2 to 4 weeks after starting an antipsychotic.
Perform blood tests weekly during the first 6 months of clozapine therapy and bi-weekly thereafter to check for abnormally low white blood cell counts that might suggest agranulocytosis.
Myocarditis is a potentially fatal inflammation of the myocardium that can result from a viral infection, toxins, medications, or hypersensitive immune reactions.
Data on myocarditis prevalence are scarce because no relatively noninvasive assessment tools exist. Among 2,200 patients with unexplained heart failure occurring over 5
An FDA-mandated “black box” in clozapine’s package insert describes an “increased risk of fatal myocarditis, especially during—but not limited to—the first month of therapy.”6 Proposed explanations of how clozapine causes myocarditis include:
- direct toxic effect on cardiac myocytes related to impaired clozapine metabolism in some patients7,8
- myocardial damage mediated by clozapine blockade of a muscarinic M2 receptor subtype9
- selenium deficiency or presence of reactive clozapine nitrenium metabolites contributing to myocardial toxicity.10,11
The common presence of peripheral eosinophilia on autopsy—including diffuse eosinophilic infiltrates in myocardial and perivascular areas—might suggest a hypereosinophilic syndrome or a type II hypersensitive immune reaction mediated by clozapine.7,12 Similar immune-mediated conditions of acute, progressive myocarditis have been noted after exposure to other medications such as penicillin or sulfonamides.13
Noting that clozapine increases inflammatory cytokines, some authors believe TNF-alpha and other inflammatory cytokines contribute to myocarditis.14
TREATMENT: New regimen
After discussing clozapine’s risks and benefits with Mr. Z and his parents, we start the medication at 25 mg/d to gauge tolerability, then titrate to 300 mg/d over 10 days. Mr. Z tolerates clozapine well, with some sedation and sialorrhea. A blood test taken 7 days after we start clozapine shows a normal white blood cell count.
After 10 days on clozapine, Mr. Z’s delusions and hallucinations are considerably less intense. He is no longer suicidal and visits his former college with his parents without thinking about his past acquaintance. We discharge him on clozapine, 300 mg/d, and refer him to the local mental health agency.
Two days later, Mr. Z’s parents report that since discharge their son has had extreme fatigue, shortness of breath, leg edema, and chest pain. We advise them to immediately take their son to the ER for cardiac workup.
The authors’ observations
Mr. Z’s sudden-onset physical symptoms suggest myocarditis, a rare but potentially fatal side effect of clozapine whose specific cause is unclear (Box 1).5-14 Myocarditis has been reported in 0.02% to 0.18% of patients exposed to clozapine,15-18 with incidence as high as 1.3% per 235 patients.19
Affected patients typically have been taking clozapine at therapeutic dosages (100 to 450 mg/d).7 Clozapine use is most prevalent among men ages 20 to 40, who tend to have more severe schizophrenia and lower cardiac risk than other populations. Correspondingly, clozapine-induced myocarditis is most prevalent in younger men,20 although what specifically causes this susceptibility is unknown.
Nonspecific symptoms such as dyspnea, tachycardia, chest pain, or fever can signal myocarditis (Table)7,21 and can surface within 4 to 8 weeks of starting clozapine.22 Haas et al20 reported other symptoms—such as leukocytosis—in young (median age 30), predominantly male patients with clozapine-induced myocarditis. Symptoms that typically occur during clozapine titration—such as fever and tachycardia—can mask “subclinical” myocarditis.22
Mr. Z’s nonspecific symptoms could signal clozapine-induced agranulocytosis or a viral syndrome, or could be delusional. The patient’s acute, sudden symptom onset strongly suggests a cardiac cause. Also, his delusions subsided, and normal blood readings helped us rule out agranulocytosis.
Coulter et al23 associated myocarditis and cardiomyopathy, a noninflammatory heart muscle disease, with several antipsychotics—including clozapine, chlorpromazine, fluphenazine, haloperidol, and risperidone—as well as lithium. More research is needed to confirm this association.
Order a cardiology consult and workup including:
- serum electrolytes
- complete blood count
- ECG21
- tests for myocardial damage including creatine kinase with MB fractionation (CK-MB) and testing for serum troponin I,25 lactic dehydrogenase, and aspartate transaminase (SGOT)21
- assessment for immune activation and peripheral eosinophilia.25
Table
Symptoms that could signal myocarditis in patients taking clozapine
|
| Source: Reference 7 |
TESTING: ‘Is this necessary?’
We contact the ER physician to request the above-mentioned tests, but he questions the need for such extensive and costly testing in a psychiatric patient with nonspecific symptoms.
After several phone conversations to review our recommendations, the emergency physician suggests sending Mr. Z home on a watch-and-wait protocol. We politely but firmly emphasize that Mr. Z needs a full cardiac workup, after which the physician consents to the tests (Box 2).
FINDINGS: suspicious readings
Mr. Z’s cardiac imaging results suggest a cardiopathy:
- echocardiogram shows mild ventricular enlargement with a decreased ejection fraction of 45% (normal reading, 55% to 60%)
- ECG shows normal sinus rhythm with low-voltage diffuse T-wave flattening throughout all leads without ST elevation
- creatine phosphokinase (CPK) and CKMB are within normal ranges
- troponin I is 0.33 ng/mL, a high-normal reading.
Based on these readings, the cardiology service admits Mr. Z with a presumptive diagnosis of clozapine-induced cardiomyopathy. The attending cardiologist stops clozapine and starts the angiotensin-converting enzyme inhibitor enalapril, 2.5 mg bid, for ventricular remodeling. Medical workup includes cytologic testing to rule out immunologic or viral disease.
Five days later, Mr. Z’s cardiac symptoms have resolved. The cardiology unit discharges him on enalapril, 2.5 mg bid, and schedules a cardiac ultrasound for 2 weeks after discharge to confirm progress.
The authors’ observations
Maintain high clinical suspicion while using clozapine. Similar to other patients with a clozapine-induced cardiopathy,16 Mr. Z showed rapid symptomatic changes after a benign initial course and experienced fairly vague symptoms that raised limited clinical concern at first.
Before starting clozapine therapy, screen all patients for pre-existing cardiac disease, which contraindicates this medication. Alert patients and caregivers to the risks and symptoms that require close monitoring early in treatment.
Many researchers suggest monitoring for myocarditis during the first month of therapy and ordering ECG at baseline and 2 and 4 weeks after starting clozapine.21,22 Berk et al26 suggest more aggressive monitoring, including:
- baseline ECG
- transthoracic echocardiogram
- baseline troponin/CK-MB
- ECG and troponin/CK-MB at 7 and 14 days
- echocardiogram at 6 and 12 months and then annually.
RELAPSE: Return of the ‘freak’
Immediately after Mr. Z’s discharge from the cardiology unit, we readmit him to inpatient psychiatry. His parents and case manager say he is again becoming preoccupied with his brief college relationship. He has been off clozapine for 5 days.
The authors’ observations
The American Psychiatric Association27 (see http://www.psych.org/psych_pract/treatg/pg/SchizPG-Complete-Feb04.pdf) recommends maximizing 1 medication for at least 2 to 4 weeks to assess schizophrenia symptom response and urges clinicians to consider adverse effects, medical comorbidities, and patient preference before continuing the medication.
These recommendations highlight the challenges of treating medication-resistant schizophrenia. Relapse is common after a serious reaction to clozapine, and combining 2 or more other antipsychotics could lead to significantly greater side effects. A time-limited trial with an antipsychotic and an adjunctive agent might be attempted while carefully weighing the combination’s risks and benefits.27
Clozapine reduced Mr. Z’s psychosis, but rechallenge would likely cause his potentially fatal cardiomyopathy to re-emerge. His sensitivity to adverse antipsychotic effects discourages polypharmacy and further complicates our decision.
How to convince other specialists
Many physicians are reluctant to pursue additional tests or procedures—and risk a confrontation with a consultant, insurer, or ER physician—especially when the risk of abnormality is extremely low. Advocating for cardiac workup in patients with vague symptoms is challenging, particularly if the suspected side effect is rare.
Taking the path of least resistance can increase the risk of a serious—albeit rare—adverse event. Failure to test could prolong a potentially harmful treatment, and the test results—even if negative—could be critical to planning care.
Calmly but firmly spell out the risks of missing a suspected cardiac problem (death, proceeding with potentially harmful treatment). Tell the ER manager or consultant, “I realize this is a very rare side effect, but not catching it could be life-threatening.”
Be circumspect when pleading your case—an overaggressive approach might cause the ER doctor to “dig in his heels” and reject your request. Use a medically focused response such as, “This is a known complication of this medicine with this common time course and presentation.”
TREATMENT: Another trial
We start olanzapine, 5 mg/d, and titrate to 20 mg/d over 1 week. We add sustained-release bupropion, 200 mg bid, for associated dysphoria.
Mr. Z’s symptoms and paranoia gradually decline, and he tolerates off-unit passes with friends and family before discharge. Staff works closely with him to develop cognitive-behavioral strategies to manage residual paranoia and hallucinations, such as assessing evidence for his delusional beliefs and developing tools to distract him from remaining “voices.” He reports no cardiac symptoms and continues taking enalapril, 2.5 mg bid.
We discharge Mr. Z after 1 week, at which point he shows no suicidal or homicidal thoughts. Follow-up echocardiogram 2 weeks later shows ejection fraction has improved to 60%, suggesting absence of cardiomyopathy.
Related resource
- Clozapine safety information.
www.clozaril.com/.
- Bupropion • Wellbutrin
- Chlorpromazine • Thorazine
- Clozapine • Clozaril
- Enalapril • Vasotec
- Fluphenazine • Prolixin, Permitil
- Haloperidol • Haldol
- Lithium • Eskalith, others
- Olanzapine • Zyprexa
- Propranolol • Inderal
- Quetiapine • Seroquel
- Risperidone • Risperdal
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
Case: You’re a ‘freak’
A local mental health agency refers Mr. Z, age 23, to our inpatient psychiatry service because of increasing suicidality and psychosis. He began receiving care from the mental health agency 3 years ago, after a psychiatrist diagnosed paranoid schizophrenia.
At presentation, Mr. Z is delusionally preoccupied with a brief relationship he had with a young woman at college 2 years ago. He feels embarrassed about his conduct toward her during a psychotic episode and her subsequent response. He believes strangers are ridiculing him, and he hears voices calling him a “freak” and making crude references to the encounter. He is also contemplating suicide and endorses a suicide plan.
Mr. Z was hospitalized for 1 month last year with schizophrenia symptoms. He is medically healthy and does not abuse alcohol or drugs.
We admit Mr. Z because of his suicidality. Four weeks later, he remains suicidal and hears voices telling him to “rape” and “kill.” Successive 2-week trials of risperidone, 1 mg/d titrated to 5 mg/d, and quetiapine, 200 mg/d titrated to 700 mg/d, cause intolerable akathisia. We try adding propranolol, 20 mg every 8 hours, to alleviate akathisia, but to no avail. Previous trials of olanzapine, 30 mg/d, and haloperidol, dosage unknown, were unsuccessful or caused akathisia.
The authors’ observations
Substantial evidence supports clozapine’s efficacy in treatment-resistant schizophrenia, and this second-generation antipsychotic (SGA) also might reduce suicidality.1,2 Clinicians often combine antipsychotics, switch to an antidepressant, or add a mood stabilizer for treatment-resistant schizophrenia,3 but little evidence supports these options.
Mr. Z had failed at least 4 antipsychotic trials. We consider clozapine for patients with severe psychosis who have failed 2 or 3 antipsychotic trials or cannot tolerate these medications. Severity of psychosis and presence of suicidality warrant use of clozapine in treatment-resistant cases.
If Mr. Z had tolerated risperidone or quetiapine, we would have waited as long as 8 weeks before switching to clozapine. In inpatients, improvement should be seen 2 to 4 weeks after starting an antipsychotic.
Perform blood tests weekly during the first 6 months of clozapine therapy and bi-weekly thereafter to check for abnormally low white blood cell counts that might suggest agranulocytosis.
Myocarditis is a potentially fatal inflammation of the myocardium that can result from a viral infection, toxins, medications, or hypersensitive immune reactions.
Data on myocarditis prevalence are scarce because no relatively noninvasive assessment tools exist. Among 2,200 patients with unexplained heart failure occurring over 5
An FDA-mandated “black box” in clozapine’s package insert describes an “increased risk of fatal myocarditis, especially during—but not limited to—the first month of therapy.”6 Proposed explanations of how clozapine causes myocarditis include:
- direct toxic effect on cardiac myocytes related to impaired clozapine metabolism in some patients7,8
- myocardial damage mediated by clozapine blockade of a muscarinic M2 receptor subtype9
- selenium deficiency or presence of reactive clozapine nitrenium metabolites contributing to myocardial toxicity.10,11
The common presence of peripheral eosinophilia on autopsy—including diffuse eosinophilic infiltrates in myocardial and perivascular areas—might suggest a hypereosinophilic syndrome or a type II hypersensitive immune reaction mediated by clozapine.7,12 Similar immune-mediated conditions of acute, progressive myocarditis have been noted after exposure to other medications such as penicillin or sulfonamides.13
Noting that clozapine increases inflammatory cytokines, some authors believe TNF-alpha and other inflammatory cytokines contribute to myocarditis.14
TREATMENT: New regimen
After discussing clozapine’s risks and benefits with Mr. Z and his parents, we start the medication at 25 mg/d to gauge tolerability, then titrate to 300 mg/d over 10 days. Mr. Z tolerates clozapine well, with some sedation and sialorrhea. A blood test taken 7 days after we start clozapine shows a normal white blood cell count.
After 10 days on clozapine, Mr. Z’s delusions and hallucinations are considerably less intense. He is no longer suicidal and visits his former college with his parents without thinking about his past acquaintance. We discharge him on clozapine, 300 mg/d, and refer him to the local mental health agency.
Two days later, Mr. Z’s parents report that since discharge their son has had extreme fatigue, shortness of breath, leg edema, and chest pain. We advise them to immediately take their son to the ER for cardiac workup.
The authors’ observations
Mr. Z’s sudden-onset physical symptoms suggest myocarditis, a rare but potentially fatal side effect of clozapine whose specific cause is unclear (Box 1).5-14 Myocarditis has been reported in 0.02% to 0.18% of patients exposed to clozapine,15-18 with incidence as high as 1.3% per 235 patients.19
Affected patients typically have been taking clozapine at therapeutic dosages (100 to 450 mg/d).7 Clozapine use is most prevalent among men ages 20 to 40, who tend to have more severe schizophrenia and lower cardiac risk than other populations. Correspondingly, clozapine-induced myocarditis is most prevalent in younger men,20 although what specifically causes this susceptibility is unknown.
Nonspecific symptoms such as dyspnea, tachycardia, chest pain, or fever can signal myocarditis (Table)7,21 and can surface within 4 to 8 weeks of starting clozapine.22 Haas et al20 reported other symptoms—such as leukocytosis—in young (median age 30), predominantly male patients with clozapine-induced myocarditis. Symptoms that typically occur during clozapine titration—such as fever and tachycardia—can mask “subclinical” myocarditis.22
Mr. Z’s nonspecific symptoms could signal clozapine-induced agranulocytosis or a viral syndrome, or could be delusional. The patient’s acute, sudden symptom onset strongly suggests a cardiac cause. Also, his delusions subsided, and normal blood readings helped us rule out agranulocytosis.
Coulter et al23 associated myocarditis and cardiomyopathy, a noninflammatory heart muscle disease, with several antipsychotics—including clozapine, chlorpromazine, fluphenazine, haloperidol, and risperidone—as well as lithium. More research is needed to confirm this association.
Order a cardiology consult and workup including:
- serum electrolytes
- complete blood count
- ECG21
- tests for myocardial damage including creatine kinase with MB fractionation (CK-MB) and testing for serum troponin I,25 lactic dehydrogenase, and aspartate transaminase (SGOT)21
- assessment for immune activation and peripheral eosinophilia.25
Table
Symptoms that could signal myocarditis in patients taking clozapine
|
| Source: Reference 7 |
TESTING: ‘Is this necessary?’
We contact the ER physician to request the above-mentioned tests, but he questions the need for such extensive and costly testing in a psychiatric patient with nonspecific symptoms.
After several phone conversations to review our recommendations, the emergency physician suggests sending Mr. Z home on a watch-and-wait protocol. We politely but firmly emphasize that Mr. Z needs a full cardiac workup, after which the physician consents to the tests (Box 2).
FINDINGS: suspicious readings
Mr. Z’s cardiac imaging results suggest a cardiopathy:
- echocardiogram shows mild ventricular enlargement with a decreased ejection fraction of 45% (normal reading, 55% to 60%)
- ECG shows normal sinus rhythm with low-voltage diffuse T-wave flattening throughout all leads without ST elevation
- creatine phosphokinase (CPK) and CKMB are within normal ranges
- troponin I is 0.33 ng/mL, a high-normal reading.
Based on these readings, the cardiology service admits Mr. Z with a presumptive diagnosis of clozapine-induced cardiomyopathy. The attending cardiologist stops clozapine and starts the angiotensin-converting enzyme inhibitor enalapril, 2.5 mg bid, for ventricular remodeling. Medical workup includes cytologic testing to rule out immunologic or viral disease.
Five days later, Mr. Z’s cardiac symptoms have resolved. The cardiology unit discharges him on enalapril, 2.5 mg bid, and schedules a cardiac ultrasound for 2 weeks after discharge to confirm progress.
The authors’ observations
Maintain high clinical suspicion while using clozapine. Similar to other patients with a clozapine-induced cardiopathy,16 Mr. Z showed rapid symptomatic changes after a benign initial course and experienced fairly vague symptoms that raised limited clinical concern at first.
Before starting clozapine therapy, screen all patients for pre-existing cardiac disease, which contraindicates this medication. Alert patients and caregivers to the risks and symptoms that require close monitoring early in treatment.
Many researchers suggest monitoring for myocarditis during the first month of therapy and ordering ECG at baseline and 2 and 4 weeks after starting clozapine.21,22 Berk et al26 suggest more aggressive monitoring, including:
- baseline ECG
- transthoracic echocardiogram
- baseline troponin/CK-MB
- ECG and troponin/CK-MB at 7 and 14 days
- echocardiogram at 6 and 12 months and then annually.
RELAPSE: Return of the ‘freak’
Immediately after Mr. Z’s discharge from the cardiology unit, we readmit him to inpatient psychiatry. His parents and case manager say he is again becoming preoccupied with his brief college relationship. He has been off clozapine for 5 days.
The authors’ observations
The American Psychiatric Association27 (see http://www.psych.org/psych_pract/treatg/pg/SchizPG-Complete-Feb04.pdf) recommends maximizing 1 medication for at least 2 to 4 weeks to assess schizophrenia symptom response and urges clinicians to consider adverse effects, medical comorbidities, and patient preference before continuing the medication.
These recommendations highlight the challenges of treating medication-resistant schizophrenia. Relapse is common after a serious reaction to clozapine, and combining 2 or more other antipsychotics could lead to significantly greater side effects. A time-limited trial with an antipsychotic and an adjunctive agent might be attempted while carefully weighing the combination’s risks and benefits.27
Clozapine reduced Mr. Z’s psychosis, but rechallenge would likely cause his potentially fatal cardiomyopathy to re-emerge. His sensitivity to adverse antipsychotic effects discourages polypharmacy and further complicates our decision.
How to convince other specialists
Many physicians are reluctant to pursue additional tests or procedures—and risk a confrontation with a consultant, insurer, or ER physician—especially when the risk of abnormality is extremely low. Advocating for cardiac workup in patients with vague symptoms is challenging, particularly if the suspected side effect is rare.
Taking the path of least resistance can increase the risk of a serious—albeit rare—adverse event. Failure to test could prolong a potentially harmful treatment, and the test results—even if negative—could be critical to planning care.
Calmly but firmly spell out the risks of missing a suspected cardiac problem (death, proceeding with potentially harmful treatment). Tell the ER manager or consultant, “I realize this is a very rare side effect, but not catching it could be life-threatening.”
Be circumspect when pleading your case—an overaggressive approach might cause the ER doctor to “dig in his heels” and reject your request. Use a medically focused response such as, “This is a known complication of this medicine with this common time course and presentation.”
TREATMENT: Another trial
We start olanzapine, 5 mg/d, and titrate to 20 mg/d over 1 week. We add sustained-release bupropion, 200 mg bid, for associated dysphoria.
Mr. Z’s symptoms and paranoia gradually decline, and he tolerates off-unit passes with friends and family before discharge. Staff works closely with him to develop cognitive-behavioral strategies to manage residual paranoia and hallucinations, such as assessing evidence for his delusional beliefs and developing tools to distract him from remaining “voices.” He reports no cardiac symptoms and continues taking enalapril, 2.5 mg bid.
We discharge Mr. Z after 1 week, at which point he shows no suicidal or homicidal thoughts. Follow-up echocardiogram 2 weeks later shows ejection fraction has improved to 60%, suggesting absence of cardiomyopathy.
Related resource
- Clozapine safety information.
www.clozaril.com/.
- Bupropion • Wellbutrin
- Chlorpromazine • Thorazine
- Clozapine • Clozaril
- Enalapril • Vasotec
- Fluphenazine • Prolixin, Permitil
- Haloperidol • Haldol
- Lithium • Eskalith, others
- Olanzapine • Zyprexa
- Propranolol • Inderal
- Quetiapine • Seroquel
- Risperidone • Risperdal
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Kane J, Honigfeld G, Singer J, Meltzer H. Clozapine for the treatment-resistant schizophrenic: a double-blind comparison with chlorpromazine. Arch Gen Psychiatry 1988;45:789-96.
2. Meltzer HY, Alphs L, Green AI, et al. International Suicide Prevention Trial Study Group. Clozapine treatment for suicidality in schizophrenia: International Suicide Prevention Trial (InterSePT). Arch Gen Psychiatry 2003;60:82-91.
3. Stahl SM. Antipsychotic polypharmacy, part 1: therapeutic option or dirty little secret? J Clin Psychiatry 1999;60:425-6.
4. Clozaril monograph. Novartis Phamaceuticals Corp.; April 12, 2006. Available at http://www.novartis.ca/downloads/en/products/clozaril_scrip_e.pdf. Accessed August 13, 2007.
5. Mason JW, O’Connell JB, Herskowitz A, et al. A clinical trial of immunosuppressive therapy for myocarditis. N Engl J Med 1995;333:269.-
6. Physicians’ desk reference. 61st ed. Montvale, NJ: Thomson PDR; 2007.
7. Merrill DB, Dec GW, Goff DC. Adverse cardiac effects associated with clozapine. J Clin Psychopharmacol 2005;25:32-41.
8. Jenie LE. Cardiovascular toxicity with clozapine therapy. Riverview Hospital Pharmacy Newsletter 2002;22:1-3.
9. Devarajan S, Kutcher SP, Dursun SM. Clozapine and sudden death. Lancet 2000;355:841.-
10. Vaddadi KS, Soosai E, Vaddadi G. Low blood selenium concentrations in schizophrenic patients on clozapine. Br J Clin Pharmacol 2003;55:307-9.
11. Williams DP, O’Donnell CJ, Maggs JL, et al. Bioactivation of clozapine by murine cardiac tissue in vivo and in vitro. Chem Res Toxicol 2003;16:1359-64.
12. Fineschi V, Neri M, Riezzo I, Turillazzi E. Sudden cardiac death due to hypersensitivity myocarditis during clozapine treatment. Int J Legal Med 2004;118:307-9.
13. Kendell KR, Day JD, Hruban RH, et al. Intimate association of eosinophils to collagen bundles in eosinophilic myocarditis and ranitidine induced hypersensitivity myocarditis. Arch Pathol Lab Med 1995;119:1154-60.
14. Pollmacher T, Schuld A, Kraus T, et al. On the clinical relevance of clozapine-triggered release of cytokines and soluble cytokine-receptors [in German]. Fortschr Neurol Psychiatr 2001;69(suppl 2):S65-S74.
15. Killian JG, Kerr K, Lawrence C, Celermajer DS. Myocarditis and cardiomyopathy associated with clozapine. Lancet 1999;354:1841-5.
16. Committee on Safety of Medicines Myocarditis with antipsychotics: recent cases with clozapine (Clozaril). Curr Probl Pharmacovigilance 1993;19:9.-
17. Degner D, Bleich S, Grohmann R, et al. Myocarditis associated with clozapine treatment. Aust NZ J Psychiatry 2000;34:880.-
18. La Grenade L, Graham D, Trontell A. Myocarditis and cardiomyopathy associated with clozapine use in the United States (letter). N Engl J Med 2001;345:224-5.
19. Reinders J, Parsonage W, Lange D, et al. Clozapinerelated myocarditis and cardiomyopathy in an Australian metropolitan psychiatric service. Aust NZ J Psychiatry 2004;38:915-22.
20. Haas SJ, Hill R, Krum H, et al. Clozapine-associated myocarditis: a review of 116 cases of suspected myocarditis associated with the use of clozapine in Australia during 1993-2003. Drug Saf 2007;30:47-57.
21. Wehmeier PM, Heiser P, Remschmidt H. Myocarditis, pericarditis and cardiomyopathy in patients treated with clozapine. J Clin Pharm Ther 2005;30:91-6.
22. Merrill DB, Ahmari SE, Bradford JM, Lieberman JA. Myocarditis during clozapine treatment. Am J Psychiatry 2006;163:204-8. Erratum in Am J Psychiatry 2006;163:556.-
23. Coulter DM, Bate A, Meyboom RH, et al. Antipsychotic drugs and heart muscle disorder in international pharmacovigilance: data mining study. BMJ 2001;322:1207-9.
24. Wooltorton E. Antipsychotic clozapine (Clozaril): myocarditis and cardiovascular toxicity. CMAJ 2002;166:1185-6.
25. Kay SE, Doery J, Sholl D. Clozapine associated pericarditis and elevated troponin I. Aust NZ J Psychiatry 2002;36:143-4.
26. Berk M, Fitzsimons J, Lambert T, et al. Monitoring the safe use of clozapine: a consensus view from Victoria, Australia. CNS Drugs 2007;21:117-27.
27. American Psychiatric Association Work Group on Schizophrenia, Lehman AF, chair. Practice guideline for the treatment of patients with schizophrenia, 2nd ed, 2004. Available at: http://www.psych.org/psych_pract/treatg/pg/SchizPG-Complete-Feb04.pdf. Accessed August 15, 2007.
1. Kane J, Honigfeld G, Singer J, Meltzer H. Clozapine for the treatment-resistant schizophrenic: a double-blind comparison with chlorpromazine. Arch Gen Psychiatry 1988;45:789-96.
2. Meltzer HY, Alphs L, Green AI, et al. International Suicide Prevention Trial Study Group. Clozapine treatment for suicidality in schizophrenia: International Suicide Prevention Trial (InterSePT). Arch Gen Psychiatry 2003;60:82-91.
3. Stahl SM. Antipsychotic polypharmacy, part 1: therapeutic option or dirty little secret? J Clin Psychiatry 1999;60:425-6.
4. Clozaril monograph. Novartis Phamaceuticals Corp.; April 12, 2006. Available at http://www.novartis.ca/downloads/en/products/clozaril_scrip_e.pdf. Accessed August 13, 2007.
5. Mason JW, O’Connell JB, Herskowitz A, et al. A clinical trial of immunosuppressive therapy for myocarditis. N Engl J Med 1995;333:269.-
6. Physicians’ desk reference. 61st ed. Montvale, NJ: Thomson PDR; 2007.
7. Merrill DB, Dec GW, Goff DC. Adverse cardiac effects associated with clozapine. J Clin Psychopharmacol 2005;25:32-41.
8. Jenie LE. Cardiovascular toxicity with clozapine therapy. Riverview Hospital Pharmacy Newsletter 2002;22:1-3.
9. Devarajan S, Kutcher SP, Dursun SM. Clozapine and sudden death. Lancet 2000;355:841.-
10. Vaddadi KS, Soosai E, Vaddadi G. Low blood selenium concentrations in schizophrenic patients on clozapine. Br J Clin Pharmacol 2003;55:307-9.
11. Williams DP, O’Donnell CJ, Maggs JL, et al. Bioactivation of clozapine by murine cardiac tissue in vivo and in vitro. Chem Res Toxicol 2003;16:1359-64.
12. Fineschi V, Neri M, Riezzo I, Turillazzi E. Sudden cardiac death due to hypersensitivity myocarditis during clozapine treatment. Int J Legal Med 2004;118:307-9.
13. Kendell KR, Day JD, Hruban RH, et al. Intimate association of eosinophils to collagen bundles in eosinophilic myocarditis and ranitidine induced hypersensitivity myocarditis. Arch Pathol Lab Med 1995;119:1154-60.
14. Pollmacher T, Schuld A, Kraus T, et al. On the clinical relevance of clozapine-triggered release of cytokines and soluble cytokine-receptors [in German]. Fortschr Neurol Psychiatr 2001;69(suppl 2):S65-S74.
15. Killian JG, Kerr K, Lawrence C, Celermajer DS. Myocarditis and cardiomyopathy associated with clozapine. Lancet 1999;354:1841-5.
16. Committee on Safety of Medicines Myocarditis with antipsychotics: recent cases with clozapine (Clozaril). Curr Probl Pharmacovigilance 1993;19:9.-
17. Degner D, Bleich S, Grohmann R, et al. Myocarditis associated with clozapine treatment. Aust NZ J Psychiatry 2000;34:880.-
18. La Grenade L, Graham D, Trontell A. Myocarditis and cardiomyopathy associated with clozapine use in the United States (letter). N Engl J Med 2001;345:224-5.
19. Reinders J, Parsonage W, Lange D, et al. Clozapinerelated myocarditis and cardiomyopathy in an Australian metropolitan psychiatric service. Aust NZ J Psychiatry 2004;38:915-22.
20. Haas SJ, Hill R, Krum H, et al. Clozapine-associated myocarditis: a review of 116 cases of suspected myocarditis associated with the use of clozapine in Australia during 1993-2003. Drug Saf 2007;30:47-57.
21. Wehmeier PM, Heiser P, Remschmidt H. Myocarditis, pericarditis and cardiomyopathy in patients treated with clozapine. J Clin Pharm Ther 2005;30:91-6.
22. Merrill DB, Ahmari SE, Bradford JM, Lieberman JA. Myocarditis during clozapine treatment. Am J Psychiatry 2006;163:204-8. Erratum in Am J Psychiatry 2006;163:556.-
23. Coulter DM, Bate A, Meyboom RH, et al. Antipsychotic drugs and heart muscle disorder in international pharmacovigilance: data mining study. BMJ 2001;322:1207-9.
24. Wooltorton E. Antipsychotic clozapine (Clozaril): myocarditis and cardiovascular toxicity. CMAJ 2002;166:1185-6.
25. Kay SE, Doery J, Sholl D. Clozapine associated pericarditis and elevated troponin I. Aust NZ J Psychiatry 2002;36:143-4.
26. Berk M, Fitzsimons J, Lambert T, et al. Monitoring the safe use of clozapine: a consensus view from Victoria, Australia. CNS Drugs 2007;21:117-27.
27. American Psychiatric Association Work Group on Schizophrenia, Lehman AF, chair. Practice guideline for the treatment of patients with schizophrenia, 2nd ed, 2004. Available at: http://www.psych.org/psych_pract/treatg/pg/SchizPG-Complete-Feb04.pdf. Accessed August 15, 2007.
Paliperidone ER: Reformulated antipsychotic for schizophrenia Tx
In the 9 months since paliperidone extended-release was FDA-approved for schizophrenia, the 3 acute pivotal trials supporting its approval have been published.1-3 They join a handful of post hoc analyses of this second-generation antipsychotic (SGA) in schizophrenia subgroups, including patients over age 65, recently diagnosed patients, and those with predominant negative symptoms.
This article discusses the evidence and paliperidone ER’s probable clinical benefits and adverse effects, with focus on its:
- pharmacodynamics and pharmacokinetics
- potential efficacy in schizophrenia and for specific patients and symptoms
- safety and tolerability.
How does paliperidone ER work?
Paliperidone ER was approved for schizophrenia treatment in December 2006 based on three 6-week, randomized, placebo-controlled trials. Paliperidone ER is the active metabolite of risperidone (9-OH risperidone) delivered in a once-daily, time-released formulation (Table 1).
Pharmacodynamics. Similar to risperidone, paliperidone ER has high binding affinity for dopamine (D2) and serotonin (5-HT2A) receptors, with additional affinity for histaminic (H1) and adrenergic receptors (alpha1 and alpha2) but not for muscarinic-cholinergic receptors.
Pharmacokinetics. After oral administration, the medication is widely and rapidly distributed. The drug’s terminal half-life is about 23 hours, and steady-state concentration is reached in 4 to 5 days.4,5
Thus paliperidone ER—when compared with risperidone and other antipsychotics that are metabolized primarily in the liver—is less likely to be involved in hepatic drug-drug or drug-disease interactions. However, some drugs that can induce CYP-450 enzymes—such as carbamazepine—may affect paliperidone’s metabolism.7
Paliperidone has an osmotic controlled-release oral delivery system (OROS®) for steady medication delivery across 24 hours8 (Table 2).1-3 The tablet consists of an osmotically active tri-layer core surrounded by a semipermeable membrane. When the tablet is swallowed, the membrane controls the rate of water reaching the tablet core, which determines the rate of drug delivery.6 The result is less variation between peak and trough drug concentrations, compared with immediate-release formulations.
Table 1
How paliperidone ER compares with risperidone
| Characteristic | Paliperidone ER | Risperidone |
|---|---|---|
| Formulation | OROS extended-release | Immediate release |
| Active moiety | 9-OH risperidone | Risperidone plus 9-OH risperidone |
| Metabolism | Primarily renal | Primarily hepatic |
| Drug interactions | Minimal | Primarily through cytochrome P-450 enzyme 2D6 |
| Dosing | Start at target dose | Titrate to target dose |
| OROS: osmotic controlled-release oral delivery system | ||
Paliperidone ER’s clinical characteristics
| Second-generation antipsychotic approved for schizophrenia |
| 9-OH active metabolite of risperidone |
| Osmotic controlled-release system provides steady-state drug delivery over 24 hours |
| Terminal half-life (time for 50% of drug to be eliminated from the body) ~23 hours |
| Available in 3-mg, 6-mg, and 9-mg tablets; recommended starting dose is 6 mg/d, and labeled dose range is 3 to 12 mg/d |
| Excreted primarily by the kidney; maximum recommended dose for patients with oderate to severe renal impairment is 3 mg/d |
| Source: References 1-3 |
Clinical use
Paliperidone ER offers potential therapeutic benefits in treating schizophrenia patients, although not without the risk of adverse events such as extrapyramidal symptoms (EPS) (Table 3).1-3
Patient selection. Because of its slow-release formulation and relatively stable plasma concentrations, paliperidone ER might be useful for patients who are highly sensitive to antipsychotics’ side effects. In particular, paliperidone ER might be ideal for patients who respond to but may not tolerate risperidone.
Paliperidone ER appears to be safe in patients with liver disease. Its primary renal excretion should minimize the risk of hepatic-related drug interactions in patients taking multiple medications.
Dosage and titration. For treating schizophrenia, the suggested starting dose of paliperidone ER is 6 mg/d taken in the morning. In the 3 pivotal trials, 6 mg was the lowest dose to show broad efficacy with minimal adverse events.9
For many patients, the 6-mg starting dose will be the therapeutic dose. When needed, the dose may be increased in 3-mg increments every 1 to 2 weeks to a maximum 12 mg/d (a 15-mg dose was used in clinical trials, but the adverse effects out-weighed the benefits). Lower maximum doses are recommended for patients with renal impairment:
- 6 mg/d for those with creatinine clearance ≥50 to
- 3 mg/d for those with creatinine clearance 10 to 10
Safety and tolerability. Pooled data from the 3 trials indicate that adverse events (AEs) occurred during treatment in 66% to 77% of patients receiving paliperidone ER vs 66% in placebo groups. The most common AEs were headache (11% to 18%), insomnia (4% to 12%), and anxiety (6% to 9%).9
EPS. Risk of EPS-related AEs (such as akathisia and parkinsonian symptoms) with 3-mg and 6-mg paliperidone ER doses (13% and 10%, respectively) was similar to placebo (11%) but increased with the 9-mg, 12-mg, and 15-mg doses (25%, 26%, and 24%, respectively). Should EPS occur, reduce the paliperidone ER dose or consider adding antiparkinsonian medications.
Lab values. No clinically relevant changes were noted in blood glucose, insulin, or lipids.12 Similar to risperidone, paliperidone ER elevated prolactin levels.
Weight gain with paliperidone ER is dose-dependent; in the clinical trials, mean body weight change for all doses was ≤1.9 kg, which is similar to the weight gain seen with risperidone and in the moderate range compared with other SGAs. When using paliperidone ER, follow the American Diabetes Association/American Psychiatric Association guidelines13 for monitoring weight gain and metabolic parameters with antipsychotics. Also monitor patients for clinical symptoms of hyperprolactinemia, and—if intolerable—adjust the dose or switch to another SGA.
Tachycardia. Advise patients that they may experience a rapid heart rate while taking paliperidone ER. In clinical trials, tachycardia occurred in ≤14% of patients—twice the rate with placebo—but did not contribute to more serious cardiac rhythm disturbances or to discontinuation. Incidence of prolonged corrected QT interval (QTc) was 3% to 5% in the paliperidone ER group vs 3% in the placebo group.
Patient education. Because of paliperidone ER’s pharmacokinetic properties, counsel patients to:
- take 1 tablet each day in the morning
- not chew, split, or crush the tablets but swallow whole to preserve the controlled-release delivery.
Table 3
Paliperidone ER’s potential benefits and risks in clinical practice
| Potential benefits | Details |
|---|---|
| Efficacy | Data support acute (6 weeks) and chronic (up to 24 weeks) improvement in schizophrenia symptoms, patient function, and quality of life |
| Pharmacokinetics | Primarily renal excretion decreases risk of hepatic drug-drug or drug-disease interactions |
| Long-acting formulation | Once-daily dosing simplifies treatment and may improve adherence |
| EPS | Risk similar to placebo at 3-mg and 6-mg doses, but increased at higher doses |
| Weight gain | Similar to risperidone |
| Hyperprolactinemia | Similar to risperidone |
| Tachycardia | Occurred in up to 14% of patients in clinical trials (twice the rate of placebo [7%]) |
| QTc prolongation | Increase up to 12 msec on average, with no patients exceeding 500 msec and no clinically adverse events during trials; use paliperidone with caution in patients with arrhythmias or cardiovascular disease or who are taking other medication that can prolong the QT interval |
| EPS: extrapyramidal symptoms | |
| Source: References 1-3 | |
Efficacy trials in schizophrenia
Three 6-week trials1-3 examined paliperidone ER’s efficacy in a total of 1,692 patients with chronic schizophrenia who were hospitalized ≥14 days with acute exacerbations. The trials were double-blind, randomized, fixed-dose, parallel-group, and placebo- and active-controlled (compared with olanzapine, 10 mg/d). Patients showed no significant differences in demographic or baseline characteristics or in the use of rescue medications.
The primary outcome measure was mean change in Positive and Negative Syndrome Scale (PANSS) total score, which quantifies positive, negative, and global psychopathologic symptom severity. Secondary outcome measures included:
- PANSS Marder factor scores14 (derived from PANSS items that reflect positive and negative symptoms, anxiety and depression, hostility, and thought disorganization).
- Clinical Global Impressions-Severity (CGI-S) score, which measures overall illness severity.15
- Personal and Social Performance (PSP) scores, which rate socially useful activities, relationships, self-care, and disturbing and aggressive behaviors; improvement by 1 category (10 points) reflects a clinically meaningful change.16,17
A total of 43% of patients completed the study—34% taking placebo; 46% taking paliperidone ER, 6 mg; 48% taking paliperidone ER, 12 mg; and 45% taking olanzapine. Demographic and baseline characteristics of the 432 patients who received ≥1 dose were similar across all groups. Approximately 75% of patients in each group used rescue medications—primarily lorazepam—for agitation, restlessness, or insomnia for a mean of 8 days.
Patients taking either paliperidone ER dose showed statistically significant greater improvement in PANSS total score compared with those taking placebo (6 mg, P = 0.006; 12 mg, P
Clinical response rates were similar with the 6-mg and 12-mg paliperidone ER doses—50% and 51%, respectively—and greater than with placebo (34%). The higher response rates with paliperidone ER were statistically significant compared with placebo (6 mg, P
Discontinuation rates for lack of efficacy were lower with paliperidone ER (6 mg, 23%; 12 mg, 14%) than with placebo (35%). A substantially lower percentage of patients taking this agent remained classified as “marked/severe/extremely severe” on the CGI-S score from baseline to endpoint, compared with the placebo group;
- 6 mg paliperidone ER, 58% to 26%
- 12 mg paliperidone ER, 64% to 21%
- placebo, 60% to 45%.
The second study2 included U.S. and international sites and compared 3 fixed doses of paliperidone ER (6-, 9-, and 12-mg) with placebo. Among the 630 patients enrolled, 66% completed the study. Patients were randomly assigned to 6 mg, 9 mg, or 12 mg of paliperidone ER; 10 mg of olanzapine; or placebo. The number of patients who dropped out because of adverse events was comparable across the groups.
Patient groups assigned to paliperidone ER showed significant improvement when compared with placebo (P 30% reduction in PANSS total score from baseline to endpoint included:
- 6 mg paliperidone ER, 56%
- 9 mg paliperidone ER, 51%
- 12 mg paliperidone ER, 61%
- placebo, 30%.
- 6 mg paliperidone ER, 63% at baseline to 22% at endpoint
- 9 mg paliperidone ER, 58% to 23%
- 12 mg paliperidone ER, 64% to 16%
- placebo, 60% to 51%.
The third study3 was a multicenter international trial that compared 3 fixed doses of paliperidone ER (3, 9, and 15 mg) with placebo. Among the 618 randomized patients, 365 (59%) completed the study: 70 of 127 (55%) on 3-mg paliperidone ER, 78 of 125 (62%) on 9-mg paliperidone ER, 82 of 115 (71%) on 15-mg paliperidone ER, and 47 of 123 (38%) on placebo.
- 3 mg paliperidone ER, 40%
- 9 mg paliperidone ER, 46%
- 15 mg paliperidone ER, 53%
- placebo, 18% (P ≤0.005).
- 3 mg paliperidone ER, 54% to 32%
- 9 mg paliperidone ER, 52% to 23%
- 15 mg paliperidone ER, 57% to 17%
- placebo, 56% to 50%.
Additional trial evidence
Schizophrenia subpopulations. Post hoc analyses of data reported from the 3 pivotal trials suggest that paliperidone ER may be useful for specific groups of schizophrenia patients, including those who are recently diagnosed, age >65, or severely ill or have predominant negative symptoms or sleep problems (Table 4).18-23
Efficacy in delaying recurrence. Paliperidone ER’s efficacy in delaying symptom recurrence was examined in a randomized, double-blind, placebo-controlled study of 207 patients who had been stabilized on open-label, flexible-dosed paliperidone ER.24 Time to first recurrence of schizophrenia symptoms was the primary efficacy measure. Starting dose was 9 mg/d (flexible dose range 3 to 15 mg/d).
The study was halted at a planned interim analysis because time-to-recurrence was significantly longer for patients receiving paliperidone ER compared with placebo (P
Final analysis of the 179 patients who completed the study confirmed the interim findings. Ongoing treatment maintained improvement in patients’ acute symptoms, functioning, and quality-of-life measures.
Table 4
Studies of paliperidone ER in schizophrenia subpopulations
| Patient population | Study design | Findings |
|---|---|---|
| Recently diagnosed | 413 patients diagnosed within 5 years of study entry compared with 893 patients who had been ill ≥5 years*18,19 | Tolerability was similar, but recently diagnosed patients were more likely to experience movement disorders and somnolence |
| Age ≥65 years | 114 schizophrenia patients age ≥65 given paliperidone ER, 3 to 12 mg/d, or placebo in 6-week, double-blind, randomized, placebo-controlled trial20 | Rates of cardiovascular, cerebrovascular, neuromotor, and metabolic changes similar to placebo, except for tachycardia (16% with paliperidone vs 0% with placebo) |
| Severely ill | 217 patients with marked to severe symptoms (baseline total PANSS score ≥105)*21 | Patients treated with paliperidone showed significantly greater improvement vs placebo in mean total PANSS score (–26.7 vs –5.7) and other measures |
| Substantial negative symptoms | 299 patients with predominant negative symptoms from 3 acute efficacy trials*22 | Patients treated with paliperidone showed significant improvements vs placebo on primary and secondary measures of negative symptoms |
| Sleep problems | 36 patients age 18 to 45 diagnosed with schizophrenia and schizophrenia-related insomnia*23 | In stable patients, paliperidone improved sleep architecture, continuity, and patient-rated sleep quality without producing or worsening daytime sleepiness |
| * Studies marked with asterisks represent post hoc analyses of data from the 3 clinical trials on which the FDA based its approval of paliperidone ER. | ||
| PANSS: Positive and Negative Syndrome Scale | ||
- Paliperidone extended release. Prescribing information. www.invega.com.
- Johnson & Johnson. U.S. District Court upholds Risperdal® (risperidone) patent (press release). October 16, 2006. www.jnj.com/news/jnj_news/20061016_094453.htm.
- Carbamazepine • Tegretol
- Lorazepam • Ativan
- Olanzapine • Zyprexa
- Paliperidone ER • Invega
- Risperidone • Risperdal
Dr. Rado and Dr. Dowd receive research support from Neuronetics, sanofi-aventis, Janssen Pharmaceutica, and Solvay.
Dr. Janicak receives research support from Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, Neuronetics, Solvay, and sanofi-aventis. He is a consultant to Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, Neuronetics, and Solvay, and a speaker for Abbott Laboratories, Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, and Pfizer.
1. Marder S, Kramer M, Ford L, et al. Efficacy and safety of paliperidone extended-release tablets: results of a 6-week, randomized, placebo-controlled study. Biol Psychiatry 2007; Jun 27; Epub ahead of print.
2. Kane J, Canas F, Kramer M, et al. Treatment of schizophrenia with paliperidone extended-release tablets: a 6-week placebo-controlled trial. Schizophr Res 2007;90(1-3):147-61.
3. Davidson M, Emsley R, Kramer M, et al. Efficacy, safety and early response of paliperidone extended-release tablets (paliperidone ER): results of a 6-week, randomized, placebo-controlled study. Schizophr Res 2007;93(1-3):117-30.
4. Rossenu SAC, Rusch S, Janssens L, et al. Extended release formulation of paliperidone shows dose proportional pharmacokinetics. Presented at: Annual Meeting of the American Association of Pharmaceutical Scientists; October 29, 2006; San Antonio, TX.
5. Vermeir M, Boom S, Naessens I, et al. Absorption, metabolism, and excretion of a single oral dose of 14C-paliperidone 1 mg in healthy subjects. Eur Neuropsychopharmacol 2005;15(suppl):S648-9.
6. Conley R, Gupta SK, Sathyan G. Clinical spectrum of the osmotic-controlled release oral delivery system (OROS), an advanced oral delivery form. Curr Med Res Opin 2006;22(10):1879-92.
7. Spina E, Avenoso A, Facciola G, et al. Plasma concentrations of risperidone and 9-hydroxyrisperidone: effect of comedication with carbamazepine or valproate. Ther Drug Monit 2000;22(4):481-5.
8. Paliperidone extended release. Prescribing information. Available at: http://www.invega.com. Accessed August 8, 2007.
9. Meltzer H, Kramer M, Gassmann-Mayer C, et al. Efficacy and tolerability of oral paliperidone extended-release tablets in the treatment of acute schizophrenia: pooled data from three 6-week placebo controlled studies. Int J Neuropsychopharmacol 2006;9(suppl 1):S225.-
10. Thyssen A, Cleton A, Osselae NV, et al. Effects of renal impairment on the pharmacokinetic profile of paliperidone extended-release tablets. Clin Pharmacol Ther 2007. In press.
11. Thyssen A, Crauwels H, Cleton A, et al. Effects of hepatic impairment on the pharmacokinetics of paliperidone immediate-release. Presented at: 46th Annual Meeting of the New Clinical Drug Evaluation Unit (NCDEU); June 12-15, 2006; Boca Raton, FL.
12. Meyer J, Kramer M, Lane R, et al. Metabolic outcomes in patients with schizophrenia treated with oral paliperidone extended release tablets: pooled analysis of three 6 week placebo-controlled studies. Int J Neuropsychopharmacol 2006;9(suppl 1):S282.-
13. 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. J Clin Psychiatry 2004;65:267-72.
14. Marder SR, Davis JM, Chouinard G. The effects of risperidone on the five dimensions of schizophrenia derived by factor analysis: combined results of the North American trials. J Clin Psychiatry 1997;58:538-46.
15. Guy W. Clinical Global Impressions Scale. Early clinical drug evaluation unit (ECDEU) assessment manual for psychopharmacology. Rockville, MD: National Institute of Mental Health, Department of Health, Education, and Welfare; 1976:218-22. ADM publication 76-338.
16. Morosini PL, Magliano L, Brambilla L, et al. Development, reliability and acceptability of a new version of the DSMIV Social and Occupational Functioning Assessment Scale (SOFAS) to assess routine social functioning. Acta Psychiatr Scand 2000;101:323-9.
17. Patrick D, Adriaenssen I, Morosini P, Rothman M. Reliability, validity and sensitivity to change of the Personal and Social Performance scale in patients with acute schizophrenia. Int J Neuropsychopharmacol 2006;9(suppl 1):S287-8.
18. Kostic D, Bossie C, Turkoz I, et al. Paliperidone extended-release tablets in patients recently diagnosed with schizophrenia. Int J Neuropsychopharmacol 2006;9(suppl 1):S161.-
19. Kostic D, Bossie C, Turkoz I, et al. Paliperidone extended-release tablets in patients recently diagnosed with schizophrenia. Presented at: Congress of the Collegium Internationale Neruo-Psychopharmacologicum (CINP); July 9-13, 2006; Chicago, IL.
20. Tzimos A, Kramer M, Ford L, et al. A 6-week placebo-controlled study of the safety and tolerability of flexible doses of oral paliperidone extended release tablets in the treatment of schizophrenia in elderly patients. Int J Neuropsychopharmacol 2006;9(suppl 1):S155.-
21. Canuso C, Youssef E, Dirks B, et al. Paliperidone extended-release in severely-ill patients with schizophrenia. Presented at: 58th Annual Institute on Psychiatric Services; October 5-8, 2006; New York, NY.
22. Dirks B, Eerdekens M, Turkoz I, et al. Efficacy of paliperidone extended-release tablets in patients with schizophrenia and predominant negative symptoms. Int J Neuropsychopharmacol 2006;9(suppl 1):S162.-
23. Luthringer R, Staner L, Noel N, et al. Sleep assessments in patients with schizophrenia following treatment with paliperidone extended-release tablets. Eur Neuropsychopharmacol 2006;16(suppl 4):S224.-
24. Kramer M, Simpson G, Maciulis V, et al. Paliperidone extended-release tablets for prevention of symptom recurrence in patients with schizophrenia: a randomized double-blind, placebo-controlled study [published correction appears in J Clin Psychopharmacol. 2007;27(3):258]. J Clin Psychopharmacol 2007;27(1):6-14.
In the 9 months since paliperidone extended-release was FDA-approved for schizophrenia, the 3 acute pivotal trials supporting its approval have been published.1-3 They join a handful of post hoc analyses of this second-generation antipsychotic (SGA) in schizophrenia subgroups, including patients over age 65, recently diagnosed patients, and those with predominant negative symptoms.
This article discusses the evidence and paliperidone ER’s probable clinical benefits and adverse effects, with focus on its:
- pharmacodynamics and pharmacokinetics
- potential efficacy in schizophrenia and for specific patients and symptoms
- safety and tolerability.
How does paliperidone ER work?
Paliperidone ER was approved for schizophrenia treatment in December 2006 based on three 6-week, randomized, placebo-controlled trials. Paliperidone ER is the active metabolite of risperidone (9-OH risperidone) delivered in a once-daily, time-released formulation (Table 1).
Pharmacodynamics. Similar to risperidone, paliperidone ER has high binding affinity for dopamine (D2) and serotonin (5-HT2A) receptors, with additional affinity for histaminic (H1) and adrenergic receptors (alpha1 and alpha2) but not for muscarinic-cholinergic receptors.
Pharmacokinetics. After oral administration, the medication is widely and rapidly distributed. The drug’s terminal half-life is about 23 hours, and steady-state concentration is reached in 4 to 5 days.4,5
Thus paliperidone ER—when compared with risperidone and other antipsychotics that are metabolized primarily in the liver—is less likely to be involved in hepatic drug-drug or drug-disease interactions. However, some drugs that can induce CYP-450 enzymes—such as carbamazepine—may affect paliperidone’s metabolism.7
Paliperidone has an osmotic controlled-release oral delivery system (OROS®) for steady medication delivery across 24 hours8 (Table 2).1-3 The tablet consists of an osmotically active tri-layer core surrounded by a semipermeable membrane. When the tablet is swallowed, the membrane controls the rate of water reaching the tablet core, which determines the rate of drug delivery.6 The result is less variation between peak and trough drug concentrations, compared with immediate-release formulations.
Table 1
How paliperidone ER compares with risperidone
| Characteristic | Paliperidone ER | Risperidone |
|---|---|---|
| Formulation | OROS extended-release | Immediate release |
| Active moiety | 9-OH risperidone | Risperidone plus 9-OH risperidone |
| Metabolism | Primarily renal | Primarily hepatic |
| Drug interactions | Minimal | Primarily through cytochrome P-450 enzyme 2D6 |
| Dosing | Start at target dose | Titrate to target dose |
| OROS: osmotic controlled-release oral delivery system | ||
Paliperidone ER’s clinical characteristics
| Second-generation antipsychotic approved for schizophrenia |
| 9-OH active metabolite of risperidone |
| Osmotic controlled-release system provides steady-state drug delivery over 24 hours |
| Terminal half-life (time for 50% of drug to be eliminated from the body) ~23 hours |
| Available in 3-mg, 6-mg, and 9-mg tablets; recommended starting dose is 6 mg/d, and labeled dose range is 3 to 12 mg/d |
| Excreted primarily by the kidney; maximum recommended dose for patients with oderate to severe renal impairment is 3 mg/d |
| Source: References 1-3 |
Clinical use
Paliperidone ER offers potential therapeutic benefits in treating schizophrenia patients, although not without the risk of adverse events such as extrapyramidal symptoms (EPS) (Table 3).1-3
Patient selection. Because of its slow-release formulation and relatively stable plasma concentrations, paliperidone ER might be useful for patients who are highly sensitive to antipsychotics’ side effects. In particular, paliperidone ER might be ideal for patients who respond to but may not tolerate risperidone.
Paliperidone ER appears to be safe in patients with liver disease. Its primary renal excretion should minimize the risk of hepatic-related drug interactions in patients taking multiple medications.
Dosage and titration. For treating schizophrenia, the suggested starting dose of paliperidone ER is 6 mg/d taken in the morning. In the 3 pivotal trials, 6 mg was the lowest dose to show broad efficacy with minimal adverse events.9
For many patients, the 6-mg starting dose will be the therapeutic dose. When needed, the dose may be increased in 3-mg increments every 1 to 2 weeks to a maximum 12 mg/d (a 15-mg dose was used in clinical trials, but the adverse effects out-weighed the benefits). Lower maximum doses are recommended for patients with renal impairment:
- 6 mg/d for those with creatinine clearance ≥50 to
- 3 mg/d for those with creatinine clearance 10 to 10
Safety and tolerability. Pooled data from the 3 trials indicate that adverse events (AEs) occurred during treatment in 66% to 77% of patients receiving paliperidone ER vs 66% in placebo groups. The most common AEs were headache (11% to 18%), insomnia (4% to 12%), and anxiety (6% to 9%).9
EPS. Risk of EPS-related AEs (such as akathisia and parkinsonian symptoms) with 3-mg and 6-mg paliperidone ER doses (13% and 10%, respectively) was similar to placebo (11%) but increased with the 9-mg, 12-mg, and 15-mg doses (25%, 26%, and 24%, respectively). Should EPS occur, reduce the paliperidone ER dose or consider adding antiparkinsonian medications.
Lab values. No clinically relevant changes were noted in blood glucose, insulin, or lipids.12 Similar to risperidone, paliperidone ER elevated prolactin levels.
Weight gain with paliperidone ER is dose-dependent; in the clinical trials, mean body weight change for all doses was ≤1.9 kg, which is similar to the weight gain seen with risperidone and in the moderate range compared with other SGAs. When using paliperidone ER, follow the American Diabetes Association/American Psychiatric Association guidelines13 for monitoring weight gain and metabolic parameters with antipsychotics. Also monitor patients for clinical symptoms of hyperprolactinemia, and—if intolerable—adjust the dose or switch to another SGA.
Tachycardia. Advise patients that they may experience a rapid heart rate while taking paliperidone ER. In clinical trials, tachycardia occurred in ≤14% of patients—twice the rate with placebo—but did not contribute to more serious cardiac rhythm disturbances or to discontinuation. Incidence of prolonged corrected QT interval (QTc) was 3% to 5% in the paliperidone ER group vs 3% in the placebo group.
Patient education. Because of paliperidone ER’s pharmacokinetic properties, counsel patients to:
- take 1 tablet each day in the morning
- not chew, split, or crush the tablets but swallow whole to preserve the controlled-release delivery.
Table 3
Paliperidone ER’s potential benefits and risks in clinical practice
| Potential benefits | Details |
|---|---|
| Efficacy | Data support acute (6 weeks) and chronic (up to 24 weeks) improvement in schizophrenia symptoms, patient function, and quality of life |
| Pharmacokinetics | Primarily renal excretion decreases risk of hepatic drug-drug or drug-disease interactions |
| Long-acting formulation | Once-daily dosing simplifies treatment and may improve adherence |
| EPS | Risk similar to placebo at 3-mg and 6-mg doses, but increased at higher doses |
| Weight gain | Similar to risperidone |
| Hyperprolactinemia | Similar to risperidone |
| Tachycardia | Occurred in up to 14% of patients in clinical trials (twice the rate of placebo [7%]) |
| QTc prolongation | Increase up to 12 msec on average, with no patients exceeding 500 msec and no clinically adverse events during trials; use paliperidone with caution in patients with arrhythmias or cardiovascular disease or who are taking other medication that can prolong the QT interval |
| EPS: extrapyramidal symptoms | |
| Source: References 1-3 | |
Efficacy trials in schizophrenia
Three 6-week trials1-3 examined paliperidone ER’s efficacy in a total of 1,692 patients with chronic schizophrenia who were hospitalized ≥14 days with acute exacerbations. The trials were double-blind, randomized, fixed-dose, parallel-group, and placebo- and active-controlled (compared with olanzapine, 10 mg/d). Patients showed no significant differences in demographic or baseline characteristics or in the use of rescue medications.
The primary outcome measure was mean change in Positive and Negative Syndrome Scale (PANSS) total score, which quantifies positive, negative, and global psychopathologic symptom severity. Secondary outcome measures included:
- PANSS Marder factor scores14 (derived from PANSS items that reflect positive and negative symptoms, anxiety and depression, hostility, and thought disorganization).
- Clinical Global Impressions-Severity (CGI-S) score, which measures overall illness severity.15
- Personal and Social Performance (PSP) scores, which rate socially useful activities, relationships, self-care, and disturbing and aggressive behaviors; improvement by 1 category (10 points) reflects a clinically meaningful change.16,17
A total of 43% of patients completed the study—34% taking placebo; 46% taking paliperidone ER, 6 mg; 48% taking paliperidone ER, 12 mg; and 45% taking olanzapine. Demographic and baseline characteristics of the 432 patients who received ≥1 dose were similar across all groups. Approximately 75% of patients in each group used rescue medications—primarily lorazepam—for agitation, restlessness, or insomnia for a mean of 8 days.
Patients taking either paliperidone ER dose showed statistically significant greater improvement in PANSS total score compared with those taking placebo (6 mg, P = 0.006; 12 mg, P
Clinical response rates were similar with the 6-mg and 12-mg paliperidone ER doses—50% and 51%, respectively—and greater than with placebo (34%). The higher response rates with paliperidone ER were statistically significant compared with placebo (6 mg, P
Discontinuation rates for lack of efficacy were lower with paliperidone ER (6 mg, 23%; 12 mg, 14%) than with placebo (35%). A substantially lower percentage of patients taking this agent remained classified as “marked/severe/extremely severe” on the CGI-S score from baseline to endpoint, compared with the placebo group;
- 6 mg paliperidone ER, 58% to 26%
- 12 mg paliperidone ER, 64% to 21%
- placebo, 60% to 45%.
The second study2 included U.S. and international sites and compared 3 fixed doses of paliperidone ER (6-, 9-, and 12-mg) with placebo. Among the 630 patients enrolled, 66% completed the study. Patients were randomly assigned to 6 mg, 9 mg, or 12 mg of paliperidone ER; 10 mg of olanzapine; or placebo. The number of patients who dropped out because of adverse events was comparable across the groups.
Patient groups assigned to paliperidone ER showed significant improvement when compared with placebo (P 30% reduction in PANSS total score from baseline to endpoint included:
- 6 mg paliperidone ER, 56%
- 9 mg paliperidone ER, 51%
- 12 mg paliperidone ER, 61%
- placebo, 30%.
- 6 mg paliperidone ER, 63% at baseline to 22% at endpoint
- 9 mg paliperidone ER, 58% to 23%
- 12 mg paliperidone ER, 64% to 16%
- placebo, 60% to 51%.
The third study3 was a multicenter international trial that compared 3 fixed doses of paliperidone ER (3, 9, and 15 mg) with placebo. Among the 618 randomized patients, 365 (59%) completed the study: 70 of 127 (55%) on 3-mg paliperidone ER, 78 of 125 (62%) on 9-mg paliperidone ER, 82 of 115 (71%) on 15-mg paliperidone ER, and 47 of 123 (38%) on placebo.
- 3 mg paliperidone ER, 40%
- 9 mg paliperidone ER, 46%
- 15 mg paliperidone ER, 53%
- placebo, 18% (P ≤0.005).
- 3 mg paliperidone ER, 54% to 32%
- 9 mg paliperidone ER, 52% to 23%
- 15 mg paliperidone ER, 57% to 17%
- placebo, 56% to 50%.
Additional trial evidence
Schizophrenia subpopulations. Post hoc analyses of data reported from the 3 pivotal trials suggest that paliperidone ER may be useful for specific groups of schizophrenia patients, including those who are recently diagnosed, age >65, or severely ill or have predominant negative symptoms or sleep problems (Table 4).18-23
Efficacy in delaying recurrence. Paliperidone ER’s efficacy in delaying symptom recurrence was examined in a randomized, double-blind, placebo-controlled study of 207 patients who had been stabilized on open-label, flexible-dosed paliperidone ER.24 Time to first recurrence of schizophrenia symptoms was the primary efficacy measure. Starting dose was 9 mg/d (flexible dose range 3 to 15 mg/d).
The study was halted at a planned interim analysis because time-to-recurrence was significantly longer for patients receiving paliperidone ER compared with placebo (P
Final analysis of the 179 patients who completed the study confirmed the interim findings. Ongoing treatment maintained improvement in patients’ acute symptoms, functioning, and quality-of-life measures.
Table 4
Studies of paliperidone ER in schizophrenia subpopulations
| Patient population | Study design | Findings |
|---|---|---|
| Recently diagnosed | 413 patients diagnosed within 5 years of study entry compared with 893 patients who had been ill ≥5 years*18,19 | Tolerability was similar, but recently diagnosed patients were more likely to experience movement disorders and somnolence |
| Age ≥65 years | 114 schizophrenia patients age ≥65 given paliperidone ER, 3 to 12 mg/d, or placebo in 6-week, double-blind, randomized, placebo-controlled trial20 | Rates of cardiovascular, cerebrovascular, neuromotor, and metabolic changes similar to placebo, except for tachycardia (16% with paliperidone vs 0% with placebo) |
| Severely ill | 217 patients with marked to severe symptoms (baseline total PANSS score ≥105)*21 | Patients treated with paliperidone showed significantly greater improvement vs placebo in mean total PANSS score (–26.7 vs –5.7) and other measures |
| Substantial negative symptoms | 299 patients with predominant negative symptoms from 3 acute efficacy trials*22 | Patients treated with paliperidone showed significant improvements vs placebo on primary and secondary measures of negative symptoms |
| Sleep problems | 36 patients age 18 to 45 diagnosed with schizophrenia and schizophrenia-related insomnia*23 | In stable patients, paliperidone improved sleep architecture, continuity, and patient-rated sleep quality without producing or worsening daytime sleepiness |
| * Studies marked with asterisks represent post hoc analyses of data from the 3 clinical trials on which the FDA based its approval of paliperidone ER. | ||
| PANSS: Positive and Negative Syndrome Scale | ||
- Paliperidone extended release. Prescribing information. www.invega.com.
- Johnson & Johnson. U.S. District Court upholds Risperdal® (risperidone) patent (press release). October 16, 2006. www.jnj.com/news/jnj_news/20061016_094453.htm.
- Carbamazepine • Tegretol
- Lorazepam • Ativan
- Olanzapine • Zyprexa
- Paliperidone ER • Invega
- Risperidone • Risperdal
Dr. Rado and Dr. Dowd receive research support from Neuronetics, sanofi-aventis, Janssen Pharmaceutica, and Solvay.
Dr. Janicak receives research support from Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, Neuronetics, Solvay, and sanofi-aventis. He is a consultant to Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, Neuronetics, and Solvay, and a speaker for Abbott Laboratories, Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, and Pfizer.
In the 9 months since paliperidone extended-release was FDA-approved for schizophrenia, the 3 acute pivotal trials supporting its approval have been published.1-3 They join a handful of post hoc analyses of this second-generation antipsychotic (SGA) in schizophrenia subgroups, including patients over age 65, recently diagnosed patients, and those with predominant negative symptoms.
This article discusses the evidence and paliperidone ER’s probable clinical benefits and adverse effects, with focus on its:
- pharmacodynamics and pharmacokinetics
- potential efficacy in schizophrenia and for specific patients and symptoms
- safety and tolerability.
How does paliperidone ER work?
Paliperidone ER was approved for schizophrenia treatment in December 2006 based on three 6-week, randomized, placebo-controlled trials. Paliperidone ER is the active metabolite of risperidone (9-OH risperidone) delivered in a once-daily, time-released formulation (Table 1).
Pharmacodynamics. Similar to risperidone, paliperidone ER has high binding affinity for dopamine (D2) and serotonin (5-HT2A) receptors, with additional affinity for histaminic (H1) and adrenergic receptors (alpha1 and alpha2) but not for muscarinic-cholinergic receptors.
Pharmacokinetics. After oral administration, the medication is widely and rapidly distributed. The drug’s terminal half-life is about 23 hours, and steady-state concentration is reached in 4 to 5 days.4,5
Thus paliperidone ER—when compared with risperidone and other antipsychotics that are metabolized primarily in the liver—is less likely to be involved in hepatic drug-drug or drug-disease interactions. However, some drugs that can induce CYP-450 enzymes—such as carbamazepine—may affect paliperidone’s metabolism.7
Paliperidone has an osmotic controlled-release oral delivery system (OROS®) for steady medication delivery across 24 hours8 (Table 2).1-3 The tablet consists of an osmotically active tri-layer core surrounded by a semipermeable membrane. When the tablet is swallowed, the membrane controls the rate of water reaching the tablet core, which determines the rate of drug delivery.6 The result is less variation between peak and trough drug concentrations, compared with immediate-release formulations.
Table 1
How paliperidone ER compares with risperidone
| Characteristic | Paliperidone ER | Risperidone |
|---|---|---|
| Formulation | OROS extended-release | Immediate release |
| Active moiety | 9-OH risperidone | Risperidone plus 9-OH risperidone |
| Metabolism | Primarily renal | Primarily hepatic |
| Drug interactions | Minimal | Primarily through cytochrome P-450 enzyme 2D6 |
| Dosing | Start at target dose | Titrate to target dose |
| OROS: osmotic controlled-release oral delivery system | ||
Paliperidone ER’s clinical characteristics
| Second-generation antipsychotic approved for schizophrenia |
| 9-OH active metabolite of risperidone |
| Osmotic controlled-release system provides steady-state drug delivery over 24 hours |
| Terminal half-life (time for 50% of drug to be eliminated from the body) ~23 hours |
| Available in 3-mg, 6-mg, and 9-mg tablets; recommended starting dose is 6 mg/d, and labeled dose range is 3 to 12 mg/d |
| Excreted primarily by the kidney; maximum recommended dose for patients with oderate to severe renal impairment is 3 mg/d |
| Source: References 1-3 |
Clinical use
Paliperidone ER offers potential therapeutic benefits in treating schizophrenia patients, although not without the risk of adverse events such as extrapyramidal symptoms (EPS) (Table 3).1-3
Patient selection. Because of its slow-release formulation and relatively stable plasma concentrations, paliperidone ER might be useful for patients who are highly sensitive to antipsychotics’ side effects. In particular, paliperidone ER might be ideal for patients who respond to but may not tolerate risperidone.
Paliperidone ER appears to be safe in patients with liver disease. Its primary renal excretion should minimize the risk of hepatic-related drug interactions in patients taking multiple medications.
Dosage and titration. For treating schizophrenia, the suggested starting dose of paliperidone ER is 6 mg/d taken in the morning. In the 3 pivotal trials, 6 mg was the lowest dose to show broad efficacy with minimal adverse events.9
For many patients, the 6-mg starting dose will be the therapeutic dose. When needed, the dose may be increased in 3-mg increments every 1 to 2 weeks to a maximum 12 mg/d (a 15-mg dose was used in clinical trials, but the adverse effects out-weighed the benefits). Lower maximum doses are recommended for patients with renal impairment:
- 6 mg/d for those with creatinine clearance ≥50 to
- 3 mg/d for those with creatinine clearance 10 to 10
Safety and tolerability. Pooled data from the 3 trials indicate that adverse events (AEs) occurred during treatment in 66% to 77% of patients receiving paliperidone ER vs 66% in placebo groups. The most common AEs were headache (11% to 18%), insomnia (4% to 12%), and anxiety (6% to 9%).9
EPS. Risk of EPS-related AEs (such as akathisia and parkinsonian symptoms) with 3-mg and 6-mg paliperidone ER doses (13% and 10%, respectively) was similar to placebo (11%) but increased with the 9-mg, 12-mg, and 15-mg doses (25%, 26%, and 24%, respectively). Should EPS occur, reduce the paliperidone ER dose or consider adding antiparkinsonian medications.
Lab values. No clinically relevant changes were noted in blood glucose, insulin, or lipids.12 Similar to risperidone, paliperidone ER elevated prolactin levels.
Weight gain with paliperidone ER is dose-dependent; in the clinical trials, mean body weight change for all doses was ≤1.9 kg, which is similar to the weight gain seen with risperidone and in the moderate range compared with other SGAs. When using paliperidone ER, follow the American Diabetes Association/American Psychiatric Association guidelines13 for monitoring weight gain and metabolic parameters with antipsychotics. Also monitor patients for clinical symptoms of hyperprolactinemia, and—if intolerable—adjust the dose or switch to another SGA.
Tachycardia. Advise patients that they may experience a rapid heart rate while taking paliperidone ER. In clinical trials, tachycardia occurred in ≤14% of patients—twice the rate with placebo—but did not contribute to more serious cardiac rhythm disturbances or to discontinuation. Incidence of prolonged corrected QT interval (QTc) was 3% to 5% in the paliperidone ER group vs 3% in the placebo group.
Patient education. Because of paliperidone ER’s pharmacokinetic properties, counsel patients to:
- take 1 tablet each day in the morning
- not chew, split, or crush the tablets but swallow whole to preserve the controlled-release delivery.
Table 3
Paliperidone ER’s potential benefits and risks in clinical practice
| Potential benefits | Details |
|---|---|
| Efficacy | Data support acute (6 weeks) and chronic (up to 24 weeks) improvement in schizophrenia symptoms, patient function, and quality of life |
| Pharmacokinetics | Primarily renal excretion decreases risk of hepatic drug-drug or drug-disease interactions |
| Long-acting formulation | Once-daily dosing simplifies treatment and may improve adherence |
| EPS | Risk similar to placebo at 3-mg and 6-mg doses, but increased at higher doses |
| Weight gain | Similar to risperidone |
| Hyperprolactinemia | Similar to risperidone |
| Tachycardia | Occurred in up to 14% of patients in clinical trials (twice the rate of placebo [7%]) |
| QTc prolongation | Increase up to 12 msec on average, with no patients exceeding 500 msec and no clinically adverse events during trials; use paliperidone with caution in patients with arrhythmias or cardiovascular disease or who are taking other medication that can prolong the QT interval |
| EPS: extrapyramidal symptoms | |
| Source: References 1-3 | |
Efficacy trials in schizophrenia
Three 6-week trials1-3 examined paliperidone ER’s efficacy in a total of 1,692 patients with chronic schizophrenia who were hospitalized ≥14 days with acute exacerbations. The trials were double-blind, randomized, fixed-dose, parallel-group, and placebo- and active-controlled (compared with olanzapine, 10 mg/d). Patients showed no significant differences in demographic or baseline characteristics or in the use of rescue medications.
The primary outcome measure was mean change in Positive and Negative Syndrome Scale (PANSS) total score, which quantifies positive, negative, and global psychopathologic symptom severity. Secondary outcome measures included:
- PANSS Marder factor scores14 (derived from PANSS items that reflect positive and negative symptoms, anxiety and depression, hostility, and thought disorganization).
- Clinical Global Impressions-Severity (CGI-S) score, which measures overall illness severity.15
- Personal and Social Performance (PSP) scores, which rate socially useful activities, relationships, self-care, and disturbing and aggressive behaviors; improvement by 1 category (10 points) reflects a clinically meaningful change.16,17
A total of 43% of patients completed the study—34% taking placebo; 46% taking paliperidone ER, 6 mg; 48% taking paliperidone ER, 12 mg; and 45% taking olanzapine. Demographic and baseline characteristics of the 432 patients who received ≥1 dose were similar across all groups. Approximately 75% of patients in each group used rescue medications—primarily lorazepam—for agitation, restlessness, or insomnia for a mean of 8 days.
Patients taking either paliperidone ER dose showed statistically significant greater improvement in PANSS total score compared with those taking placebo (6 mg, P = 0.006; 12 mg, P
Clinical response rates were similar with the 6-mg and 12-mg paliperidone ER doses—50% and 51%, respectively—and greater than with placebo (34%). The higher response rates with paliperidone ER were statistically significant compared with placebo (6 mg, P
Discontinuation rates for lack of efficacy were lower with paliperidone ER (6 mg, 23%; 12 mg, 14%) than with placebo (35%). A substantially lower percentage of patients taking this agent remained classified as “marked/severe/extremely severe” on the CGI-S score from baseline to endpoint, compared with the placebo group;
- 6 mg paliperidone ER, 58% to 26%
- 12 mg paliperidone ER, 64% to 21%
- placebo, 60% to 45%.
The second study2 included U.S. and international sites and compared 3 fixed doses of paliperidone ER (6-, 9-, and 12-mg) with placebo. Among the 630 patients enrolled, 66% completed the study. Patients were randomly assigned to 6 mg, 9 mg, or 12 mg of paliperidone ER; 10 mg of olanzapine; or placebo. The number of patients who dropped out because of adverse events was comparable across the groups.
Patient groups assigned to paliperidone ER showed significant improvement when compared with placebo (P 30% reduction in PANSS total score from baseline to endpoint included:
- 6 mg paliperidone ER, 56%
- 9 mg paliperidone ER, 51%
- 12 mg paliperidone ER, 61%
- placebo, 30%.
- 6 mg paliperidone ER, 63% at baseline to 22% at endpoint
- 9 mg paliperidone ER, 58% to 23%
- 12 mg paliperidone ER, 64% to 16%
- placebo, 60% to 51%.
The third study3 was a multicenter international trial that compared 3 fixed doses of paliperidone ER (3, 9, and 15 mg) with placebo. Among the 618 randomized patients, 365 (59%) completed the study: 70 of 127 (55%) on 3-mg paliperidone ER, 78 of 125 (62%) on 9-mg paliperidone ER, 82 of 115 (71%) on 15-mg paliperidone ER, and 47 of 123 (38%) on placebo.
- 3 mg paliperidone ER, 40%
- 9 mg paliperidone ER, 46%
- 15 mg paliperidone ER, 53%
- placebo, 18% (P ≤0.005).
- 3 mg paliperidone ER, 54% to 32%
- 9 mg paliperidone ER, 52% to 23%
- 15 mg paliperidone ER, 57% to 17%
- placebo, 56% to 50%.
Additional trial evidence
Schizophrenia subpopulations. Post hoc analyses of data reported from the 3 pivotal trials suggest that paliperidone ER may be useful for specific groups of schizophrenia patients, including those who are recently diagnosed, age >65, or severely ill or have predominant negative symptoms or sleep problems (Table 4).18-23
Efficacy in delaying recurrence. Paliperidone ER’s efficacy in delaying symptom recurrence was examined in a randomized, double-blind, placebo-controlled study of 207 patients who had been stabilized on open-label, flexible-dosed paliperidone ER.24 Time to first recurrence of schizophrenia symptoms was the primary efficacy measure. Starting dose was 9 mg/d (flexible dose range 3 to 15 mg/d).
The study was halted at a planned interim analysis because time-to-recurrence was significantly longer for patients receiving paliperidone ER compared with placebo (P
Final analysis of the 179 patients who completed the study confirmed the interim findings. Ongoing treatment maintained improvement in patients’ acute symptoms, functioning, and quality-of-life measures.
Table 4
Studies of paliperidone ER in schizophrenia subpopulations
| Patient population | Study design | Findings |
|---|---|---|
| Recently diagnosed | 413 patients diagnosed within 5 years of study entry compared with 893 patients who had been ill ≥5 years*18,19 | Tolerability was similar, but recently diagnosed patients were more likely to experience movement disorders and somnolence |
| Age ≥65 years | 114 schizophrenia patients age ≥65 given paliperidone ER, 3 to 12 mg/d, or placebo in 6-week, double-blind, randomized, placebo-controlled trial20 | Rates of cardiovascular, cerebrovascular, neuromotor, and metabolic changes similar to placebo, except for tachycardia (16% with paliperidone vs 0% with placebo) |
| Severely ill | 217 patients with marked to severe symptoms (baseline total PANSS score ≥105)*21 | Patients treated with paliperidone showed significantly greater improvement vs placebo in mean total PANSS score (–26.7 vs –5.7) and other measures |
| Substantial negative symptoms | 299 patients with predominant negative symptoms from 3 acute efficacy trials*22 | Patients treated with paliperidone showed significant improvements vs placebo on primary and secondary measures of negative symptoms |
| Sleep problems | 36 patients age 18 to 45 diagnosed with schizophrenia and schizophrenia-related insomnia*23 | In stable patients, paliperidone improved sleep architecture, continuity, and patient-rated sleep quality without producing or worsening daytime sleepiness |
| * Studies marked with asterisks represent post hoc analyses of data from the 3 clinical trials on which the FDA based its approval of paliperidone ER. | ||
| PANSS: Positive and Negative Syndrome Scale | ||
- Paliperidone extended release. Prescribing information. www.invega.com.
- Johnson & Johnson. U.S. District Court upholds Risperdal® (risperidone) patent (press release). October 16, 2006. www.jnj.com/news/jnj_news/20061016_094453.htm.
- Carbamazepine • Tegretol
- Lorazepam • Ativan
- Olanzapine • Zyprexa
- Paliperidone ER • Invega
- Risperidone • Risperdal
Dr. Rado and Dr. Dowd receive research support from Neuronetics, sanofi-aventis, Janssen Pharmaceutica, and Solvay.
Dr. Janicak receives research support from Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, Neuronetics, Solvay, and sanofi-aventis. He is a consultant to Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, Neuronetics, and Solvay, and a speaker for Abbott Laboratories, Astra-Zeneca, Bristol-Myers Squibb, Janssen Pharmaceutica, and Pfizer.
1. Marder S, Kramer M, Ford L, et al. Efficacy and safety of paliperidone extended-release tablets: results of a 6-week, randomized, placebo-controlled study. Biol Psychiatry 2007; Jun 27; Epub ahead of print.
2. Kane J, Canas F, Kramer M, et al. Treatment of schizophrenia with paliperidone extended-release tablets: a 6-week placebo-controlled trial. Schizophr Res 2007;90(1-3):147-61.
3. Davidson M, Emsley R, Kramer M, et al. Efficacy, safety and early response of paliperidone extended-release tablets (paliperidone ER): results of a 6-week, randomized, placebo-controlled study. Schizophr Res 2007;93(1-3):117-30.
4. Rossenu SAC, Rusch S, Janssens L, et al. Extended release formulation of paliperidone shows dose proportional pharmacokinetics. Presented at: Annual Meeting of the American Association of Pharmaceutical Scientists; October 29, 2006; San Antonio, TX.
5. Vermeir M, Boom S, Naessens I, et al. Absorption, metabolism, and excretion of a single oral dose of 14C-paliperidone 1 mg in healthy subjects. Eur Neuropsychopharmacol 2005;15(suppl):S648-9.
6. Conley R, Gupta SK, Sathyan G. Clinical spectrum of the osmotic-controlled release oral delivery system (OROS), an advanced oral delivery form. Curr Med Res Opin 2006;22(10):1879-92.
7. Spina E, Avenoso A, Facciola G, et al. Plasma concentrations of risperidone and 9-hydroxyrisperidone: effect of comedication with carbamazepine or valproate. Ther Drug Monit 2000;22(4):481-5.
8. Paliperidone extended release. Prescribing information. Available at: http://www.invega.com. Accessed August 8, 2007.
9. Meltzer H, Kramer M, Gassmann-Mayer C, et al. Efficacy and tolerability of oral paliperidone extended-release tablets in the treatment of acute schizophrenia: pooled data from three 6-week placebo controlled studies. Int J Neuropsychopharmacol 2006;9(suppl 1):S225.-
10. Thyssen A, Cleton A, Osselae NV, et al. Effects of renal impairment on the pharmacokinetic profile of paliperidone extended-release tablets. Clin Pharmacol Ther 2007. In press.
11. Thyssen A, Crauwels H, Cleton A, et al. Effects of hepatic impairment on the pharmacokinetics of paliperidone immediate-release. Presented at: 46th Annual Meeting of the New Clinical Drug Evaluation Unit (NCDEU); June 12-15, 2006; Boca Raton, FL.
12. Meyer J, Kramer M, Lane R, et al. Metabolic outcomes in patients with schizophrenia treated with oral paliperidone extended release tablets: pooled analysis of three 6 week placebo-controlled studies. Int J Neuropsychopharmacol 2006;9(suppl 1):S282.-
13. 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. J Clin Psychiatry 2004;65:267-72.
14. Marder SR, Davis JM, Chouinard G. The effects of risperidone on the five dimensions of schizophrenia derived by factor analysis: combined results of the North American trials. J Clin Psychiatry 1997;58:538-46.
15. Guy W. Clinical Global Impressions Scale. Early clinical drug evaluation unit (ECDEU) assessment manual for psychopharmacology. Rockville, MD: National Institute of Mental Health, Department of Health, Education, and Welfare; 1976:218-22. ADM publication 76-338.
16. Morosini PL, Magliano L, Brambilla L, et al. Development, reliability and acceptability of a new version of the DSMIV Social and Occupational Functioning Assessment Scale (SOFAS) to assess routine social functioning. Acta Psychiatr Scand 2000;101:323-9.
17. Patrick D, Adriaenssen I, Morosini P, Rothman M. Reliability, validity and sensitivity to change of the Personal and Social Performance scale in patients with acute schizophrenia. Int J Neuropsychopharmacol 2006;9(suppl 1):S287-8.
18. Kostic D, Bossie C, Turkoz I, et al. Paliperidone extended-release tablets in patients recently diagnosed with schizophrenia. Int J Neuropsychopharmacol 2006;9(suppl 1):S161.-
19. Kostic D, Bossie C, Turkoz I, et al. Paliperidone extended-release tablets in patients recently diagnosed with schizophrenia. Presented at: Congress of the Collegium Internationale Neruo-Psychopharmacologicum (CINP); July 9-13, 2006; Chicago, IL.
20. Tzimos A, Kramer M, Ford L, et al. A 6-week placebo-controlled study of the safety and tolerability of flexible doses of oral paliperidone extended release tablets in the treatment of schizophrenia in elderly patients. Int J Neuropsychopharmacol 2006;9(suppl 1):S155.-
21. Canuso C, Youssef E, Dirks B, et al. Paliperidone extended-release in severely-ill patients with schizophrenia. Presented at: 58th Annual Institute on Psychiatric Services; October 5-8, 2006; New York, NY.
22. Dirks B, Eerdekens M, Turkoz I, et al. Efficacy of paliperidone extended-release tablets in patients with schizophrenia and predominant negative symptoms. Int J Neuropsychopharmacol 2006;9(suppl 1):S162.-
23. Luthringer R, Staner L, Noel N, et al. Sleep assessments in patients with schizophrenia following treatment with paliperidone extended-release tablets. Eur Neuropsychopharmacol 2006;16(suppl 4):S224.-
24. Kramer M, Simpson G, Maciulis V, et al. Paliperidone extended-release tablets for prevention of symptom recurrence in patients with schizophrenia: a randomized double-blind, placebo-controlled study [published correction appears in J Clin Psychopharmacol. 2007;27(3):258]. J Clin Psychopharmacol 2007;27(1):6-14.
1. Marder S, Kramer M, Ford L, et al. Efficacy and safety of paliperidone extended-release tablets: results of a 6-week, randomized, placebo-controlled study. Biol Psychiatry 2007; Jun 27; Epub ahead of print.
2. Kane J, Canas F, Kramer M, et al. Treatment of schizophrenia with paliperidone extended-release tablets: a 6-week placebo-controlled trial. Schizophr Res 2007;90(1-3):147-61.
3. Davidson M, Emsley R, Kramer M, et al. Efficacy, safety and early response of paliperidone extended-release tablets (paliperidone ER): results of a 6-week, randomized, placebo-controlled study. Schizophr Res 2007;93(1-3):117-30.
4. Rossenu SAC, Rusch S, Janssens L, et al. Extended release formulation of paliperidone shows dose proportional pharmacokinetics. Presented at: Annual Meeting of the American Association of Pharmaceutical Scientists; October 29, 2006; San Antonio, TX.
5. Vermeir M, Boom S, Naessens I, et al. Absorption, metabolism, and excretion of a single oral dose of 14C-paliperidone 1 mg in healthy subjects. Eur Neuropsychopharmacol 2005;15(suppl):S648-9.
6. Conley R, Gupta SK, Sathyan G. Clinical spectrum of the osmotic-controlled release oral delivery system (OROS), an advanced oral delivery form. Curr Med Res Opin 2006;22(10):1879-92.
7. Spina E, Avenoso A, Facciola G, et al. Plasma concentrations of risperidone and 9-hydroxyrisperidone: effect of comedication with carbamazepine or valproate. Ther Drug Monit 2000;22(4):481-5.
8. Paliperidone extended release. Prescribing information. Available at: http://www.invega.com. Accessed August 8, 2007.
9. Meltzer H, Kramer M, Gassmann-Mayer C, et al. Efficacy and tolerability of oral paliperidone extended-release tablets in the treatment of acute schizophrenia: pooled data from three 6-week placebo controlled studies. Int J Neuropsychopharmacol 2006;9(suppl 1):S225.-
10. Thyssen A, Cleton A, Osselae NV, et al. Effects of renal impairment on the pharmacokinetic profile of paliperidone extended-release tablets. Clin Pharmacol Ther 2007. In press.
11. Thyssen A, Crauwels H, Cleton A, et al. Effects of hepatic impairment on the pharmacokinetics of paliperidone immediate-release. Presented at: 46th Annual Meeting of the New Clinical Drug Evaluation Unit (NCDEU); June 12-15, 2006; Boca Raton, FL.
12. Meyer J, Kramer M, Lane R, et al. Metabolic outcomes in patients with schizophrenia treated with oral paliperidone extended release tablets: pooled analysis of three 6 week placebo-controlled studies. Int J Neuropsychopharmacol 2006;9(suppl 1):S282.-
13. 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. J Clin Psychiatry 2004;65:267-72.
14. Marder SR, Davis JM, Chouinard G. The effects of risperidone on the five dimensions of schizophrenia derived by factor analysis: combined results of the North American trials. J Clin Psychiatry 1997;58:538-46.
15. Guy W. Clinical Global Impressions Scale. Early clinical drug evaluation unit (ECDEU) assessment manual for psychopharmacology. Rockville, MD: National Institute of Mental Health, Department of Health, Education, and Welfare; 1976:218-22. ADM publication 76-338.
16. Morosini PL, Magliano L, Brambilla L, et al. Development, reliability and acceptability of a new version of the DSMIV Social and Occupational Functioning Assessment Scale (SOFAS) to assess routine social functioning. Acta Psychiatr Scand 2000;101:323-9.
17. Patrick D, Adriaenssen I, Morosini P, Rothman M. Reliability, validity and sensitivity to change of the Personal and Social Performance scale in patients with acute schizophrenia. Int J Neuropsychopharmacol 2006;9(suppl 1):S287-8.
18. Kostic D, Bossie C, Turkoz I, et al. Paliperidone extended-release tablets in patients recently diagnosed with schizophrenia. Int J Neuropsychopharmacol 2006;9(suppl 1):S161.-
19. Kostic D, Bossie C, Turkoz I, et al. Paliperidone extended-release tablets in patients recently diagnosed with schizophrenia. Presented at: Congress of the Collegium Internationale Neruo-Psychopharmacologicum (CINP); July 9-13, 2006; Chicago, IL.
20. Tzimos A, Kramer M, Ford L, et al. A 6-week placebo-controlled study of the safety and tolerability of flexible doses of oral paliperidone extended release tablets in the treatment of schizophrenia in elderly patients. Int J Neuropsychopharmacol 2006;9(suppl 1):S155.-
21. Canuso C, Youssef E, Dirks B, et al. Paliperidone extended-release in severely-ill patients with schizophrenia. Presented at: 58th Annual Institute on Psychiatric Services; October 5-8, 2006; New York, NY.
22. Dirks B, Eerdekens M, Turkoz I, et al. Efficacy of paliperidone extended-release tablets in patients with schizophrenia and predominant negative symptoms. Int J Neuropsychopharmacol 2006;9(suppl 1):S162.-
23. Luthringer R, Staner L, Noel N, et al. Sleep assessments in patients with schizophrenia following treatment with paliperidone extended-release tablets. Eur Neuropsychopharmacol 2006;16(suppl 4):S224.-
24. Kramer M, Simpson G, Maciulis V, et al. Paliperidone extended-release tablets for prevention of symptom recurrence in patients with schizophrenia: a randomized double-blind, placebo-controlled study [published correction appears in J Clin Psychopharmacol. 2007;27(3):258]. J Clin Psychopharmacol 2007;27(1):6-14.
