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')]
Risperidone’s 2 new pediatric indications
Risperidone is the first second-generation antipsychotic (SGA) to receive FDA approval for treating children and adolescents with bipolar mania or schizophrenia. Specifically, the SGA is indicated for treating schizophrenia in patients age 13 to 17 and as monotherapy in short-term treatment of manic or mixed episodes of bipolar I disorder in patients age 10 to 17 (Table 1).
Risperidone also is approved for:
- schizophrenia in adults
- acute mania or mixed episodes associated with bipolar I disorder in adults, alone or in combination with lithium or valproate
- irritability associated with autistic disorder in patients age 5 to 16.
Table 1
Risperidone: Fast facts
| Brand name: Risperdal |
| Class: Second-generation antipsychotic |
| New indications: Schizophrenia in adolescents age 13 to 17 and monotherapy in short-term treatment of manic or mixed episodes of bipolar I disorder in children and adolescents age 10 to 17. (Risperidone had been approved for schizophrenia and short-term treatment of acute manic or mixed episodes associated with bipolar I disorder in adults and treatment of irritability associated with autistic disorder in children and adolescents.) |
| Approval date: August 22, 2007 for pediatric schizophrenia and mania indications |
| Manufacturer: Janssen, L.P. |
| Dosing forms: 0.25-, 0.5-, 1-, 2-, 3-, and 4-mg tablets; 0.5-, 1-, 2-, 3-, and 4-mg orally disintegrating tablets; 1 mg/mL oral solution |
| Recommended target dosage: 3 mg/d (pediatric schizophrenia) or 2.5 mg/d (pediatric bipolar mania). See Table 2 for initial dosages and titration |
Clinical implications
Risperidone is widely used off-label to treat irritability in children with pervasive developmental disorders,1,2 aggressive behaviors associated with conduct disorder,3 psychotic disorders,4 and bipolar disorder.5 It also has been used off-label to treat pediatric schizophrenia and bipolar disorder for many years.
These 2 new indications give clinicians additional support for using SGAs in children and adolescents with these serious psychiatric disorders.
How it works
Risperidone’s therapeutic activity in schizophrenia seems to be mediated through a combination of dopamine type 2 (D2) and serotonin type 2 (5HT2) receptor antagonism. Antagonism at receptors other than D2 and 5HT2 may explain some of risperidone’s other therapeutic effects.
Pharmacokinetics
In children, the half-lives of risperidone and its major active metabolite 9-hydroxyrisperidone are 3±2.3 hours and 22±46 hours, respectively.6 The pharmacologic activity of 9-hydroxyrisperidone is similar to that of risperidone.
Risperidone is extensively metabolized in the liver by the cytochrome P-450 (CYP) 2D6 enzyme system. The main metabolic pathway is through hydroxylation of risperidone to 9-hydroxyrisperidone by CYP 2D6. Food does not affect the rate or extent of the drug’s absorption.6
Efficacy studies
In schizophrenia. Approval of the indication for pediatric schizophrenia was based on data from 2 short-term (6 and 8 weeks) randomized, double-blind, controlled trials involving a total of 416 patients age 13 to 17 who met DSM-IV-TR criteria for schizophrenia and were experiencing an acute episode at enrollment.7 In one study, patients received risperidone, 1 to 3 mg/d, 4 to 6 mg/d, or placebo. In the other study, dosages were 0.15 to 0.6 mg/d or 1.5 to 6 mg/d. Except for patients in the 0.15 to 0.6 mg group (who initially received 0.05 mg/d), most patients started risperidone at 0.5 mg/d. In both trials, starting dosages were titrated to the target range in approximately 7 days.
Outcomes were measured as changes in total Positive and Negative Syndrome Scale (PANSS) and Personal and Social Performance (PSP) scale scores. The multi-item PANSS inventory measures positive and negative schizophrenia symptoms, disorganized thoughts, uncontrolled hostility/excitement, and anxiety/depression. The PSP gauges personal and social functioning in socially useful activities (work and study), personal and social relationships, self-care, and disturbing/ aggressive behaviors.
Risperidone, 1 to 6 mg/d, improved schizophrenia symptoms significantly more than placebo, as measured by PANSS scores. Doses >3 mg/d did not show greater efficacy than lower doses, as evaluated by PANSS and PSP scores.
Adverse reactions experienced by >5% of patients treated with risperidone included somnolence, parkinsonism, tremor, dystonia, dizziness, akathisia, increased salivation, and anxiety.7
In bipolar I disorder. Risperidone’s efficacy for short-term treatment of mania in children and adolescents was demonstrated in a 3-week, randomized, double-blind, placebo-controlled, multi-center study of 169 patients age 10 to 17 who were experiencing a manic or mixed episode of bipolar I disorder.7 Patients were randomly assigned to risperidone, 0.5 to 2.5 mg/d or 3 to 6 mg/d, or placebo. All patients were started at 0.5 mg/d and this dose was titrated to the target dosage range in 7 days.
Risperidone, 0.5 to 6 mg/d, significantly decreased the total Young Mania Rating Scale score—a measure of the severity of elevated mood, increased motor activity energy, sexual interest, sleep, irritability, speech (rate/amount), language (thought disorder, content, disruptive), aggressive behavior, appearance, and insight. No evidence of increased efficacy was observed at doses >2.5 mg/d. In this trial, symptoms reported by >5% of patients included fatigue, dizziness, dystonia, parkinsonism, akathisia, abdominal pain, dyspepsia, nausea, vomiting, and diarrhea.7
Pediatric dosing. Based on these studies, the recommended starting dose for children and adolescents is 0.5 mg/d, with titration in 0.5-to 1-mg increments to targets of:
Table 2
Recommended dosing of risperidone
for pediatric schizophrenia and bipolar mania
| Indication | Initial dose | Titration | Target dose | Effective dose range |
|---|---|---|---|---|
| Schizophrenia, adolescents age 13 to 17 | 0.5 mg/d | 0.5 to 1 mg/d | 3 mg/d | 1 to 6 mg/d |
| Bipolar mania, children and adolescents age 10 to 17 | 0.5 mg/d | 0.5 to 1 mg/d | 2.5 mg/d | 0.5 to 6 mg/d |
| Source:Reference 7 | ||||
Tolerability studies
In long-term studies, the most commonly reported adverse events associated with risperidone in children and adolescents have been rhinitis, abdominal pain, increased saliva, body pain, gynecomastia, and weight increase.8 Specific adverse effects that pose long-term concerns are:
- tardive dyskinesia (TD)
- weight gain
- increased prolactin levels
Tardive dyskinesia. In clinical trials that included 1,885 children and adolescents with autistic disorder or other psychiatric disorders treated with risperidone, 2 patients (0.1%) were reported to have TD, which resolved when risperidone was discontinued.7 To monitor for TD, administer the Abnormal Involuntary Movement Scale at baseline and every 6 months while using risperidone in pediatric patients.
Weight gain. In long-term, open-label trials, patients with autistic or other psychiatric disorders gained an average 7.5
kg after 12 months of risperidone treatment. Most of the weight gain occurred in the first 6 months.9 Expected normal weight gain in children is 3 to 3.5 kg/year adjusted for age, based on Centers for Disease Control and Prevention normative data.
Follow the American Diabetes Association guidelines10 for monitoring metabolic parameters during antipsychotic
treatment, and intervene if clinically significant weight gain occurs.
In a 16-week, placebo-controlled study,11 metformin reversed weight gain associated with SGAs in children and adolescents. Metformin’s potential side effects include hypoglycemia, diarrhea, nausea/vomiting, and (rarely) lactic acidosis, but no adverse events were attributed to metformin.
Increased prolactin. As in adults, risperidone elevates serum prolactin in children and adolescents. All pediatric risperidone trials—of autism,2 disruptive behavior disorders in children with subaverage intelligence,9 schizophrenia,7 and bipolar mania—have shown increased serum prolactin. Risperidone’s long-term effects on growth and sexual maturation have not been fully evaluated, but hyperprolactinemia may inhibit reproductive function.
Findling et al12 analyzed data from 5 clinical trials (total 700 patients) in which children and adolescents age 5 to 15 years with subaverage IQs and conduct or other disruptive behavior disorders received risperidone for up to 55 weeks. Mean prolactin levels rose from 7.8 ng/mL
at baseline to 29.4 ng/mL at weeks 4 to 7, then progressively decreased to 16.1 ng/mL at weeks 40 to 48 (n=358) and 13.0 ng/mL at weeks 52 to 55 (n=42). Girls returned to a mean value within the normal range (≤30 ng/mL) by weeks 8 to 12, and boys were close to normal values (≤18 ng/mL) by weeks 16 to 24.
The researchers concluded that serum prolactin levels in children tend to rise and peak within the first 1 to 2 months of risperidone treatment and then steadily decline to values within or very close to normal range by 3 to 5 months.
The biological significance of chronic, mild prolactin elevations is unknown.13 Children entering puberty appear to be at highest risk for elevated prolactin and clinical symptoms while treated with risperidone.14 Therefore, ask all adolescents treated with risperidone about increases in breast size and galactorrhea. Switch those who develop these symptoms to an SGA that does not increase serum prolactin.
Contraindications. Risperidone is contraindicated in patients with a known hypersensitivity to the drug.
- Risperdal prescribing information. www.risperdal.com/risperdal/shared/pi/risperdal.pdf.
Drug brand names
- Lithium • Eskalith, Lithobid
- Risperidone • Risperdal
- Metformin • Glucophage, Fortamet
- Valproate • Depakote
Disclosures
Dr. Kowatch receives research support from Bristol-Meyers Squibb, Stanley Research Foundation, National Institute of Mental Health, and National Institute of Child Health and Human Development. He is a consultant for Creative Educational Concepts, Child and Adolescent Bipolar Foundation, Abbott Laboratories, and sanofi-aventis, and a speaker for Abbott Laboratories and AstraZeneca.
1. Aman MG, De Smedt G, Derivan A, et al. Double-blind, placebo-controlled study of risperidone for the treatment of disruptive behaviors in children with subaverage intelligence. Am J Psychiatry 2002;159:1337-46.
2. McCracken JT, McGough J, Shah B, et al. Risperidone in children with autism and serious behavioral problems. N Engl J Med 2002;347(5):314-21.
3. Findling RL, McNamara NK, Branicky LA, et al. A double-blind pilot study of risperidone in the treatment of conduct disorder. J Am Acad Child Adolesc Psychiatry 2000;39(4):509-16.
4. Sikich L, Hamer R, Malekpour AH, et al. Double-blind trial comparing risperidone, olanzapine, and haloperidol in the treatment of psychotic children and adolescents. Paper presented at: Society of Biological Psychiatry Annual Meeting; May 16-18, 2002; Philadelphia, PA.
5. Frazier JA, Meyer MC, Biederman J, et al. Risperidone treatment for juvenile bipolar disorder: a retrospective chart review. J Am Acad Child Adolesc Psychiatry 1999;38(8):960-5.
6. Aman MG, Vinks AA, Remmerie B, et al. Plasma pharmacokinetic characteristics of risperidone and their relationship to saliva concentrations in children with psychiatric or neurodevelopmental disorders. Clin Ther 2007;29(7):1476-86.
7. Risperdal [package insert]. Titusville, NJ: Janssen, L.P; 2007.
8. Reyes MR, Olah R, Csaba K, et al. Long-term safety and efficacy of risperidone in children with disruptive behaviour disorders. Results of a 2-year extension study. Eur Child Adolesc Psychiatry 2006;15(2):97-104.
9. Croonenberghs J, Fegert JM, Findling RL, et al. Risperidone in children with disruptive behavior disorders and subaverage intelligence: a 1-year, open-label study of 504 patients. J Am Acad Child Adolesc Psychiatry 2005;44(1):64-72.
10. 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(2):267-72.
11. Klein DJ, Cottingham EM, Sorter M, et al. A randomized, double-blind, placebo-controlled trial of metformin treatment of weight gain associated with initiation of atypical antipsychotic therapy in children and adolescents. Am J Psychiatry 2006;163(12):2072-9.
12. Findling RL, Kusumakar V, Daneman D, et al. Prolactin levels during long-term risperidone treatment in children and adolescents. J Clin Psychiatry 2003;64(11):1362-9.
13. Staller J. The effect of long-term antipsychotic treatment on prolactin. J Child Adolesc Psychopharmacol 2006;16(3):317-26.
14. Holzer L, Eap CB. Risperidone-induced symptomatic hyperprolactinaemia in adolescents. J Clin Psychopharmacol 2006;26(2):167-71.
Dr. Kowatch is professor of psychiatry and pediatrics at Cincinnati Children’s Hospital Medical Center and a Section Editor for Current Psychiatry.
Risperidone is the first second-generation antipsychotic (SGA) to receive FDA approval for treating children and adolescents with bipolar mania or schizophrenia. Specifically, the SGA is indicated for treating schizophrenia in patients age 13 to 17 and as monotherapy in short-term treatment of manic or mixed episodes of bipolar I disorder in patients age 10 to 17 (Table 1).
Risperidone also is approved for:
- schizophrenia in adults
- acute mania or mixed episodes associated with bipolar I disorder in adults, alone or in combination with lithium or valproate
- irritability associated with autistic disorder in patients age 5 to 16.
Table 1
Risperidone: Fast facts
| Brand name: Risperdal |
| Class: Second-generation antipsychotic |
| New indications: Schizophrenia in adolescents age 13 to 17 and monotherapy in short-term treatment of manic or mixed episodes of bipolar I disorder in children and adolescents age 10 to 17. (Risperidone had been approved for schizophrenia and short-term treatment of acute manic or mixed episodes associated with bipolar I disorder in adults and treatment of irritability associated with autistic disorder in children and adolescents.) |
| Approval date: August 22, 2007 for pediatric schizophrenia and mania indications |
| Manufacturer: Janssen, L.P. |
| Dosing forms: 0.25-, 0.5-, 1-, 2-, 3-, and 4-mg tablets; 0.5-, 1-, 2-, 3-, and 4-mg orally disintegrating tablets; 1 mg/mL oral solution |
| Recommended target dosage: 3 mg/d (pediatric schizophrenia) or 2.5 mg/d (pediatric bipolar mania). See Table 2 for initial dosages and titration |
Clinical implications
Risperidone is widely used off-label to treat irritability in children with pervasive developmental disorders,1,2 aggressive behaviors associated with conduct disorder,3 psychotic disorders,4 and bipolar disorder.5 It also has been used off-label to treat pediatric schizophrenia and bipolar disorder for many years.
These 2 new indications give clinicians additional support for using SGAs in children and adolescents with these serious psychiatric disorders.
How it works
Risperidone’s therapeutic activity in schizophrenia seems to be mediated through a combination of dopamine type 2 (D2) and serotonin type 2 (5HT2) receptor antagonism. Antagonism at receptors other than D2 and 5HT2 may explain some of risperidone’s other therapeutic effects.
Pharmacokinetics
In children, the half-lives of risperidone and its major active metabolite 9-hydroxyrisperidone are 3±2.3 hours and 22±46 hours, respectively.6 The pharmacologic activity of 9-hydroxyrisperidone is similar to that of risperidone.
Risperidone is extensively metabolized in the liver by the cytochrome P-450 (CYP) 2D6 enzyme system. The main metabolic pathway is through hydroxylation of risperidone to 9-hydroxyrisperidone by CYP 2D6. Food does not affect the rate or extent of the drug’s absorption.6
Efficacy studies
In schizophrenia. Approval of the indication for pediatric schizophrenia was based on data from 2 short-term (6 and 8 weeks) randomized, double-blind, controlled trials involving a total of 416 patients age 13 to 17 who met DSM-IV-TR criteria for schizophrenia and were experiencing an acute episode at enrollment.7 In one study, patients received risperidone, 1 to 3 mg/d, 4 to 6 mg/d, or placebo. In the other study, dosages were 0.15 to 0.6 mg/d or 1.5 to 6 mg/d. Except for patients in the 0.15 to 0.6 mg group (who initially received 0.05 mg/d), most patients started risperidone at 0.5 mg/d. In both trials, starting dosages were titrated to the target range in approximately 7 days.
Outcomes were measured as changes in total Positive and Negative Syndrome Scale (PANSS) and Personal and Social Performance (PSP) scale scores. The multi-item PANSS inventory measures positive and negative schizophrenia symptoms, disorganized thoughts, uncontrolled hostility/excitement, and anxiety/depression. The PSP gauges personal and social functioning in socially useful activities (work and study), personal and social relationships, self-care, and disturbing/ aggressive behaviors.
Risperidone, 1 to 6 mg/d, improved schizophrenia symptoms significantly more than placebo, as measured by PANSS scores. Doses >3 mg/d did not show greater efficacy than lower doses, as evaluated by PANSS and PSP scores.
Adverse reactions experienced by >5% of patients treated with risperidone included somnolence, parkinsonism, tremor, dystonia, dizziness, akathisia, increased salivation, and anxiety.7
In bipolar I disorder. Risperidone’s efficacy for short-term treatment of mania in children and adolescents was demonstrated in a 3-week, randomized, double-blind, placebo-controlled, multi-center study of 169 patients age 10 to 17 who were experiencing a manic or mixed episode of bipolar I disorder.7 Patients were randomly assigned to risperidone, 0.5 to 2.5 mg/d or 3 to 6 mg/d, or placebo. All patients were started at 0.5 mg/d and this dose was titrated to the target dosage range in 7 days.
Risperidone, 0.5 to 6 mg/d, significantly decreased the total Young Mania Rating Scale score—a measure of the severity of elevated mood, increased motor activity energy, sexual interest, sleep, irritability, speech (rate/amount), language (thought disorder, content, disruptive), aggressive behavior, appearance, and insight. No evidence of increased efficacy was observed at doses >2.5 mg/d. In this trial, symptoms reported by >5% of patients included fatigue, dizziness, dystonia, parkinsonism, akathisia, abdominal pain, dyspepsia, nausea, vomiting, and diarrhea.7
Pediatric dosing. Based on these studies, the recommended starting dose for children and adolescents is 0.5 mg/d, with titration in 0.5-to 1-mg increments to targets of:
Table 2
Recommended dosing of risperidone
for pediatric schizophrenia and bipolar mania
| Indication | Initial dose | Titration | Target dose | Effective dose range |
|---|---|---|---|---|
| Schizophrenia, adolescents age 13 to 17 | 0.5 mg/d | 0.5 to 1 mg/d | 3 mg/d | 1 to 6 mg/d |
| Bipolar mania, children and adolescents age 10 to 17 | 0.5 mg/d | 0.5 to 1 mg/d | 2.5 mg/d | 0.5 to 6 mg/d |
| Source:Reference 7 | ||||
Tolerability studies
In long-term studies, the most commonly reported adverse events associated with risperidone in children and adolescents have been rhinitis, abdominal pain, increased saliva, body pain, gynecomastia, and weight increase.8 Specific adverse effects that pose long-term concerns are:
- tardive dyskinesia (TD)
- weight gain
- increased prolactin levels
Tardive dyskinesia. In clinical trials that included 1,885 children and adolescents with autistic disorder or other psychiatric disorders treated with risperidone, 2 patients (0.1%) were reported to have TD, which resolved when risperidone was discontinued.7 To monitor for TD, administer the Abnormal Involuntary Movement Scale at baseline and every 6 months while using risperidone in pediatric patients.
Weight gain. In long-term, open-label trials, patients with autistic or other psychiatric disorders gained an average 7.5
kg after 12 months of risperidone treatment. Most of the weight gain occurred in the first 6 months.9 Expected normal weight gain in children is 3 to 3.5 kg/year adjusted for age, based on Centers for Disease Control and Prevention normative data.
Follow the American Diabetes Association guidelines10 for monitoring metabolic parameters during antipsychotic
treatment, and intervene if clinically significant weight gain occurs.
In a 16-week, placebo-controlled study,11 metformin reversed weight gain associated with SGAs in children and adolescents. Metformin’s potential side effects include hypoglycemia, diarrhea, nausea/vomiting, and (rarely) lactic acidosis, but no adverse events were attributed to metformin.
Increased prolactin. As in adults, risperidone elevates serum prolactin in children and adolescents. All pediatric risperidone trials—of autism,2 disruptive behavior disorders in children with subaverage intelligence,9 schizophrenia,7 and bipolar mania—have shown increased serum prolactin. Risperidone’s long-term effects on growth and sexual maturation have not been fully evaluated, but hyperprolactinemia may inhibit reproductive function.
Findling et al12 analyzed data from 5 clinical trials (total 700 patients) in which children and adolescents age 5 to 15 years with subaverage IQs and conduct or other disruptive behavior disorders received risperidone for up to 55 weeks. Mean prolactin levels rose from 7.8 ng/mL
at baseline to 29.4 ng/mL at weeks 4 to 7, then progressively decreased to 16.1 ng/mL at weeks 40 to 48 (n=358) and 13.0 ng/mL at weeks 52 to 55 (n=42). Girls returned to a mean value within the normal range (≤30 ng/mL) by weeks 8 to 12, and boys were close to normal values (≤18 ng/mL) by weeks 16 to 24.
The researchers concluded that serum prolactin levels in children tend to rise and peak within the first 1 to 2 months of risperidone treatment and then steadily decline to values within or very close to normal range by 3 to 5 months.
The biological significance of chronic, mild prolactin elevations is unknown.13 Children entering puberty appear to be at highest risk for elevated prolactin and clinical symptoms while treated with risperidone.14 Therefore, ask all adolescents treated with risperidone about increases in breast size and galactorrhea. Switch those who develop these symptoms to an SGA that does not increase serum prolactin.
Contraindications. Risperidone is contraindicated in patients with a known hypersensitivity to the drug.
- Risperdal prescribing information. www.risperdal.com/risperdal/shared/pi/risperdal.pdf.
Drug brand names
- Lithium • Eskalith, Lithobid
- Risperidone • Risperdal
- Metformin • Glucophage, Fortamet
- Valproate • Depakote
Disclosures
Dr. Kowatch receives research support from Bristol-Meyers Squibb, Stanley Research Foundation, National Institute of Mental Health, and National Institute of Child Health and Human Development. He is a consultant for Creative Educational Concepts, Child and Adolescent Bipolar Foundation, Abbott Laboratories, and sanofi-aventis, and a speaker for Abbott Laboratories and AstraZeneca.
Risperidone is the first second-generation antipsychotic (SGA) to receive FDA approval for treating children and adolescents with bipolar mania or schizophrenia. Specifically, the SGA is indicated for treating schizophrenia in patients age 13 to 17 and as monotherapy in short-term treatment of manic or mixed episodes of bipolar I disorder in patients age 10 to 17 (Table 1).
Risperidone also is approved for:
- schizophrenia in adults
- acute mania or mixed episodes associated with bipolar I disorder in adults, alone or in combination with lithium or valproate
- irritability associated with autistic disorder in patients age 5 to 16.
Table 1
Risperidone: Fast facts
| Brand name: Risperdal |
| Class: Second-generation antipsychotic |
| New indications: Schizophrenia in adolescents age 13 to 17 and monotherapy in short-term treatment of manic or mixed episodes of bipolar I disorder in children and adolescents age 10 to 17. (Risperidone had been approved for schizophrenia and short-term treatment of acute manic or mixed episodes associated with bipolar I disorder in adults and treatment of irritability associated with autistic disorder in children and adolescents.) |
| Approval date: August 22, 2007 for pediatric schizophrenia and mania indications |
| Manufacturer: Janssen, L.P. |
| Dosing forms: 0.25-, 0.5-, 1-, 2-, 3-, and 4-mg tablets; 0.5-, 1-, 2-, 3-, and 4-mg orally disintegrating tablets; 1 mg/mL oral solution |
| Recommended target dosage: 3 mg/d (pediatric schizophrenia) or 2.5 mg/d (pediatric bipolar mania). See Table 2 for initial dosages and titration |
Clinical implications
Risperidone is widely used off-label to treat irritability in children with pervasive developmental disorders,1,2 aggressive behaviors associated with conduct disorder,3 psychotic disorders,4 and bipolar disorder.5 It also has been used off-label to treat pediatric schizophrenia and bipolar disorder for many years.
These 2 new indications give clinicians additional support for using SGAs in children and adolescents with these serious psychiatric disorders.
How it works
Risperidone’s therapeutic activity in schizophrenia seems to be mediated through a combination of dopamine type 2 (D2) and serotonin type 2 (5HT2) receptor antagonism. Antagonism at receptors other than D2 and 5HT2 may explain some of risperidone’s other therapeutic effects.
Pharmacokinetics
In children, the half-lives of risperidone and its major active metabolite 9-hydroxyrisperidone are 3±2.3 hours and 22±46 hours, respectively.6 The pharmacologic activity of 9-hydroxyrisperidone is similar to that of risperidone.
Risperidone is extensively metabolized in the liver by the cytochrome P-450 (CYP) 2D6 enzyme system. The main metabolic pathway is through hydroxylation of risperidone to 9-hydroxyrisperidone by CYP 2D6. Food does not affect the rate or extent of the drug’s absorption.6
Efficacy studies
In schizophrenia. Approval of the indication for pediatric schizophrenia was based on data from 2 short-term (6 and 8 weeks) randomized, double-blind, controlled trials involving a total of 416 patients age 13 to 17 who met DSM-IV-TR criteria for schizophrenia and were experiencing an acute episode at enrollment.7 In one study, patients received risperidone, 1 to 3 mg/d, 4 to 6 mg/d, or placebo. In the other study, dosages were 0.15 to 0.6 mg/d or 1.5 to 6 mg/d. Except for patients in the 0.15 to 0.6 mg group (who initially received 0.05 mg/d), most patients started risperidone at 0.5 mg/d. In both trials, starting dosages were titrated to the target range in approximately 7 days.
Outcomes were measured as changes in total Positive and Negative Syndrome Scale (PANSS) and Personal and Social Performance (PSP) scale scores. The multi-item PANSS inventory measures positive and negative schizophrenia symptoms, disorganized thoughts, uncontrolled hostility/excitement, and anxiety/depression. The PSP gauges personal and social functioning in socially useful activities (work and study), personal and social relationships, self-care, and disturbing/ aggressive behaviors.
Risperidone, 1 to 6 mg/d, improved schizophrenia symptoms significantly more than placebo, as measured by PANSS scores. Doses >3 mg/d did not show greater efficacy than lower doses, as evaluated by PANSS and PSP scores.
Adverse reactions experienced by >5% of patients treated with risperidone included somnolence, parkinsonism, tremor, dystonia, dizziness, akathisia, increased salivation, and anxiety.7
In bipolar I disorder. Risperidone’s efficacy for short-term treatment of mania in children and adolescents was demonstrated in a 3-week, randomized, double-blind, placebo-controlled, multi-center study of 169 patients age 10 to 17 who were experiencing a manic or mixed episode of bipolar I disorder.7 Patients were randomly assigned to risperidone, 0.5 to 2.5 mg/d or 3 to 6 mg/d, or placebo. All patients were started at 0.5 mg/d and this dose was titrated to the target dosage range in 7 days.
Risperidone, 0.5 to 6 mg/d, significantly decreased the total Young Mania Rating Scale score—a measure of the severity of elevated mood, increased motor activity energy, sexual interest, sleep, irritability, speech (rate/amount), language (thought disorder, content, disruptive), aggressive behavior, appearance, and insight. No evidence of increased efficacy was observed at doses >2.5 mg/d. In this trial, symptoms reported by >5% of patients included fatigue, dizziness, dystonia, parkinsonism, akathisia, abdominal pain, dyspepsia, nausea, vomiting, and diarrhea.7
Pediatric dosing. Based on these studies, the recommended starting dose for children and adolescents is 0.5 mg/d, with titration in 0.5-to 1-mg increments to targets of:
Table 2
Recommended dosing of risperidone
for pediatric schizophrenia and bipolar mania
| Indication | Initial dose | Titration | Target dose | Effective dose range |
|---|---|---|---|---|
| Schizophrenia, adolescents age 13 to 17 | 0.5 mg/d | 0.5 to 1 mg/d | 3 mg/d | 1 to 6 mg/d |
| Bipolar mania, children and adolescents age 10 to 17 | 0.5 mg/d | 0.5 to 1 mg/d | 2.5 mg/d | 0.5 to 6 mg/d |
| Source:Reference 7 | ||||
Tolerability studies
In long-term studies, the most commonly reported adverse events associated with risperidone in children and adolescents have been rhinitis, abdominal pain, increased saliva, body pain, gynecomastia, and weight increase.8 Specific adverse effects that pose long-term concerns are:
- tardive dyskinesia (TD)
- weight gain
- increased prolactin levels
Tardive dyskinesia. In clinical trials that included 1,885 children and adolescents with autistic disorder or other psychiatric disorders treated with risperidone, 2 patients (0.1%) were reported to have TD, which resolved when risperidone was discontinued.7 To monitor for TD, administer the Abnormal Involuntary Movement Scale at baseline and every 6 months while using risperidone in pediatric patients.
Weight gain. In long-term, open-label trials, patients with autistic or other psychiatric disorders gained an average 7.5
kg after 12 months of risperidone treatment. Most of the weight gain occurred in the first 6 months.9 Expected normal weight gain in children is 3 to 3.5 kg/year adjusted for age, based on Centers for Disease Control and Prevention normative data.
Follow the American Diabetes Association guidelines10 for monitoring metabolic parameters during antipsychotic
treatment, and intervene if clinically significant weight gain occurs.
In a 16-week, placebo-controlled study,11 metformin reversed weight gain associated with SGAs in children and adolescents. Metformin’s potential side effects include hypoglycemia, diarrhea, nausea/vomiting, and (rarely) lactic acidosis, but no adverse events were attributed to metformin.
Increased prolactin. As in adults, risperidone elevates serum prolactin in children and adolescents. All pediatric risperidone trials—of autism,2 disruptive behavior disorders in children with subaverage intelligence,9 schizophrenia,7 and bipolar mania—have shown increased serum prolactin. Risperidone’s long-term effects on growth and sexual maturation have not been fully evaluated, but hyperprolactinemia may inhibit reproductive function.
Findling et al12 analyzed data from 5 clinical trials (total 700 patients) in which children and adolescents age 5 to 15 years with subaverage IQs and conduct or other disruptive behavior disorders received risperidone for up to 55 weeks. Mean prolactin levels rose from 7.8 ng/mL
at baseline to 29.4 ng/mL at weeks 4 to 7, then progressively decreased to 16.1 ng/mL at weeks 40 to 48 (n=358) and 13.0 ng/mL at weeks 52 to 55 (n=42). Girls returned to a mean value within the normal range (≤30 ng/mL) by weeks 8 to 12, and boys were close to normal values (≤18 ng/mL) by weeks 16 to 24.
The researchers concluded that serum prolactin levels in children tend to rise and peak within the first 1 to 2 months of risperidone treatment and then steadily decline to values within or very close to normal range by 3 to 5 months.
The biological significance of chronic, mild prolactin elevations is unknown.13 Children entering puberty appear to be at highest risk for elevated prolactin and clinical symptoms while treated with risperidone.14 Therefore, ask all adolescents treated with risperidone about increases in breast size and galactorrhea. Switch those who develop these symptoms to an SGA that does not increase serum prolactin.
Contraindications. Risperidone is contraindicated in patients with a known hypersensitivity to the drug.
- Risperdal prescribing information. www.risperdal.com/risperdal/shared/pi/risperdal.pdf.
Drug brand names
- Lithium • Eskalith, Lithobid
- Risperidone • Risperdal
- Metformin • Glucophage, Fortamet
- Valproate • Depakote
Disclosures
Dr. Kowatch receives research support from Bristol-Meyers Squibb, Stanley Research Foundation, National Institute of Mental Health, and National Institute of Child Health and Human Development. He is a consultant for Creative Educational Concepts, Child and Adolescent Bipolar Foundation, Abbott Laboratories, and sanofi-aventis, and a speaker for Abbott Laboratories and AstraZeneca.
1. Aman MG, De Smedt G, Derivan A, et al. Double-blind, placebo-controlled study of risperidone for the treatment of disruptive behaviors in children with subaverage intelligence. Am J Psychiatry 2002;159:1337-46.
2. McCracken JT, McGough J, Shah B, et al. Risperidone in children with autism and serious behavioral problems. N Engl J Med 2002;347(5):314-21.
3. Findling RL, McNamara NK, Branicky LA, et al. A double-blind pilot study of risperidone in the treatment of conduct disorder. J Am Acad Child Adolesc Psychiatry 2000;39(4):509-16.
4. Sikich L, Hamer R, Malekpour AH, et al. Double-blind trial comparing risperidone, olanzapine, and haloperidol in the treatment of psychotic children and adolescents. Paper presented at: Society of Biological Psychiatry Annual Meeting; May 16-18, 2002; Philadelphia, PA.
5. Frazier JA, Meyer MC, Biederman J, et al. Risperidone treatment for juvenile bipolar disorder: a retrospective chart review. J Am Acad Child Adolesc Psychiatry 1999;38(8):960-5.
6. Aman MG, Vinks AA, Remmerie B, et al. Plasma pharmacokinetic characteristics of risperidone and their relationship to saliva concentrations in children with psychiatric or neurodevelopmental disorders. Clin Ther 2007;29(7):1476-86.
7. Risperdal [package insert]. Titusville, NJ: Janssen, L.P; 2007.
8. Reyes MR, Olah R, Csaba K, et al. Long-term safety and efficacy of risperidone in children with disruptive behaviour disorders. Results of a 2-year extension study. Eur Child Adolesc Psychiatry 2006;15(2):97-104.
9. Croonenberghs J, Fegert JM, Findling RL, et al. Risperidone in children with disruptive behavior disorders and subaverage intelligence: a 1-year, open-label study of 504 patients. J Am Acad Child Adolesc Psychiatry 2005;44(1):64-72.
10. 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(2):267-72.
11. Klein DJ, Cottingham EM, Sorter M, et al. A randomized, double-blind, placebo-controlled trial of metformin treatment of weight gain associated with initiation of atypical antipsychotic therapy in children and adolescents. Am J Psychiatry 2006;163(12):2072-9.
12. Findling RL, Kusumakar V, Daneman D, et al. Prolactin levels during long-term risperidone treatment in children and adolescents. J Clin Psychiatry 2003;64(11):1362-9.
13. Staller J. The effect of long-term antipsychotic treatment on prolactin. J Child Adolesc Psychopharmacol 2006;16(3):317-26.
14. Holzer L, Eap CB. Risperidone-induced symptomatic hyperprolactinaemia in adolescents. J Clin Psychopharmacol 2006;26(2):167-71.
Dr. Kowatch is professor of psychiatry and pediatrics at Cincinnati Children’s Hospital Medical Center and a Section Editor for Current Psychiatry.
1. Aman MG, De Smedt G, Derivan A, et al. Double-blind, placebo-controlled study of risperidone for the treatment of disruptive behaviors in children with subaverage intelligence. Am J Psychiatry 2002;159:1337-46.
2. McCracken JT, McGough J, Shah B, et al. Risperidone in children with autism and serious behavioral problems. N Engl J Med 2002;347(5):314-21.
3. Findling RL, McNamara NK, Branicky LA, et al. A double-blind pilot study of risperidone in the treatment of conduct disorder. J Am Acad Child Adolesc Psychiatry 2000;39(4):509-16.
4. Sikich L, Hamer R, Malekpour AH, et al. Double-blind trial comparing risperidone, olanzapine, and haloperidol in the treatment of psychotic children and adolescents. Paper presented at: Society of Biological Psychiatry Annual Meeting; May 16-18, 2002; Philadelphia, PA.
5. Frazier JA, Meyer MC, Biederman J, et al. Risperidone treatment for juvenile bipolar disorder: a retrospective chart review. J Am Acad Child Adolesc Psychiatry 1999;38(8):960-5.
6. Aman MG, Vinks AA, Remmerie B, et al. Plasma pharmacokinetic characteristics of risperidone and their relationship to saliva concentrations in children with psychiatric or neurodevelopmental disorders. Clin Ther 2007;29(7):1476-86.
7. Risperdal [package insert]. Titusville, NJ: Janssen, L.P; 2007.
8. Reyes MR, Olah R, Csaba K, et al. Long-term safety and efficacy of risperidone in children with disruptive behaviour disorders. Results of a 2-year extension study. Eur Child Adolesc Psychiatry 2006;15(2):97-104.
9. Croonenberghs J, Fegert JM, Findling RL, et al. Risperidone in children with disruptive behavior disorders and subaverage intelligence: a 1-year, open-label study of 504 patients. J Am Acad Child Adolesc Psychiatry 2005;44(1):64-72.
10. 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(2):267-72.
11. Klein DJ, Cottingham EM, Sorter M, et al. A randomized, double-blind, placebo-controlled trial of metformin treatment of weight gain associated with initiation of atypical antipsychotic therapy in children and adolescents. Am J Psychiatry 2006;163(12):2072-9.
12. Findling RL, Kusumakar V, Daneman D, et al. Prolactin levels during long-term risperidone treatment in children and adolescents. J Clin Psychiatry 2003;64(11):1362-9.
13. Staller J. The effect of long-term antipsychotic treatment on prolactin. J Child Adolesc Psychopharmacol 2006;16(3):317-26.
14. Holzer L, Eap CB. Risperidone-induced symptomatic hyperprolactinaemia in adolescents. J Clin Psychopharmacol 2006;26(2):167-71.
Dr. Kowatch is professor of psychiatry and pediatrics at Cincinnati Children’s Hospital Medical Center and a Section Editor for Current Psychiatry.
Lost in translation: Be leery of lay language
Patients may use diagnostic labels of psychiatric disorders when describing their mental distress to clinicians. Sometimes they use these words appropriately, but often they don’t understand the meaning of psychiatric terms they have read or heard (Table). For example, a study of U.S. newspapers found that 28% of articles incorrectly used “schizophrenic” to refer to a “split” or inconsistent personality.1
Terminology confusion could lead to 2 clinical problems:
- The patient may be confident in his or her self-diagnosis, which can strain the therapeutic relationship.
- The clinician may passively accept that the patient’s use of terms is accurate, resulting in a distorted diagnosis.
Table
Psychiatric terms patients misuse to describe symptoms
| Psychiatric term | Possible implied symptoms |
|---|---|
| ‘ADHD’ or ‘ADD’ | Poor concentration or forgetfulness, excessive energy or restlessness |
| ‘Antisocial’ | Social withdrawal, unpleasant interpersonal relationships |
| ‘Bipolar’ | Mood lability, unpredictability, anxiety attacks |
| ‘Depression’ | Grief, remorse, loneliness, disappointment |
| ‘Obsessing’ | Ruminating, worrying |
| ‘OCD’ | Meticulousness, excessive worrying |
| ‘Panic attack’ | Intense anxiety, even without physical symptoms |
| ‘Paranoid’ | Worry, dread, pessimism |
| ‘Psychotic’ | Enraged, unpredictable |
| ‘PTSD’ | Emotional change following a significant, though not necessarily traumatic, event |
| ‘Schizophrenic’ | Indecisive, ‘split personality’ |
| ADHD: attention-deficit/hyperactivity disorder; OCD: obsessive-compulsive disorder; PTSD: posttraumatic stress disorder | |
Searching for meaning
When a patient uses a psychiatric term to describe symptoms, clarify what he or she means by asking; “Can you tell me more about your experience of (the term) without using that word to describe it?” Alternately, you could say, “Let’s not worry about applying a label right now, just describe what you are going through.”
Another approach is to offer phrases that are synonyms of the term’s standard use. For example, ask your patient, “By ‘schizophrenic’ do you mean ‘being in 2 minds’ or ‘having really unusual experiences?’” Using popular culture references also may be helpful. For example, “When I use the term ‘schizophrenia,’ I think of someone like Russell Crowe’s character in the movie A Beautiful Mind.” Similarly, for obsessive-compulsive disorder, reference Jack Nicholson’s character in As Good as it Gets.
Clarifying terminology also can help you gather a complete family history. A patient may say “Oh, that sounds like my mother” when you accurately describe a psychiatric symptom or disorder.
1. Duckworth K, Halpern JH, Schutt K, et al. Use of schizophrenia as a metaphor in U.S. newspapers. Psychiatr Serv 2003;54:1402-4.
Dr. Dunlop is assistant professor, department of psychiatry and behavioral sciences, Emory University, Atlanta, GA.
Patients may use diagnostic labels of psychiatric disorders when describing their mental distress to clinicians. Sometimes they use these words appropriately, but often they don’t understand the meaning of psychiatric terms they have read or heard (Table). For example, a study of U.S. newspapers found that 28% of articles incorrectly used “schizophrenic” to refer to a “split” or inconsistent personality.1
Terminology confusion could lead to 2 clinical problems:
- The patient may be confident in his or her self-diagnosis, which can strain the therapeutic relationship.
- The clinician may passively accept that the patient’s use of terms is accurate, resulting in a distorted diagnosis.
Table
Psychiatric terms patients misuse to describe symptoms
| Psychiatric term | Possible implied symptoms |
|---|---|
| ‘ADHD’ or ‘ADD’ | Poor concentration or forgetfulness, excessive energy or restlessness |
| ‘Antisocial’ | Social withdrawal, unpleasant interpersonal relationships |
| ‘Bipolar’ | Mood lability, unpredictability, anxiety attacks |
| ‘Depression’ | Grief, remorse, loneliness, disappointment |
| ‘Obsessing’ | Ruminating, worrying |
| ‘OCD’ | Meticulousness, excessive worrying |
| ‘Panic attack’ | Intense anxiety, even without physical symptoms |
| ‘Paranoid’ | Worry, dread, pessimism |
| ‘Psychotic’ | Enraged, unpredictable |
| ‘PTSD’ | Emotional change following a significant, though not necessarily traumatic, event |
| ‘Schizophrenic’ | Indecisive, ‘split personality’ |
| ADHD: attention-deficit/hyperactivity disorder; OCD: obsessive-compulsive disorder; PTSD: posttraumatic stress disorder | |
Searching for meaning
When a patient uses a psychiatric term to describe symptoms, clarify what he or she means by asking; “Can you tell me more about your experience of (the term) without using that word to describe it?” Alternately, you could say, “Let’s not worry about applying a label right now, just describe what you are going through.”
Another approach is to offer phrases that are synonyms of the term’s standard use. For example, ask your patient, “By ‘schizophrenic’ do you mean ‘being in 2 minds’ or ‘having really unusual experiences?’” Using popular culture references also may be helpful. For example, “When I use the term ‘schizophrenia,’ I think of someone like Russell Crowe’s character in the movie A Beautiful Mind.” Similarly, for obsessive-compulsive disorder, reference Jack Nicholson’s character in As Good as it Gets.
Clarifying terminology also can help you gather a complete family history. A patient may say “Oh, that sounds like my mother” when you accurately describe a psychiatric symptom or disorder.
Patients may use diagnostic labels of psychiatric disorders when describing their mental distress to clinicians. Sometimes they use these words appropriately, but often they don’t understand the meaning of psychiatric terms they have read or heard (Table). For example, a study of U.S. newspapers found that 28% of articles incorrectly used “schizophrenic” to refer to a “split” or inconsistent personality.1
Terminology confusion could lead to 2 clinical problems:
- The patient may be confident in his or her self-diagnosis, which can strain the therapeutic relationship.
- The clinician may passively accept that the patient’s use of terms is accurate, resulting in a distorted diagnosis.
Table
Psychiatric terms patients misuse to describe symptoms
| Psychiatric term | Possible implied symptoms |
|---|---|
| ‘ADHD’ or ‘ADD’ | Poor concentration or forgetfulness, excessive energy or restlessness |
| ‘Antisocial’ | Social withdrawal, unpleasant interpersonal relationships |
| ‘Bipolar’ | Mood lability, unpredictability, anxiety attacks |
| ‘Depression’ | Grief, remorse, loneliness, disappointment |
| ‘Obsessing’ | Ruminating, worrying |
| ‘OCD’ | Meticulousness, excessive worrying |
| ‘Panic attack’ | Intense anxiety, even without physical symptoms |
| ‘Paranoid’ | Worry, dread, pessimism |
| ‘Psychotic’ | Enraged, unpredictable |
| ‘PTSD’ | Emotional change following a significant, though not necessarily traumatic, event |
| ‘Schizophrenic’ | Indecisive, ‘split personality’ |
| ADHD: attention-deficit/hyperactivity disorder; OCD: obsessive-compulsive disorder; PTSD: posttraumatic stress disorder | |
Searching for meaning
When a patient uses a psychiatric term to describe symptoms, clarify what he or she means by asking; “Can you tell me more about your experience of (the term) without using that word to describe it?” Alternately, you could say, “Let’s not worry about applying a label right now, just describe what you are going through.”
Another approach is to offer phrases that are synonyms of the term’s standard use. For example, ask your patient, “By ‘schizophrenic’ do you mean ‘being in 2 minds’ or ‘having really unusual experiences?’” Using popular culture references also may be helpful. For example, “When I use the term ‘schizophrenia,’ I think of someone like Russell Crowe’s character in the movie A Beautiful Mind.” Similarly, for obsessive-compulsive disorder, reference Jack Nicholson’s character in As Good as it Gets.
Clarifying terminology also can help you gather a complete family history. A patient may say “Oh, that sounds like my mother” when you accurately describe a psychiatric symptom or disorder.
1. Duckworth K, Halpern JH, Schutt K, et al. Use of schizophrenia as a metaphor in U.S. newspapers. Psychiatr Serv 2003;54:1402-4.
Dr. Dunlop is assistant professor, department of psychiatry and behavioral sciences, Emory University, Atlanta, GA.
1. Duckworth K, Halpern JH, Schutt K, et al. Use of schizophrenia as a metaphor in U.S. newspapers. Psychiatr Serv 2003;54:1402-4.
Dr. Dunlop is assistant professor, department of psychiatry and behavioral sciences, Emory University, Atlanta, GA.
Tips for telling your patients good-bye
Transferring to another psychiatrist can distress mental health patients and disrupt treatment, whether you part ways with them because of an insurance change or relocation. A smooth transfer helps maintain patients’ clinical progress and reduces the risk of losing them to follow-up. We suggest a timeline for saying good-bye (Table) and some strategies to ease the transition.
Table
Timeline for transferring your patient’s care
| Issues to discuss/explore | |
|---|---|
| 6 months before departure | Determine which issues patient would like to address before transfer Current or past medications |
| 1 month before departure | Focus on closure Avoid addressing new issues Avoid changing medications or session time, day, or frequency Go over transfer summary |
| Final session | Give 1 to 2 prescription refills Encourage patient to follow up with new doctor End session on positive note |
Starting the conversation
Inform the patient of your approximate departure date as soon as possible. Most residents, for example, should have this conversation in January, allowing approximately 6 months to address issues your departure may bring up. Don’t be surprised if your patient does not recall this conversation, however, because he or she might unconsciously repress this information. You might have to discuss your departure several times before it becomes “real” for your patient.
Identify specific issues to address before transferring the patient’s care. For example, explore whether any medications need to be changed.
Tell your patient you would like to write the transfer summary together, and encourage him or her to think about what information to include. If another physician transferred the patient to you, inquire about that process. Did the earlier physician do or say something that was helpful?
Initiating transfer of care
Encourage your patient to talk about feelings related to the transfer by asking how he or she thinks the process will go. Don’t assume your patient is anxious or upset about the change, however. Some patients “bond” to the clinic rather than to a particular doctor.
Be alert for unconscious communication about your impending departure. For example, your patient might talk about others who have left in the past. Consider these statements as opportunities to discuss your departure against the backdrop of other losses and changes.
Patients might unconsciously act out in response to your upcoming departure. For example, a patient who has faithfully attended appointments might “accidentally” miss a visit or discontinue 1 or more medications.
Examine your feelings about the impending transfer of care. Guard against attributing your feelings about the process to your patient. If you find that these feelings lead to difficulty helping your patient find closure, consider consulting with a colleague or mentor.
1 month before the transfer
Your patient might initiate more intense work than in the past. Your impending departure might make it seem safer to share previously undiscussed information because there is little time to explore it.
Although you may be tempted to take advantage of your patient’s impulse, carefully assess this strategy. This is the time to work toward closure, rather than delving into new areas. Keep treatment structured; avoid increasing or decreasing the frequency of visits as you approach the last session.
Also avoid changing the patient’s medication regimen, if possible. If your patient is anxious about your departure, new medication side effects might exacerbate this anxiety.
If possible, personally introduce your patient to the new physician and discuss the transfer summary. Don’t say that the new doctor is “really good.” The qualities you like about this clinician might not appeal to the patient. Encourage the patient to “interview” the new physician.
Don’t discuss what you will be doing after you leave. If the patient asks, talk about your plans in general terms. Detailed or persistent questioning might have psychological meaning and could be discussed in psychotherapy.
The last session
Write 1 or 2 prescription refills. Many patients are concerned about a possible delay in starting treatment with the new physician, and adequate refills may allay fears about obtaining medication. Having refills also may act as a temporary “transitional object” until the patient feels comfortable with the new physician.
Tell your patient that many individuals don’t follow up with a new physician, but it is important to do so. Discussing this phenomenon may increase the probability that your patient will follow up because you can talk about his or her concerns about seeing a new physician or ending treatment with you.
Don’t agree to correspond with the patient after you transfer care. Further communication might interfere with the new therapeutic relationship. The patient might communicate clinical concerns to you, not to the new physician.
Don’t initiate a hug at the end of the session. If your patient initiates a hug or a handshake, you may accept it if you are comfortable with physical contact. End the session on a positive note, and express your best wishes for the patient’s continued growth and well-being.
Still having problems?
If the transition of your patient’s care is unusually difficult, do not hesitate to ask a supervisor or colleague for assistance.
Dr. Kay is instructor in psychiatry and Dr. Mago is assistant professor of psychiatry, Thomas Jefferson University, Philadelphia, PA.
Transferring to another psychiatrist can distress mental health patients and disrupt treatment, whether you part ways with them because of an insurance change or relocation. A smooth transfer helps maintain patients’ clinical progress and reduces the risk of losing them to follow-up. We suggest a timeline for saying good-bye (Table) and some strategies to ease the transition.
Table
Timeline for transferring your patient’s care
| Issues to discuss/explore | |
|---|---|
| 6 months before departure | Determine which issues patient would like to address before transfer Current or past medications |
| 1 month before departure | Focus on closure Avoid addressing new issues Avoid changing medications or session time, day, or frequency Go over transfer summary |
| Final session | Give 1 to 2 prescription refills Encourage patient to follow up with new doctor End session on positive note |
Starting the conversation
Inform the patient of your approximate departure date as soon as possible. Most residents, for example, should have this conversation in January, allowing approximately 6 months to address issues your departure may bring up. Don’t be surprised if your patient does not recall this conversation, however, because he or she might unconsciously repress this information. You might have to discuss your departure several times before it becomes “real” for your patient.
Identify specific issues to address before transferring the patient’s care. For example, explore whether any medications need to be changed.
Tell your patient you would like to write the transfer summary together, and encourage him or her to think about what information to include. If another physician transferred the patient to you, inquire about that process. Did the earlier physician do or say something that was helpful?
Initiating transfer of care
Encourage your patient to talk about feelings related to the transfer by asking how he or she thinks the process will go. Don’t assume your patient is anxious or upset about the change, however. Some patients “bond” to the clinic rather than to a particular doctor.
Be alert for unconscious communication about your impending departure. For example, your patient might talk about others who have left in the past. Consider these statements as opportunities to discuss your departure against the backdrop of other losses and changes.
Patients might unconsciously act out in response to your upcoming departure. For example, a patient who has faithfully attended appointments might “accidentally” miss a visit or discontinue 1 or more medications.
Examine your feelings about the impending transfer of care. Guard against attributing your feelings about the process to your patient. If you find that these feelings lead to difficulty helping your patient find closure, consider consulting with a colleague or mentor.
1 month before the transfer
Your patient might initiate more intense work than in the past. Your impending departure might make it seem safer to share previously undiscussed information because there is little time to explore it.
Although you may be tempted to take advantage of your patient’s impulse, carefully assess this strategy. This is the time to work toward closure, rather than delving into new areas. Keep treatment structured; avoid increasing or decreasing the frequency of visits as you approach the last session.
Also avoid changing the patient’s medication regimen, if possible. If your patient is anxious about your departure, new medication side effects might exacerbate this anxiety.
If possible, personally introduce your patient to the new physician and discuss the transfer summary. Don’t say that the new doctor is “really good.” The qualities you like about this clinician might not appeal to the patient. Encourage the patient to “interview” the new physician.
Don’t discuss what you will be doing after you leave. If the patient asks, talk about your plans in general terms. Detailed or persistent questioning might have psychological meaning and could be discussed in psychotherapy.
The last session
Write 1 or 2 prescription refills. Many patients are concerned about a possible delay in starting treatment with the new physician, and adequate refills may allay fears about obtaining medication. Having refills also may act as a temporary “transitional object” until the patient feels comfortable with the new physician.
Tell your patient that many individuals don’t follow up with a new physician, but it is important to do so. Discussing this phenomenon may increase the probability that your patient will follow up because you can talk about his or her concerns about seeing a new physician or ending treatment with you.
Don’t agree to correspond with the patient after you transfer care. Further communication might interfere with the new therapeutic relationship. The patient might communicate clinical concerns to you, not to the new physician.
Don’t initiate a hug at the end of the session. If your patient initiates a hug or a handshake, you may accept it if you are comfortable with physical contact. End the session on a positive note, and express your best wishes for the patient’s continued growth and well-being.
Still having problems?
If the transition of your patient’s care is unusually difficult, do not hesitate to ask a supervisor or colleague for assistance.
Transferring to another psychiatrist can distress mental health patients and disrupt treatment, whether you part ways with them because of an insurance change or relocation. A smooth transfer helps maintain patients’ clinical progress and reduces the risk of losing them to follow-up. We suggest a timeline for saying good-bye (Table) and some strategies to ease the transition.
Table
Timeline for transferring your patient’s care
| Issues to discuss/explore | |
|---|---|
| 6 months before departure | Determine which issues patient would like to address before transfer Current or past medications |
| 1 month before departure | Focus on closure Avoid addressing new issues Avoid changing medications or session time, day, or frequency Go over transfer summary |
| Final session | Give 1 to 2 prescription refills Encourage patient to follow up with new doctor End session on positive note |
Starting the conversation
Inform the patient of your approximate departure date as soon as possible. Most residents, for example, should have this conversation in January, allowing approximately 6 months to address issues your departure may bring up. Don’t be surprised if your patient does not recall this conversation, however, because he or she might unconsciously repress this information. You might have to discuss your departure several times before it becomes “real” for your patient.
Identify specific issues to address before transferring the patient’s care. For example, explore whether any medications need to be changed.
Tell your patient you would like to write the transfer summary together, and encourage him or her to think about what information to include. If another physician transferred the patient to you, inquire about that process. Did the earlier physician do or say something that was helpful?
Initiating transfer of care
Encourage your patient to talk about feelings related to the transfer by asking how he or she thinks the process will go. Don’t assume your patient is anxious or upset about the change, however. Some patients “bond” to the clinic rather than to a particular doctor.
Be alert for unconscious communication about your impending departure. For example, your patient might talk about others who have left in the past. Consider these statements as opportunities to discuss your departure against the backdrop of other losses and changes.
Patients might unconsciously act out in response to your upcoming departure. For example, a patient who has faithfully attended appointments might “accidentally” miss a visit or discontinue 1 or more medications.
Examine your feelings about the impending transfer of care. Guard against attributing your feelings about the process to your patient. If you find that these feelings lead to difficulty helping your patient find closure, consider consulting with a colleague or mentor.
1 month before the transfer
Your patient might initiate more intense work than in the past. Your impending departure might make it seem safer to share previously undiscussed information because there is little time to explore it.
Although you may be tempted to take advantage of your patient’s impulse, carefully assess this strategy. This is the time to work toward closure, rather than delving into new areas. Keep treatment structured; avoid increasing or decreasing the frequency of visits as you approach the last session.
Also avoid changing the patient’s medication regimen, if possible. If your patient is anxious about your departure, new medication side effects might exacerbate this anxiety.
If possible, personally introduce your patient to the new physician and discuss the transfer summary. Don’t say that the new doctor is “really good.” The qualities you like about this clinician might not appeal to the patient. Encourage the patient to “interview” the new physician.
Don’t discuss what you will be doing after you leave. If the patient asks, talk about your plans in general terms. Detailed or persistent questioning might have psychological meaning and could be discussed in psychotherapy.
The last session
Write 1 or 2 prescription refills. Many patients are concerned about a possible delay in starting treatment with the new physician, and adequate refills may allay fears about obtaining medication. Having refills also may act as a temporary “transitional object” until the patient feels comfortable with the new physician.
Tell your patient that many individuals don’t follow up with a new physician, but it is important to do so. Discussing this phenomenon may increase the probability that your patient will follow up because you can talk about his or her concerns about seeing a new physician or ending treatment with you.
Don’t agree to correspond with the patient after you transfer care. Further communication might interfere with the new therapeutic relationship. The patient might communicate clinical concerns to you, not to the new physician.
Don’t initiate a hug at the end of the session. If your patient initiates a hug or a handshake, you may accept it if you are comfortable with physical contact. End the session on a positive note, and express your best wishes for the patient’s continued growth and well-being.
Still having problems?
If the transition of your patient’s care is unusually difficult, do not hesitate to ask a supervisor or colleague for assistance.
Dr. Kay is instructor in psychiatry and Dr. Mago is assistant professor of psychiatry, Thomas Jefferson University, Philadelphia, PA.
Dr. Kay is instructor in psychiatry and Dr. Mago is assistant professor of psychiatry, Thomas Jefferson University, Philadelphia, PA.
Consider adding a stimulant to antidepressant treatment
My comments are somewhat biased because my medical practice is exclusively dedicated to treating attention-deficit/hyperactivity disorder (ADHD) and the associated comorbid disorders for the last 30 years. However, Dr. James W. Jefferson’s brilliant article (Rediscovering antidepressants: The spectrum beyond depression, Current Psychiatry, October 2007) has one major omission in my opinion. My suggestion is to add methylphenidate to antidepressant treatment. Methylphenidate is in fact a mild antidepressant and was introduced as such many years ago.
Over the years I have seen hundreds of adults who have seen many doctors and received many antidepressants for depression with no or minimal benefit. The majority of these patients—with very few exceptions—are undiagnosed adults with ADHD.
One sequela of untreated ADHD is depression. When a patient is placed on an effective dose of a stimulant such as methylphenidate and monitored to optimal levels together with an antidepressant, the results are nothing less than dramatic.
There are very few resistant depressions but, rather, many missed ADHD diagnoses. The response time is also very exciting. Sometimes results as measured by the Hamilton Depression Rating Scale and the modified Conners’ Adult ADHD Rating Scale will show within 10 to 20 days. These results often are confirmed by the patient and family members, making this treatment a clear-cut success story.
My oldest patient is 84 years old, and she has told me that for the first time in her life she is living a "whole" life instead of a "half" life. I must admit I was hesitant to treat such an old patient, but her result is very gratifying indeed.
Billy Levin, MbChB
Benoni, South Africa
My comments are somewhat biased because my medical practice is exclusively dedicated to treating attention-deficit/hyperactivity disorder (ADHD) and the associated comorbid disorders for the last 30 years. However, Dr. James W. Jefferson’s brilliant article (Rediscovering antidepressants: The spectrum beyond depression, Current Psychiatry, October 2007) has one major omission in my opinion. My suggestion is to add methylphenidate to antidepressant treatment. Methylphenidate is in fact a mild antidepressant and was introduced as such many years ago.
Over the years I have seen hundreds of adults who have seen many doctors and received many antidepressants for depression with no or minimal benefit. The majority of these patients—with very few exceptions—are undiagnosed adults with ADHD.
One sequela of untreated ADHD is depression. When a patient is placed on an effective dose of a stimulant such as methylphenidate and monitored to optimal levels together with an antidepressant, the results are nothing less than dramatic.
There are very few resistant depressions but, rather, many missed ADHD diagnoses. The response time is also very exciting. Sometimes results as measured by the Hamilton Depression Rating Scale and the modified Conners’ Adult ADHD Rating Scale will show within 10 to 20 days. These results often are confirmed by the patient and family members, making this treatment a clear-cut success story.
My oldest patient is 84 years old, and she has told me that for the first time in her life she is living a "whole" life instead of a "half" life. I must admit I was hesitant to treat such an old patient, but her result is very gratifying indeed.
Billy Levin, MbChB
Benoni, South Africa
My comments are somewhat biased because my medical practice is exclusively dedicated to treating attention-deficit/hyperactivity disorder (ADHD) and the associated comorbid disorders for the last 30 years. However, Dr. James W. Jefferson’s brilliant article (Rediscovering antidepressants: The spectrum beyond depression, Current Psychiatry, October 2007) has one major omission in my opinion. My suggestion is to add methylphenidate to antidepressant treatment. Methylphenidate is in fact a mild antidepressant and was introduced as such many years ago.
Over the years I have seen hundreds of adults who have seen many doctors and received many antidepressants for depression with no or minimal benefit. The majority of these patients—with very few exceptions—are undiagnosed adults with ADHD.
One sequela of untreated ADHD is depression. When a patient is placed on an effective dose of a stimulant such as methylphenidate and monitored to optimal levels together with an antidepressant, the results are nothing less than dramatic.
There are very few resistant depressions but, rather, many missed ADHD diagnoses. The response time is also very exciting. Sometimes results as measured by the Hamilton Depression Rating Scale and the modified Conners’ Adult ADHD Rating Scale will show within 10 to 20 days. These results often are confirmed by the patient and family members, making this treatment a clear-cut success story.
My oldest patient is 84 years old, and she has told me that for the first time in her life she is living a "whole" life instead of a "half" life. I must admit I was hesitant to treat such an old patient, but her result is very gratifying indeed.
Billy Levin, MbChB
Benoni, South Africa
Brain injury: More clues to symptom validity
I appreciate Current Psychiatry’s emphasis on sharpening one’s psychiatric diagnostic skills (“Neurocognitive impairment: Feigned, exaggerated, or real?” Current Psychiatry, August 2007). I have found a few additional questions may help validate symptoms of neurocognitive impairment during a psychiatric interview. These questions were developed as a result of my informal survey of individuals with head injuries, usually months after the injury.
Ask about a patient’s job. Significant changes in employment status—such as job loss or changes in job responsibilities or how others relate to the individual in the workplace— usually occur in the first 6 months after an injury.
Has the patient thought about or obtained a gun permit? In response to their perceived weaker status, many patients acquired weapons. I have no knowledge if these guns were misused.
In an informal survey, I also asked these injured individuals to name their favorite movie and actor. The Godfather: Part II and/or the film’s actors Robert De Niro, Al Pacino, and James Caan were chosen by 85% of patients from a certain generation. One possible reason for this choice was suggested by a 14-year veteran of the Canadian Forces, who felt that the motion picture allowed one to fantasize about striking out at others without harming oneself. Positive answers to 1 or more of these questions suggested symptom validity.
The psychologist’s documentation of an organic cause of the neurocognitive impairment compared favorably to an abnormal finding on a computerized brain mapping electroencephalogram (EEG). The recent computerization of multiple EEGs will allow physicians to identify abnormal patterns of electrical activity and suggest a diagnosis. A finding of 2 standard deviations from average may indicate neurocognitive impairment.
Leonard R. Friedman, MD
Revere, MA
I appreciate Current Psychiatry’s emphasis on sharpening one’s psychiatric diagnostic skills (“Neurocognitive impairment: Feigned, exaggerated, or real?” Current Psychiatry, August 2007). I have found a few additional questions may help validate symptoms of neurocognitive impairment during a psychiatric interview. These questions were developed as a result of my informal survey of individuals with head injuries, usually months after the injury.
Ask about a patient’s job. Significant changes in employment status—such as job loss or changes in job responsibilities or how others relate to the individual in the workplace— usually occur in the first 6 months after an injury.
Has the patient thought about or obtained a gun permit? In response to their perceived weaker status, many patients acquired weapons. I have no knowledge if these guns were misused.
In an informal survey, I also asked these injured individuals to name their favorite movie and actor. The Godfather: Part II and/or the film’s actors Robert De Niro, Al Pacino, and James Caan were chosen by 85% of patients from a certain generation. One possible reason for this choice was suggested by a 14-year veteran of the Canadian Forces, who felt that the motion picture allowed one to fantasize about striking out at others without harming oneself. Positive answers to 1 or more of these questions suggested symptom validity.
The psychologist’s documentation of an organic cause of the neurocognitive impairment compared favorably to an abnormal finding on a computerized brain mapping electroencephalogram (EEG). The recent computerization of multiple EEGs will allow physicians to identify abnormal patterns of electrical activity and suggest a diagnosis. A finding of 2 standard deviations from average may indicate neurocognitive impairment.
Leonard R. Friedman, MD
Revere, MA
I appreciate Current Psychiatry’s emphasis on sharpening one’s psychiatric diagnostic skills (“Neurocognitive impairment: Feigned, exaggerated, or real?” Current Psychiatry, August 2007). I have found a few additional questions may help validate symptoms of neurocognitive impairment during a psychiatric interview. These questions were developed as a result of my informal survey of individuals with head injuries, usually months after the injury.
Ask about a patient’s job. Significant changes in employment status—such as job loss or changes in job responsibilities or how others relate to the individual in the workplace— usually occur in the first 6 months after an injury.
Has the patient thought about or obtained a gun permit? In response to their perceived weaker status, many patients acquired weapons. I have no knowledge if these guns were misused.
In an informal survey, I also asked these injured individuals to name their favorite movie and actor. The Godfather: Part II and/or the film’s actors Robert De Niro, Al Pacino, and James Caan were chosen by 85% of patients from a certain generation. One possible reason for this choice was suggested by a 14-year veteran of the Canadian Forces, who felt that the motion picture allowed one to fantasize about striking out at others without harming oneself. Positive answers to 1 or more of these questions suggested symptom validity.
The psychologist’s documentation of an organic cause of the neurocognitive impairment compared favorably to an abnormal finding on a computerized brain mapping electroencephalogram (EEG). The recent computerization of multiple EEGs will allow physicians to identify abnormal patterns of electrical activity and suggest a diagnosis. A finding of 2 standard deviations from average may indicate neurocognitive impairment.
Leonard R. Friedman, MD
Revere, MA
Identify and document suicide risk
The stepwise risk assessment in the article “Suicide intervention: How to recognize risk, focus on patient safety” is very precise and hits the bull’seye for risk assessment (Current Psychiatry, September 2007). It is important that we identify the predictors of suicide because risk is a dynamic concept and difficult to determine in all cases. Dr. David J. Muzina provided a good discussion of the risk factors associated with suicide and “protective factors.” This article will help me perform a better suicide risk assessments when on call and during routine clinical work.
Vineet Padmanabhan
Bangor, North Wales, United Kingdom
I found “Suicide intervention: How to recognize risk, focus on patient safety” to be interesting and informative. However, there is one group at high risk for suicide that was not mentioned. That is the group that suffers from gender identity dysphoria (GID) or “Harry Benjamin syndrome” (HBS) as it is now becoming known.
Major reasons for suicide in this group include feelings of helplessness, rejection by family, lack of support, and abuse from the public in general. Also, transsexuals might become depressed because of job discrimination and inability to raise required funds to pay for gender correction surgery.
I am familiar with the subject as I had GID/HBS but was fortunate enough to be able to pay for my surgery. I belong to an online suicide prevention group where we supply information, encouragement, and the telephone numbers of group members who may be contacted for immediate emotional support.
Pamela J.S. Dunn
Wesley Chapel, FL
The stepwise risk assessment in the article “Suicide intervention: How to recognize risk, focus on patient safety” is very precise and hits the bull’seye for risk assessment (Current Psychiatry, September 2007). It is important that we identify the predictors of suicide because risk is a dynamic concept and difficult to determine in all cases. Dr. David J. Muzina provided a good discussion of the risk factors associated with suicide and “protective factors.” This article will help me perform a better suicide risk assessments when on call and during routine clinical work.
Vineet Padmanabhan
Bangor, North Wales, United Kingdom
I found “Suicide intervention: How to recognize risk, focus on patient safety” to be interesting and informative. However, there is one group at high risk for suicide that was not mentioned. That is the group that suffers from gender identity dysphoria (GID) or “Harry Benjamin syndrome” (HBS) as it is now becoming known.
Major reasons for suicide in this group include feelings of helplessness, rejection by family, lack of support, and abuse from the public in general. Also, transsexuals might become depressed because of job discrimination and inability to raise required funds to pay for gender correction surgery.
I am familiar with the subject as I had GID/HBS but was fortunate enough to be able to pay for my surgery. I belong to an online suicide prevention group where we supply information, encouragement, and the telephone numbers of group members who may be contacted for immediate emotional support.
Pamela J.S. Dunn
Wesley Chapel, FL
The stepwise risk assessment in the article “Suicide intervention: How to recognize risk, focus on patient safety” is very precise and hits the bull’seye for risk assessment (Current Psychiatry, September 2007). It is important that we identify the predictors of suicide because risk is a dynamic concept and difficult to determine in all cases. Dr. David J. Muzina provided a good discussion of the risk factors associated with suicide and “protective factors.” This article will help me perform a better suicide risk assessments when on call and during routine clinical work.
Vineet Padmanabhan
Bangor, North Wales, United Kingdom
I found “Suicide intervention: How to recognize risk, focus on patient safety” to be interesting and informative. However, there is one group at high risk for suicide that was not mentioned. That is the group that suffers from gender identity dysphoria (GID) or “Harry Benjamin syndrome” (HBS) as it is now becoming known.
Major reasons for suicide in this group include feelings of helplessness, rejection by family, lack of support, and abuse from the public in general. Also, transsexuals might become depressed because of job discrimination and inability to raise required funds to pay for gender correction surgery.
I am familiar with the subject as I had GID/HBS but was fortunate enough to be able to pay for my surgery. I belong to an online suicide prevention group where we supply information, encouragement, and the telephone numbers of group members who may be contacted for immediate emotional support.
Pamela J.S. Dunn
Wesley Chapel, FL
ICD shocks and anxiety: Chicken or egg?
In “Managing anxiety in patients with implanted cardiac defibrillators” (Current Psychiatry, September 2007), Drs. Douglas P. Gibson and Kristin K. Kuntz address the potential psychiatric sequela of shocks from implanted cardiac defibrillators (ICDs). Depression is a known risk factor for developing and dying from cardiac disease,1 and recent evidence suggests that anxiety disorders also are associated with increased cardiac risk.2 Autonomic dysfunction is 1 potential mechanism that could explain the increased cardiac mortality associated with depression and anxiety. Patients with depression have a higher risk of ICD shocks induced by ventricular arrhythmias than nondepressed patients.3 This suggests a vicious circle when anxiety and depression could increase arrhythmic risk through autonomic dysfunction, and arrhythmias leading to ICD shocks may cause or worsen depression and anxiety.
Drs. Gibson and Kuntz also describe how cognitive-behavioral therapy (CBT) can reduce anxiety and depressive symptoms associated with ICD shocks. A paper published last year described how CBT may help decrease ICD shocks.4 Therefore it is possible that psychotherapeutic interventions such as CBT may do more than help improve depression and anxiety symptoms, whether preceded or caused by ICD shocks. CBT also might help reduce ventricular arrhythmias. This is important because we do not have good empiric evidence that current treatments for depression—pharmacologic or psychotherapeutic—reduce cardiac risk.1
A recent paper suggests that autonomic dysfunction may improve with sertraline treatment.5 However, the rationale for treating cardiac patients for depression and anxiety should be improvement of psychiatric symptoms and not an as-yet-unfounded belief that such treatments may also reduce the cardiac mortality and morbidity.
Jonas Hannestad, MD, PhD
Department of psychiatry
Yale University
New Haven, CT
1. Rivelli S, Jiang W. Depression and ischemic heart disease: what have we learned from clinical trials? Curr Opin Cardiol 2007;22(4):286-91.
2. Bedi US, Arora R. Cardiovascular manifestations of posttraumatic stress disorder. J Natl Med Assoc 2007;99(6):642-9.
3. Whang W, Albert CM, Sears SF, Jr, et al. Depression as a predictor for appropriate shocks among patients with implantable cardioverter-defibrillators: results from the Triggers of Ventricular Arrhythmias (TOVA) study. J Am Coll Cardiol 2005;45(7):1090-5.
4. Chevalier P, Cottraux J, Mollard E, et al. Prevention of implantable defibrillator shocks by cognitive behavioral therapy: a pilot trial. Am Heart J 2006;151(1):191.-
5. Glassman AH, Bigger JT, Gaffney M, Van Zyl LT. Heart rate variability in acute coronary syndrome patients with major depression: influence of sertra-line and mood improvement. Arch Gen Psychiatry 2007;64(9):1025-31.
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] or visit CurrentPsychiatry.com and click on the “Contact Us” link.
In “Managing anxiety in patients with implanted cardiac defibrillators” (Current Psychiatry, September 2007), Drs. Douglas P. Gibson and Kristin K. Kuntz address the potential psychiatric sequela of shocks from implanted cardiac defibrillators (ICDs). Depression is a known risk factor for developing and dying from cardiac disease,1 and recent evidence suggests that anxiety disorders also are associated with increased cardiac risk.2 Autonomic dysfunction is 1 potential mechanism that could explain the increased cardiac mortality associated with depression and anxiety. Patients with depression have a higher risk of ICD shocks induced by ventricular arrhythmias than nondepressed patients.3 This suggests a vicious circle when anxiety and depression could increase arrhythmic risk through autonomic dysfunction, and arrhythmias leading to ICD shocks may cause or worsen depression and anxiety.
Drs. Gibson and Kuntz also describe how cognitive-behavioral therapy (CBT) can reduce anxiety and depressive symptoms associated with ICD shocks. A paper published last year described how CBT may help decrease ICD shocks.4 Therefore it is possible that psychotherapeutic interventions such as CBT may do more than help improve depression and anxiety symptoms, whether preceded or caused by ICD shocks. CBT also might help reduce ventricular arrhythmias. This is important because we do not have good empiric evidence that current treatments for depression—pharmacologic or psychotherapeutic—reduce cardiac risk.1
A recent paper suggests that autonomic dysfunction may improve with sertraline treatment.5 However, the rationale for treating cardiac patients for depression and anxiety should be improvement of psychiatric symptoms and not an as-yet-unfounded belief that such treatments may also reduce the cardiac mortality and morbidity.
Jonas Hannestad, MD, PhD
Department of psychiatry
Yale University
New Haven, CT
In “Managing anxiety in patients with implanted cardiac defibrillators” (Current Psychiatry, September 2007), Drs. Douglas P. Gibson and Kristin K. Kuntz address the potential psychiatric sequela of shocks from implanted cardiac defibrillators (ICDs). Depression is a known risk factor for developing and dying from cardiac disease,1 and recent evidence suggests that anxiety disorders also are associated with increased cardiac risk.2 Autonomic dysfunction is 1 potential mechanism that could explain the increased cardiac mortality associated with depression and anxiety. Patients with depression have a higher risk of ICD shocks induced by ventricular arrhythmias than nondepressed patients.3 This suggests a vicious circle when anxiety and depression could increase arrhythmic risk through autonomic dysfunction, and arrhythmias leading to ICD shocks may cause or worsen depression and anxiety.
Drs. Gibson and Kuntz also describe how cognitive-behavioral therapy (CBT) can reduce anxiety and depressive symptoms associated with ICD shocks. A paper published last year described how CBT may help decrease ICD shocks.4 Therefore it is possible that psychotherapeutic interventions such as CBT may do more than help improve depression and anxiety symptoms, whether preceded or caused by ICD shocks. CBT also might help reduce ventricular arrhythmias. This is important because we do not have good empiric evidence that current treatments for depression—pharmacologic or psychotherapeutic—reduce cardiac risk.1
A recent paper suggests that autonomic dysfunction may improve with sertraline treatment.5 However, the rationale for treating cardiac patients for depression and anxiety should be improvement of psychiatric symptoms and not an as-yet-unfounded belief that such treatments may also reduce the cardiac mortality and morbidity.
Jonas Hannestad, MD, PhD
Department of psychiatry
Yale University
New Haven, CT
1. Rivelli S, Jiang W. Depression and ischemic heart disease: what have we learned from clinical trials? Curr Opin Cardiol 2007;22(4):286-91.
2. Bedi US, Arora R. Cardiovascular manifestations of posttraumatic stress disorder. J Natl Med Assoc 2007;99(6):642-9.
3. Whang W, Albert CM, Sears SF, Jr, et al. Depression as a predictor for appropriate shocks among patients with implantable cardioverter-defibrillators: results from the Triggers of Ventricular Arrhythmias (TOVA) study. J Am Coll Cardiol 2005;45(7):1090-5.
4. Chevalier P, Cottraux J, Mollard E, et al. Prevention of implantable defibrillator shocks by cognitive behavioral therapy: a pilot trial. Am Heart J 2006;151(1):191.-
5. Glassman AH, Bigger JT, Gaffney M, Van Zyl LT. Heart rate variability in acute coronary syndrome patients with major depression: influence of sertra-line and mood improvement. Arch Gen Psychiatry 2007;64(9):1025-31.
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] or visit CurrentPsychiatry.com and click on the “Contact Us” link.
1. Rivelli S, Jiang W. Depression and ischemic heart disease: what have we learned from clinical trials? Curr Opin Cardiol 2007;22(4):286-91.
2. Bedi US, Arora R. Cardiovascular manifestations of posttraumatic stress disorder. J Natl Med Assoc 2007;99(6):642-9.
3. Whang W, Albert CM, Sears SF, Jr, et al. Depression as a predictor for appropriate shocks among patients with implantable cardioverter-defibrillators: results from the Triggers of Ventricular Arrhythmias (TOVA) study. J Am Coll Cardiol 2005;45(7):1090-5.
4. Chevalier P, Cottraux J, Mollard E, et al. Prevention of implantable defibrillator shocks by cognitive behavioral therapy: a pilot trial. Am Heart J 2006;151(1):191.-
5. Glassman AH, Bigger JT, Gaffney M, Van Zyl LT. Heart rate variability in acute coronary syndrome patients with major depression: influence of sertra-line and mood improvement. Arch Gen Psychiatry 2007;64(9):1025-31.
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] or visit CurrentPsychiatry.com and click on the “Contact Us” link.
Innovative polypharmacy: When dopamine blockade is not enough
Polypharmacy has been a 4-letter word for a long time in schizophrenia management. Prescribing more than 1 antipsychotic to a patient with refractory symptoms has evoked images of a potentially harmful, nonevidence-based cocktail with no proven advantage over monotherapy.
Compare schizophrenia with bipolar disorder, for which combination therapy—an antipsychotic plus a mood stabilizer plus an antidepressant/ antianxiety agent—is the standard of care. Similarly, augmentation therapy is viewed as necessary for difficult cases of unipolar depression.
In the United States, approximately 40% of schizophrenia patients receive 2 or more concomitant antipsychotics (atypical and conventional agents).1 Clearly, many clinicians resort to antipsychotic polypharmacy in a desperate attempt to manage chronic, treatment-resistant illness, even though no published data support this practice.
This situation may be changing, however, because of evolving under-standings of schizophrenia’s neurobiology. Before long, clinicians may employ concomitant agents with different mechanisms in novel approaches to improve outcomes in patients with schizophrenia.
Novel polypharmacy. The dopamine pathways approach is inadequate for achieving true remission across all of chronic schizophrenia’s symptom domains. Positive and negative symptoms and cognitive impairment that persist despite antipsychotic therapy call for new treatment approaches. Here are some of my speculations—suggested by emerging data about schizophrenia’s pathophysiology—about “futuristic” adjuncts to antipsychotics:
Add a glutamate modulator (such as lamotrigine). This combination has shown benefit in patients who have not responded to clozapine.2 Many lines of evidence show that the glutamate system is impaired in schizophrenia, and this approach is promising.
Add a GABA agonist (such as valproate or benzodiazepines). Recent findings of a GABA deficit in frontal lobe chandelier cells in schizophrenia gives this combination legitimacy.3
Add an anti-inflammatory agent (such as a COX-2 inhibitor). Several studies report increased inflammatory cytokines in patients with schizophrenia. Others have found an antipsychotic/anti-inflammatory combination more efficacious than an antipsychotic alone.4
Add a cognitive enhancing agent. Antipsychotics as monotherapy fail to reverse schizophrenia’s severe cognitive deficits (~2 standard deviations below healthy individuals’ cognition).The National Institute of Mental Health-sponsored MATRICS initiative (Measurement and Treatment Research to Improve Cognition in Schizophrenia)5 is testing potential neuro-protective and myelin-repair agents to improve deficits in memory, attention, and executive function in schizophrenia. Potential mechanisms include alpha 7 nicotinic receptor agonists, D1 receptor agonists, or AMPA glutamatergic receptor agonists. These agents might become available in a few years.
Add a neuroprotective agent. As a neurodegenerative disorder, schizophrenia could benefit from induction of neurotrophic factors (such as nerve growth factor [NGF], brain-derived neurotrophic factor [BDNF], or vascular endothelial growth factor [VEGF]) and neurogenesis stimulation. Atypical antipsychotics —but not typical agents—have shown neurotrophic activity,6 but combining them with other neurotrophic agents such as lithium or selective serotonin reuptake inhibitors7 might expedite brain tissue regeneration and improve patients’ function.
Add a myelin repair agent. Many schizophrenia studies suggest impaired myelin (white-matter) structure, which may explain the “disconnection” among brain regions that results in thought disorder. A recent report indicates that citalopram restored white-matter integrity in patients with obsessive-compulsive disorder after a few weeks of use.8 If these results are replicated in schizophrenia, antipsychotic-myelin repair agent combinations may become a rational polypharmacy.
What lies ahead. Future schizophrenia treatment almost certainly will include drug combinations that:
- address clinical domains not managed with current antipsychotic monotherapy
- help override treatment resistance or refractoriness (hallucinations or delusions).
Combinations of 3 or more drugs often are used to treat serious medical disorders such as cancer, HIV, or malignant hypertension. Management of a severe, disabling psychiatric disorder such as schizophrenia should be no less aggressive.
1. Broekema WJ, de Groot IW, van Harten PN. Simultaneous prescribing of atypical antipsychotics, conventional antipsychotics and anticholinergics—a European study. Pharm World Sci 2007;29:126-30.
2. Zoccali R, Muscatello MR, Bruno A, et al. The effect of lamotrigine augmentation of clozapine in a sample of treatment-resistant schizophrenic patients: a double-blind, placebo-controlled study. Schizophr Res 2007;93:109-16.
3. Konopaske GT, Sweet RA, Wu Q, et al. Regional specificity of chandelier neuron axon terminal alterations in schizophrenia. Neuroscience 2006;90:189-95.
4. Akhondzadeh S, Tabatabaee M, Amini H, et al. Celecoxib as adjunctive therapy in schizophrenia: a double-blind, randomized and placebo-controlled trial. Schizophr Res 2007;90(1-3):179-85.
5. Marder SR. The NIMH-MATRICS project for developing cognition-enhancing agents for schizophrenia. Dialogues Clin Neurosci 2006;8:109-13.
6. Pillai A, Terry AV, Jr, Mahadik SP. Differential effects of long-term treatment with typical and atypical antipsychotics on NGF and BDNF levels in rat striatum and hippocampus. Schizophr Res 2006;82:95-106.
7. Duman RS, Monteggia LM. A neurotrophic model for stress-related mood disorders. Biol Psychiat 2006;59:1116-27.
8. Yoo SY, Jang JH, Shin YW, et al. White matter abnormalities in drug-naïve patients with obsessive-compulsive disorder: a diffusion tensor study before and after citalopram treatment. Acta Psychiatr Scand 2007;116:211-9.
Polypharmacy has been a 4-letter word for a long time in schizophrenia management. Prescribing more than 1 antipsychotic to a patient with refractory symptoms has evoked images of a potentially harmful, nonevidence-based cocktail with no proven advantage over monotherapy.
Compare schizophrenia with bipolar disorder, for which combination therapy—an antipsychotic plus a mood stabilizer plus an antidepressant/ antianxiety agent—is the standard of care. Similarly, augmentation therapy is viewed as necessary for difficult cases of unipolar depression.
In the United States, approximately 40% of schizophrenia patients receive 2 or more concomitant antipsychotics (atypical and conventional agents).1 Clearly, many clinicians resort to antipsychotic polypharmacy in a desperate attempt to manage chronic, treatment-resistant illness, even though no published data support this practice.
This situation may be changing, however, because of evolving under-standings of schizophrenia’s neurobiology. Before long, clinicians may employ concomitant agents with different mechanisms in novel approaches to improve outcomes in patients with schizophrenia.
Novel polypharmacy. The dopamine pathways approach is inadequate for achieving true remission across all of chronic schizophrenia’s symptom domains. Positive and negative symptoms and cognitive impairment that persist despite antipsychotic therapy call for new treatment approaches. Here are some of my speculations—suggested by emerging data about schizophrenia’s pathophysiology—about “futuristic” adjuncts to antipsychotics:
Add a glutamate modulator (such as lamotrigine). This combination has shown benefit in patients who have not responded to clozapine.2 Many lines of evidence show that the glutamate system is impaired in schizophrenia, and this approach is promising.
Add a GABA agonist (such as valproate or benzodiazepines). Recent findings of a GABA deficit in frontal lobe chandelier cells in schizophrenia gives this combination legitimacy.3
Add an anti-inflammatory agent (such as a COX-2 inhibitor). Several studies report increased inflammatory cytokines in patients with schizophrenia. Others have found an antipsychotic/anti-inflammatory combination more efficacious than an antipsychotic alone.4
Add a cognitive enhancing agent. Antipsychotics as monotherapy fail to reverse schizophrenia’s severe cognitive deficits (~2 standard deviations below healthy individuals’ cognition).The National Institute of Mental Health-sponsored MATRICS initiative (Measurement and Treatment Research to Improve Cognition in Schizophrenia)5 is testing potential neuro-protective and myelin-repair agents to improve deficits in memory, attention, and executive function in schizophrenia. Potential mechanisms include alpha 7 nicotinic receptor agonists, D1 receptor agonists, or AMPA glutamatergic receptor agonists. These agents might become available in a few years.
Add a neuroprotective agent. As a neurodegenerative disorder, schizophrenia could benefit from induction of neurotrophic factors (such as nerve growth factor [NGF], brain-derived neurotrophic factor [BDNF], or vascular endothelial growth factor [VEGF]) and neurogenesis stimulation. Atypical antipsychotics —but not typical agents—have shown neurotrophic activity,6 but combining them with other neurotrophic agents such as lithium or selective serotonin reuptake inhibitors7 might expedite brain tissue regeneration and improve patients’ function.
Add a myelin repair agent. Many schizophrenia studies suggest impaired myelin (white-matter) structure, which may explain the “disconnection” among brain regions that results in thought disorder. A recent report indicates that citalopram restored white-matter integrity in patients with obsessive-compulsive disorder after a few weeks of use.8 If these results are replicated in schizophrenia, antipsychotic-myelin repair agent combinations may become a rational polypharmacy.
What lies ahead. Future schizophrenia treatment almost certainly will include drug combinations that:
- address clinical domains not managed with current antipsychotic monotherapy
- help override treatment resistance or refractoriness (hallucinations or delusions).
Combinations of 3 or more drugs often are used to treat serious medical disorders such as cancer, HIV, or malignant hypertension. Management of a severe, disabling psychiatric disorder such as schizophrenia should be no less aggressive.
Polypharmacy has been a 4-letter word for a long time in schizophrenia management. Prescribing more than 1 antipsychotic to a patient with refractory symptoms has evoked images of a potentially harmful, nonevidence-based cocktail with no proven advantage over monotherapy.
Compare schizophrenia with bipolar disorder, for which combination therapy—an antipsychotic plus a mood stabilizer plus an antidepressant/ antianxiety agent—is the standard of care. Similarly, augmentation therapy is viewed as necessary for difficult cases of unipolar depression.
In the United States, approximately 40% of schizophrenia patients receive 2 or more concomitant antipsychotics (atypical and conventional agents).1 Clearly, many clinicians resort to antipsychotic polypharmacy in a desperate attempt to manage chronic, treatment-resistant illness, even though no published data support this practice.
This situation may be changing, however, because of evolving under-standings of schizophrenia’s neurobiology. Before long, clinicians may employ concomitant agents with different mechanisms in novel approaches to improve outcomes in patients with schizophrenia.
Novel polypharmacy. The dopamine pathways approach is inadequate for achieving true remission across all of chronic schizophrenia’s symptom domains. Positive and negative symptoms and cognitive impairment that persist despite antipsychotic therapy call for new treatment approaches. Here are some of my speculations—suggested by emerging data about schizophrenia’s pathophysiology—about “futuristic” adjuncts to antipsychotics:
Add a glutamate modulator (such as lamotrigine). This combination has shown benefit in patients who have not responded to clozapine.2 Many lines of evidence show that the glutamate system is impaired in schizophrenia, and this approach is promising.
Add a GABA agonist (such as valproate or benzodiazepines). Recent findings of a GABA deficit in frontal lobe chandelier cells in schizophrenia gives this combination legitimacy.3
Add an anti-inflammatory agent (such as a COX-2 inhibitor). Several studies report increased inflammatory cytokines in patients with schizophrenia. Others have found an antipsychotic/anti-inflammatory combination more efficacious than an antipsychotic alone.4
Add a cognitive enhancing agent. Antipsychotics as monotherapy fail to reverse schizophrenia’s severe cognitive deficits (~2 standard deviations below healthy individuals’ cognition).The National Institute of Mental Health-sponsored MATRICS initiative (Measurement and Treatment Research to Improve Cognition in Schizophrenia)5 is testing potential neuro-protective and myelin-repair agents to improve deficits in memory, attention, and executive function in schizophrenia. Potential mechanisms include alpha 7 nicotinic receptor agonists, D1 receptor agonists, or AMPA glutamatergic receptor agonists. These agents might become available in a few years.
Add a neuroprotective agent. As a neurodegenerative disorder, schizophrenia could benefit from induction of neurotrophic factors (such as nerve growth factor [NGF], brain-derived neurotrophic factor [BDNF], or vascular endothelial growth factor [VEGF]) and neurogenesis stimulation. Atypical antipsychotics —but not typical agents—have shown neurotrophic activity,6 but combining them with other neurotrophic agents such as lithium or selective serotonin reuptake inhibitors7 might expedite brain tissue regeneration and improve patients’ function.
Add a myelin repair agent. Many schizophrenia studies suggest impaired myelin (white-matter) structure, which may explain the “disconnection” among brain regions that results in thought disorder. A recent report indicates that citalopram restored white-matter integrity in patients with obsessive-compulsive disorder after a few weeks of use.8 If these results are replicated in schizophrenia, antipsychotic-myelin repair agent combinations may become a rational polypharmacy.
What lies ahead. Future schizophrenia treatment almost certainly will include drug combinations that:
- address clinical domains not managed with current antipsychotic monotherapy
- help override treatment resistance or refractoriness (hallucinations or delusions).
Combinations of 3 or more drugs often are used to treat serious medical disorders such as cancer, HIV, or malignant hypertension. Management of a severe, disabling psychiatric disorder such as schizophrenia should be no less aggressive.
1. Broekema WJ, de Groot IW, van Harten PN. Simultaneous prescribing of atypical antipsychotics, conventional antipsychotics and anticholinergics—a European study. Pharm World Sci 2007;29:126-30.
2. Zoccali R, Muscatello MR, Bruno A, et al. The effect of lamotrigine augmentation of clozapine in a sample of treatment-resistant schizophrenic patients: a double-blind, placebo-controlled study. Schizophr Res 2007;93:109-16.
3. Konopaske GT, Sweet RA, Wu Q, et al. Regional specificity of chandelier neuron axon terminal alterations in schizophrenia. Neuroscience 2006;90:189-95.
4. Akhondzadeh S, Tabatabaee M, Amini H, et al. Celecoxib as adjunctive therapy in schizophrenia: a double-blind, randomized and placebo-controlled trial. Schizophr Res 2007;90(1-3):179-85.
5. Marder SR. The NIMH-MATRICS project for developing cognition-enhancing agents for schizophrenia. Dialogues Clin Neurosci 2006;8:109-13.
6. Pillai A, Terry AV, Jr, Mahadik SP. Differential effects of long-term treatment with typical and atypical antipsychotics on NGF and BDNF levels in rat striatum and hippocampus. Schizophr Res 2006;82:95-106.
7. Duman RS, Monteggia LM. A neurotrophic model for stress-related mood disorders. Biol Psychiat 2006;59:1116-27.
8. Yoo SY, Jang JH, Shin YW, et al. White matter abnormalities in drug-naïve patients with obsessive-compulsive disorder: a diffusion tensor study before and after citalopram treatment. Acta Psychiatr Scand 2007;116:211-9.
1. Broekema WJ, de Groot IW, van Harten PN. Simultaneous prescribing of atypical antipsychotics, conventional antipsychotics and anticholinergics—a European study. Pharm World Sci 2007;29:126-30.
2. Zoccali R, Muscatello MR, Bruno A, et al. The effect of lamotrigine augmentation of clozapine in a sample of treatment-resistant schizophrenic patients: a double-blind, placebo-controlled study. Schizophr Res 2007;93:109-16.
3. Konopaske GT, Sweet RA, Wu Q, et al. Regional specificity of chandelier neuron axon terminal alterations in schizophrenia. Neuroscience 2006;90:189-95.
4. Akhondzadeh S, Tabatabaee M, Amini H, et al. Celecoxib as adjunctive therapy in schizophrenia: a double-blind, randomized and placebo-controlled trial. Schizophr Res 2007;90(1-3):179-85.
5. Marder SR. The NIMH-MATRICS project for developing cognition-enhancing agents for schizophrenia. Dialogues Clin Neurosci 2006;8:109-13.
6. Pillai A, Terry AV, Jr, Mahadik SP. Differential effects of long-term treatment with typical and atypical antipsychotics on NGF and BDNF levels in rat striatum and hippocampus. Schizophr Res 2006;82:95-106.
7. Duman RS, Monteggia LM. A neurotrophic model for stress-related mood disorders. Biol Psychiat 2006;59:1116-27.
8. Yoo SY, Jang JH, Shin YW, et al. White matter abnormalities in drug-naïve patients with obsessive-compulsive disorder: a diffusion tensor study before and after citalopram treatment. Acta Psychiatr Scand 2007;116:211-9.
After the ‘pink clouds,’ he sees red
HISTORY: Depressed and sick
Mr. T, age 53, was diagnosed last year with hepatitis C and for 20 years has battled recurrent major depression with euthymia between episodes. His hepatologist asks us to evaluate his recent depressed mood and erratic behavior.
Less than 2 months ago, the hepatologist prescribed ribavirin, 1,000 mg bid, and peginterferon alfa-2B, 10 million IU/1.0 mL weekly, for hepatitis C. Soon afterward, Mr. T became irritable, especially toward his wife. He now refuses to leave his house most days because of overwhelming sadness and hopelessness. Once an avid motorcycle enthusiast, Mr. T has stopped riding and complains of fatigue, “fuzzy” thinking, and diminished concentration, but he denies suicidal thoughts or intent. He weighs 232 lb but has lost 15 lb in the last 6 weeks.
Five weeks ago, the hepatologist added bupropion XL, 150 mg/d, for Mr. T’s depressive symptoms, but the patient complained that the antidepressant “amped me up” and “made my mind race.” After 3 weeks, the hepatologist switched to escitalopram, 10 mg/d, but Mr. T’s agitation continued.
Several days after starting escitalopram, Mr. T experienced what he calls a “pink cloud” period—intensely pleasurable episodes that he says began in late childhood, usually last about 4 days, and occur 6 times annually. During these episodes, his thoughts race, his speech is mildly pressured, and he sleeps 5 hours or less nightly. While euphoric, he drives his motorcycle at 100 mph, starts several projects at once, and is distractible.
Once the “pink clouds” clear, Mr. T feels fatigued and “let down” as he does now. He says he has never reported these euphoric periods because he usually enjoys them.
Mr. T also has longstanding anxiety. Most days he is “on edge” and restless, feels muscle tension in his neck, and has trouble falling and staying sleep. After changing jobs last year, he began having panic attacks triggered by excessive worry. He denies anticipatory fear or avoidance, so we rule out panic disorder.
Additionally, Mr. T has been engaging in weekly binge-eating episodes during which he consumes nearly 50 large-sized cookies and 2 to 3 2-ounce bags of potato chips in 2 hours. He is wracked with guilt after bingeing and often feels embarrassed about being overweight (body mass index, 31 kg/m2). He does not purge but moderately restricts his diet between binges. He says he started bingeing at age 20, and at one point was bingeing 3 times a week.
Mr. T also complains that ribavirin and peginterferon are causing headaches, fatigue, and myalgias. He also takes hydrochlorothiazide, 25 mg/d, for hypertension, and is allergic to sulfonamides. He denies using alcohol and drugs but smokes 2 packs of cigarettes per day.
We diagnose bipolar II disorder based on Mr. T’s extreme mood shifts, history of major depressive episodes, recent hypomania, lack of manic or mixed episodes, and significant distress. His hypomania episodes last <1 week; episodes that last ≥1 week or require hospitalization would signal bipolar I disorder.
We rule out interferon-induced depression and hypomania1 because Mr. T showed signs of mood dysfunction long before he contracted hepatitis C. We also diagnose generalized anxiety disorder and eating disorder, not otherwise specified.
The authors’ observations
Diagnosing and managing bipolar disorder is challenging, especially when hypomania is not readily apparent.2
After we discuss treatment options with Mr. T, he chooses lamotrigine because it causes relatively few side effects and is less likely than valproic acid and other mood-stabilizing anticonvulsants to cause hepatotoxicity or pancreatitis.3 Lamotrigine also might reduce Mr. T’s anxiety.4
We do not try lithium because Mr. T is taking a diuretic (hydrochlorothiazide), which can cause lithium toxicity when used concomitantly. Also, lithium requires close laboratory monitoring, interacts with many medications, and can cause drowsiness, dry mouth, blurry vision, and fatigue.5 These
factors contraindicate lithium for Mr. T, who is taking several medications and suffers side effects from ribavirin and interferon.
Olanzapine might control Mr. T’s mood swings, but the neuroleptic can cause weight gain and metabolic syndrome6 and might complicate his eating disorder.
TREATMENT: A ‘rash’ reaction
We add lamotrigine, 25 mg/d, for 2 weeks and then increase to 50 mg/d.
Two days after the lamotrigine increase, Mr. T reports a rash on the left side of his trunk and left hip, buttock, and elbow (Figure). He also complains of mild chills and night sweats, although these symptoms emerged several weeks ago. He denies blistering, fevers, dysuria, nausea, or vomiting. We see no signs of lymphadenopathy, and mucosae are unaffected. Since he started lamotrigine, he says, he has not tried unfamiliar brands of shampoos, laundry detergents, or shower gels that might irritate his skin.
Figure: Did lamotrigine cause Mr. T’s rash?
Folliculocentric pustules around patient’s left elbow and throughout his left side.We have Mr. T come in that day for an emergency physical examination. At presentation, the rash appears infectious with isolated pustules throughout. We refer him to a dermatologist for same-day evaluation.
The authors’ observations
A rash is an immunologic reaction to an offending agent. If lamotrigine were causing the rash, lowering the dosage would not mitigate it.
We continued lamotrigine because the dermatologist could examine the rash within 24 hours of Mr. T’s complaint. Also, the agent was decreasing the patient’s mood, irritability, and anger. If we believed lamotrigine was causing the rash and could not obtain an immediate dermatology consult, we would have stopped the medication.
FOLLOW-UP: ‘Hot’ findings
During the patient history interview, the dermatologist discovers that Mr. T recently installed a whirlpool bath, and that the eruption occurred 3 to 5 days after the patient first used it. Physical examination shows groups of discrete folliculocentric pustules with surrounding erythema mainly on his extensor surfaces and left buttock. These findings and Mr. T’s history suggest a skin infection.
The dermatologist diagnoses hot tub folliculitis, an infection caused by exposure to contaminated whirlpools, hot tubs, or water slides. Cultures obtained that day grow Pseudomonas aeruginosa, confirming the diagnosis. The dermatologist tells Mr. T to stop using his whirlpool bath and prescribes topical gentamicin and ciprofloxacin, 500 mg bid for 10 days. We continue lamotrigine based on the dermatologist’s recommendation.
Two weeks later, Mr. T’s eruption resolves, and we increase lamotrigine to 100 mg/d, which improves his mood and achieves steady-state effectiveness.7 We continue escitalopram, 10 mg/d, then increase to 20 mg/d to treat his generalized anxiety. Mr. T begins experiencing anorgasmia 1 week after the escitalopram increase, so we switch to buspirone, 15 mg bid. After another 4 weeks, his anger, irritability, panic attacks, anxiety, and depression have diminished.
After 3 months, Mr. T’s hepatologist stops ribavirin and peginterferon because they are not helping his hepatitis C infection. Days later, Mr. T’s chills, sweats, and fatigue remit.
The hepatologist considers an experimental hepatitis C
medication.
We see Mr. T once monthly for supportive psychotherapy and medication management. Despite divorce proceedings and persistent mild depression he is optimistic, enjoys work, and rides his motorcycle safely twice a week.
The authors’ observations
Although Mr. T’s presentation and patient history clearly suggested an independent skin infection, distinguishing between an infection and anticonvulsant-induced rash can be difficult.
Lamotrigine and other antiepileptics (Table 1)8 have been associated with morbilliform eruptions, anticonvulsant hypersensitivity syndrome, erythema multiforme, Stevens-Johnson syndrome (SJS), and toxic epidermal necrolysis (TEN), a severe form of SJS with a 20% to 30% mortality rate.9,10
Table 1
Estimated risk of severe rash among first-time antiepileptic users*
| Drug | Total new users† | Total SJS/TEN cases | Risk per 10,000 new users |
|---|---|---|---|
| Carbamazepine | 286,360 | 39 | 1.4 |
| Lamotrigine | 55,154 | 14 | 2.5 |
| Phenobarbital | 8,659 | 7 | 8.1 |
| Phenytoin | 36,171 | 30 | 8.3 |
| Valproic acid | 103,150 | 4 | 0.4 |
| * Researchers reviewed records of patients hospitalized between 1998 and 2001 with SJS or TEN after using an anticonvulsant. | |||
| † Estimates based on number of dispensed prescriptions, average prescribed dosages, and duration of anticonvulsant use as recorded in Germany’s Mediplus database. | |||
| SJS: Stevens-Johnson syndrome; TEN: toxic epidermal necrolysis | |||
| Source: Adapted from reference 8 | |||
Although most lamotrigine-induced cutaneous eruptions are mild or self-limited, some are severe and potentially fatal. In clinical trials, approximately 10% of patients receiving lamotrigine for epilepsy developed cutaneous reactions.11 Among 3,348 patients with epilepsy who received lamotrigine, 11 (0.3%) required hospitalization for SJS or TEN.11
Anticonvulsant hypersensitivity syndrome, estimated to occur once per 1,000 to 10,000 exposures to anticonvulsants,12 can lead to fever, lymphadenopathy, hepatomegaly, and arthralgias. Although hypersensitivity to aromatic anticonvulsants such as phenytoin, carbamazepine, or phenobarbital is most common, hypersensitivity to lamotrigine also has been reported.13,14
Roughly 90% of patients with anticonvulsant hypersensitivity syndrome develop leukocytosis with eosinophilia, and some develop leukocytosis with agranulocytosis.15-17 Fulminant hepatitis can occur, which leads to most deaths associated with this syndrome.
4 steps to gauging rash
Taking a thorough history, examining the eruption, ordering liver function tests (LFTs) and a complete blood count (CBC), and referring the patient to a dermatologist are key to determining the seriousness of an eruption and planning treatment in patients taking anticonvulsants (Table 2). See the patient within 12 hours after he reports the rash, as SJS and TEN often progress rapidly.
Table 2
4 steps to determining rash severity and cause
| Take a thorough history | Find out when eruption occurred and when patient started the anticonvulsant |
| Ask about past rashes, other medicines, and family history of reactions to medications | |
| Find out if patient has had fever/chills, malaise, lymphadenopathy, or mucosal symptoms such as photophobia or dysuria | |
| Examine the eruption | Examine for mucosal involvement, facial edema, and blistering; describe the symmetry and extent of involvement |
| Look for systemic findings such as fever, chills, lymphadenopathy, and organomegaly | |
| Photograph the eruption for the dermatologist if possible | |
| Order laboratory tests | Order liver function tests and complete blood count with differentials; assess for eosinophilia |
| Closely monitor patient | Stop anticonvulsant if history, physical findings suggest a drug-induced eruption |
| Refer patient to a dermatologist |
STEP 1: Take a thorough history
Ask the patient:
What medications are you taking? Because more than 100 medications could cause SJS or TEN, a detailed drug history is critical to determining whether a medication has induced the eruption.
When did you start taking the potentially offending medication? True lamotrigine-induced eruptions usually occur 5 days to 8 weeks after the first dose.10 SJS and TEN generally take 1 to 2 weeks to develop.
What is your current dosage? Has it increased or decreased recently? Rapid lamotrigine dosage escalations or use of lamotrigine with valproic acid can cause severe rash.9,10,18 Valproic acid increases serum lamotrigine by inhibiting its hepatic metabolism, thereby raising side-effect risk. In clinical trials, 30% of patients who received both anticonvulsants developed a rash.10
Have any family members had rashes after taking an anticonvulsant? Compared with the general population, siblings and first-degree relatives of patients with anticonvulsant-related eruptions are at higher risk for this complication.19 Decreased epoxide hydrolase activity might negate these patients’ ability to detoxify the arene oxide metabolite, which can cause adverse effects if it accumulates.
Do you have other medical problems? Hepatitis C, for example, can theoretically increase lamotrigine’s half-life, thereby elevating side-effect risk.11
Watch for anticonvulsant-related adverse events in patients with hepatic insufficiency because hepatitis might hinder anticonvulsant metabolism.20 Other medical comorbidities—such as HIV infection and systemic lupus erythematosus—also could increase the risk of antiepileptic-induced rash.10
Have you had fever, chills, or other symptoms? Patients with SJS and TEN usually present with systemic symptoms such as malaise, rash, lymphadenopathy, mucosal lesions, and/or symptoms such as photophobia, difficulty swallowing, rectal erosions, or dysuria. Patients with anticonvulsant hypersensitivity syndrome typically have fever and associated arthralgias, skin pain, lymphadenopathy, or a burning sensation on their skin. These symptoms generally are absent in localized cutaneous infections.
STEP 2: Examine the eruption
Cutaneous SJS and TEN findings usually include abrupt onset of erythematous macules—which progress to targetoid lesions containing central bullae—followed by extensive epidermal necrosis. Superficial lip and mouth necrosis occur early, leading to severe stomatitis.
TEN and SJS can appear similar clinically, but TEN
covers >30% of body surface area, whereas SJS covers <10%.
Rashes that cover 10% to 30% of body surface suggest SJS-TEN overlap syndrome.
Anticonvulsant hypersensitivity syndrome usually manifests as
a morbilliform eruption on the face, arms, and/or torso. The
lesions might become edematous and progress to exfoliation or vesiculobullae. Facial edema is a hallmark of anticonvulsant hypersensitivity,15,16 and pustules and/or erythroderma might also appear. Other warning signs include symmetrical widespread eruption and organomegaly.
STEP 3: Order laboratory tests
Check liver function and order a CBC with differential to measure eosinophils. Eosinophilia and abnormal LFT results can signal anticonvulsant hypersensitivity.
Eosinophils. A normal eosinophil count ranges between 0% and 5% of peripheral blood leukocytes in adults, at a count of 350 to 650/cm. Although upper limits of normal vary, values >500/cm suggest hypereosinophilia.21
LFTs. Normal aspartate aminotransferase and alanine aminotransferase levels are 0 to 42 U/L and 0 to 48 U/L, respectively. Any LFT elevation could signal anticonvulsant hypersensitivity syndrome.
STEP 4: Closely monitor the patient
Discontinue the anticonvulsant if findings suggest a cutaneous drug reaction, and contact the patient’s primary care physician or dermatologist immediately. Early consultation with a dermatologist can help determine the eruption’s cause and reveal therapeutic options.
Dr. Pejic is chief resident in the adult psychiatry residency program, Louisiana State University Health Sciences Center and Ochsner Clinic Foundation, New Orleans.
Dr. Klinger is a third-year dermatology resident, Dr. Conrad is assistant professor of clinical psychiatry, and Dr. Nesbitt is chairman, department of dermatology, Louisiana State University Health Sciences Center.
Related Resources
- High WA. Stevens-Johnson syndrome and toxic epidermal necrolysis in adults. UpToDate Online (version 15.1); June 9, 2007. www.uptodate.com.
- Martin KA, Krahn LE, Rosati MJ, Balan V. Hepatitis C: How to manage mood during interferon treatment. Current Psychiatry 2006;5(11):69-80. http://www.currentpsychiatry.com/article_pages.asp?AID=4553.
Drug Brand Names
- Bupropion • Wellbutrin
- Buspirone • BuSpar
- Carbamazepine • Tegretol, Equetro, others
- Ciprofloxacin • Cipro, Proquin
- Escitalopram • Lexapro
- Hydrochlorothiazide • various
- Lamotrigine • Lamictal
- Lithium • Eskalith, others
- Olanzapine • Zyprexa
- Peginterferon alfa-2B • PEG-Intron
- Phenytoin • Dilantin
- Ribavirin • Copegus, Rebetol, others
- Valproic acid • Depakote
Disclosure
Dr. Conrad receives research/grant support from AstraZeneca, Bristol-Myers Squibb, Forest Pharmaceuticals, GlaxoSmithKline, and Wyeth.
Drs. Pejic, Nesbitt, and Klinger report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Crone CC, Gabriel GM, Wise TN. Managing the neuropsychiatric side effects of interferon-based therapy for hepatitis C. Cleve Clin J Med 2004;71(suppl 3):S27-S32.
2. Phelps JR, Ghaemi SN. Improving the diagnosis of bipolar disorder: predictive value of screening tests. J Affect Disord 2006;92:141-8.
3. Lacerda G, Krummel T, Sabourdy C, et al. Optimizing therapy of seizures in patients with renal or hepatic dysfunction. Neurology 2006;67(suppl 4):S28-S33.
4. Yumru M, Savas HA, Kurt E, et al. Atypical antipsychotics related metabolic syndrome in bipolar patients. J Affect Disord 2007;98:247-52.
5. Finley PR, Warner MD, Peabody CA. Clinical relevance of drug interactions with lithium. Clin Pharmacokinet 1995;3:172-91.
6. Keck PE, Jr, Strawn JR, McElroy SL. Pharmacologic treatment considerations in co-occurring bipolar and anxiety disorders. J Clin Psychiatry 2006;67(suppl 1):S8-S15.
7. Sachs GS, Printz DJ, Kahn DA, et al. The expert consensus guidelines: medication treatment of bipolar disorder 2000. A postgraduate medicine special report. New York: McGraw-Hill; 2000:24.
8. Mockenhaupt M, Messenheimer J, Tennis P, Schlingmann J. Risk of Stevens-Johnson syndrome and toxic epidermal necrolysis in new users of antiepileptics. Neurology 2005;64:1134-8.
9. Schlienger RG, Shapiro LE, Shear NH. Lamotrigine-induced severe cutaneous adverse reactions. Epilepsia 1998;29(suppl 7):S22-S26.
10. Calabrese JR, Sullivan JR, Bowden CL, et al. Rash in multicenter trials of lamotrigine in mood disorders: clinical relevance and management. J Clin Psychiatry 2002;63:1012-19.
11. Physicians’ desk reference. 61st ed. Montvale, NJ: Thomson PDR; 2007:1483-4, 1488.
12. Knowles SR, Shapiro LE, Shear NH. Anticonvulsant hypersensitivity syndrome: incidence, prevention and management. Drug Saf 1999;21:489-501.
13. Tennis P, Stern RS. Risk of serious cutaneous disorders after initiation of use of phenytoin, carbamazepine, or sodium valproate: a record linkage study. Neurology 1997;49:542-6.
14. Knowles SR, Shapiro LE, Shear NH. Anticonvulsant hypersensitivity syndrome: incidence, prevention and management. Drug Saf 1999;21:489-501.
15. Chang DK, Shear NH. Cutaneous reactions to anticonvulsants. Semin Neurol 1992;12:329-7.
16. Vittorio CC, Muglia JJ. Anticonvulsant hypersensitivity syndrome. Arch Intern Med 1995;155:2285-90.
17. Callot V, Roujeau JC, Bagot M, et al. Drug induced pseudo-lymphoma and hypersensitivity syndrome. Two different clinical entities. Arch Dermatol 1996;132:1315-21.
18. Yalcin B, Karaduman A. Stevens-Johnson syndrome associated with concomitant use of lamotrigine and valproic acid. J Am Acad Dermatol 2000;43:898-9.
19. Gennis MA, Vemuri R, Burns EA, et al. Familial occurrence of hypersensitivity to phenytoin. Am J Med 1991;91:631-4.
20. McLaren KD, Marangell LB. Special considerations in the treatment of patients with bipolar disorder and medical comorbidities. Ann Gen Hosp Psychiatry 2004;3:7.-
21. Valencak J, Ortiz-Urda S, Heere-Ress E, et al. Carbamazepine-induced DRESS syndrome with recurrent fever and exanthema. Int J Dermatol 2004;43:51-4.
HISTORY: Depressed and sick
Mr. T, age 53, was diagnosed last year with hepatitis C and for 20 years has battled recurrent major depression with euthymia between episodes. His hepatologist asks us to evaluate his recent depressed mood and erratic behavior.
Less than 2 months ago, the hepatologist prescribed ribavirin, 1,000 mg bid, and peginterferon alfa-2B, 10 million IU/1.0 mL weekly, for hepatitis C. Soon afterward, Mr. T became irritable, especially toward his wife. He now refuses to leave his house most days because of overwhelming sadness and hopelessness. Once an avid motorcycle enthusiast, Mr. T has stopped riding and complains of fatigue, “fuzzy” thinking, and diminished concentration, but he denies suicidal thoughts or intent. He weighs 232 lb but has lost 15 lb in the last 6 weeks.
Five weeks ago, the hepatologist added bupropion XL, 150 mg/d, for Mr. T’s depressive symptoms, but the patient complained that the antidepressant “amped me up” and “made my mind race.” After 3 weeks, the hepatologist switched to escitalopram, 10 mg/d, but Mr. T’s agitation continued.
Several days after starting escitalopram, Mr. T experienced what he calls a “pink cloud” period—intensely pleasurable episodes that he says began in late childhood, usually last about 4 days, and occur 6 times annually. During these episodes, his thoughts race, his speech is mildly pressured, and he sleeps 5 hours or less nightly. While euphoric, he drives his motorcycle at 100 mph, starts several projects at once, and is distractible.
Once the “pink clouds” clear, Mr. T feels fatigued and “let down” as he does now. He says he has never reported these euphoric periods because he usually enjoys them.
Mr. T also has longstanding anxiety. Most days he is “on edge” and restless, feels muscle tension in his neck, and has trouble falling and staying sleep. After changing jobs last year, he began having panic attacks triggered by excessive worry. He denies anticipatory fear or avoidance, so we rule out panic disorder.
Additionally, Mr. T has been engaging in weekly binge-eating episodes during which he consumes nearly 50 large-sized cookies and 2 to 3 2-ounce bags of potato chips in 2 hours. He is wracked with guilt after bingeing and often feels embarrassed about being overweight (body mass index, 31 kg/m2). He does not purge but moderately restricts his diet between binges. He says he started bingeing at age 20, and at one point was bingeing 3 times a week.
Mr. T also complains that ribavirin and peginterferon are causing headaches, fatigue, and myalgias. He also takes hydrochlorothiazide, 25 mg/d, for hypertension, and is allergic to sulfonamides. He denies using alcohol and drugs but smokes 2 packs of cigarettes per day.
We diagnose bipolar II disorder based on Mr. T’s extreme mood shifts, history of major depressive episodes, recent hypomania, lack of manic or mixed episodes, and significant distress. His hypomania episodes last <1 week; episodes that last ≥1 week or require hospitalization would signal bipolar I disorder.
We rule out interferon-induced depression and hypomania1 because Mr. T showed signs of mood dysfunction long before he contracted hepatitis C. We also diagnose generalized anxiety disorder and eating disorder, not otherwise specified.
The authors’ observations
Diagnosing and managing bipolar disorder is challenging, especially when hypomania is not readily apparent.2
After we discuss treatment options with Mr. T, he chooses lamotrigine because it causes relatively few side effects and is less likely than valproic acid and other mood-stabilizing anticonvulsants to cause hepatotoxicity or pancreatitis.3 Lamotrigine also might reduce Mr. T’s anxiety.4
We do not try lithium because Mr. T is taking a diuretic (hydrochlorothiazide), which can cause lithium toxicity when used concomitantly. Also, lithium requires close laboratory monitoring, interacts with many medications, and can cause drowsiness, dry mouth, blurry vision, and fatigue.5 These
factors contraindicate lithium for Mr. T, who is taking several medications and suffers side effects from ribavirin and interferon.
Olanzapine might control Mr. T’s mood swings, but the neuroleptic can cause weight gain and metabolic syndrome6 and might complicate his eating disorder.
TREATMENT: A ‘rash’ reaction
We add lamotrigine, 25 mg/d, for 2 weeks and then increase to 50 mg/d.
Two days after the lamotrigine increase, Mr. T reports a rash on the left side of his trunk and left hip, buttock, and elbow (Figure). He also complains of mild chills and night sweats, although these symptoms emerged several weeks ago. He denies blistering, fevers, dysuria, nausea, or vomiting. We see no signs of lymphadenopathy, and mucosae are unaffected. Since he started lamotrigine, he says, he has not tried unfamiliar brands of shampoos, laundry detergents, or shower gels that might irritate his skin.
Figure: Did lamotrigine cause Mr. T’s rash?
Folliculocentric pustules around patient’s left elbow and throughout his left side.We have Mr. T come in that day for an emergency physical examination. At presentation, the rash appears infectious with isolated pustules throughout. We refer him to a dermatologist for same-day evaluation.
The authors’ observations
A rash is an immunologic reaction to an offending agent. If lamotrigine were causing the rash, lowering the dosage would not mitigate it.
We continued lamotrigine because the dermatologist could examine the rash within 24 hours of Mr. T’s complaint. Also, the agent was decreasing the patient’s mood, irritability, and anger. If we believed lamotrigine was causing the rash and could not obtain an immediate dermatology consult, we would have stopped the medication.
FOLLOW-UP: ‘Hot’ findings
During the patient history interview, the dermatologist discovers that Mr. T recently installed a whirlpool bath, and that the eruption occurred 3 to 5 days after the patient first used it. Physical examination shows groups of discrete folliculocentric pustules with surrounding erythema mainly on his extensor surfaces and left buttock. These findings and Mr. T’s history suggest a skin infection.
The dermatologist diagnoses hot tub folliculitis, an infection caused by exposure to contaminated whirlpools, hot tubs, or water slides. Cultures obtained that day grow Pseudomonas aeruginosa, confirming the diagnosis. The dermatologist tells Mr. T to stop using his whirlpool bath and prescribes topical gentamicin and ciprofloxacin, 500 mg bid for 10 days. We continue lamotrigine based on the dermatologist’s recommendation.
Two weeks later, Mr. T’s eruption resolves, and we increase lamotrigine to 100 mg/d, which improves his mood and achieves steady-state effectiveness.7 We continue escitalopram, 10 mg/d, then increase to 20 mg/d to treat his generalized anxiety. Mr. T begins experiencing anorgasmia 1 week after the escitalopram increase, so we switch to buspirone, 15 mg bid. After another 4 weeks, his anger, irritability, panic attacks, anxiety, and depression have diminished.
After 3 months, Mr. T’s hepatologist stops ribavirin and peginterferon because they are not helping his hepatitis C infection. Days later, Mr. T’s chills, sweats, and fatigue remit.
The hepatologist considers an experimental hepatitis C
medication.
We see Mr. T once monthly for supportive psychotherapy and medication management. Despite divorce proceedings and persistent mild depression he is optimistic, enjoys work, and rides his motorcycle safely twice a week.
The authors’ observations
Although Mr. T’s presentation and patient history clearly suggested an independent skin infection, distinguishing between an infection and anticonvulsant-induced rash can be difficult.
Lamotrigine and other antiepileptics (Table 1)8 have been associated with morbilliform eruptions, anticonvulsant hypersensitivity syndrome, erythema multiforme, Stevens-Johnson syndrome (SJS), and toxic epidermal necrolysis (TEN), a severe form of SJS with a 20% to 30% mortality rate.9,10
Table 1
Estimated risk of severe rash among first-time antiepileptic users*
| Drug | Total new users† | Total SJS/TEN cases | Risk per 10,000 new users |
|---|---|---|---|
| Carbamazepine | 286,360 | 39 | 1.4 |
| Lamotrigine | 55,154 | 14 | 2.5 |
| Phenobarbital | 8,659 | 7 | 8.1 |
| Phenytoin | 36,171 | 30 | 8.3 |
| Valproic acid | 103,150 | 4 | 0.4 |
| * Researchers reviewed records of patients hospitalized between 1998 and 2001 with SJS or TEN after using an anticonvulsant. | |||
| † Estimates based on number of dispensed prescriptions, average prescribed dosages, and duration of anticonvulsant use as recorded in Germany’s Mediplus database. | |||
| SJS: Stevens-Johnson syndrome; TEN: toxic epidermal necrolysis | |||
| Source: Adapted from reference 8 | |||
Although most lamotrigine-induced cutaneous eruptions are mild or self-limited, some are severe and potentially fatal. In clinical trials, approximately 10% of patients receiving lamotrigine for epilepsy developed cutaneous reactions.11 Among 3,348 patients with epilepsy who received lamotrigine, 11 (0.3%) required hospitalization for SJS or TEN.11
Anticonvulsant hypersensitivity syndrome, estimated to occur once per 1,000 to 10,000 exposures to anticonvulsants,12 can lead to fever, lymphadenopathy, hepatomegaly, and arthralgias. Although hypersensitivity to aromatic anticonvulsants such as phenytoin, carbamazepine, or phenobarbital is most common, hypersensitivity to lamotrigine also has been reported.13,14
Roughly 90% of patients with anticonvulsant hypersensitivity syndrome develop leukocytosis with eosinophilia, and some develop leukocytosis with agranulocytosis.15-17 Fulminant hepatitis can occur, which leads to most deaths associated with this syndrome.
4 steps to gauging rash
Taking a thorough history, examining the eruption, ordering liver function tests (LFTs) and a complete blood count (CBC), and referring the patient to a dermatologist are key to determining the seriousness of an eruption and planning treatment in patients taking anticonvulsants (Table 2). See the patient within 12 hours after he reports the rash, as SJS and TEN often progress rapidly.
Table 2
4 steps to determining rash severity and cause
| Take a thorough history | Find out when eruption occurred and when patient started the anticonvulsant |
| Ask about past rashes, other medicines, and family history of reactions to medications | |
| Find out if patient has had fever/chills, malaise, lymphadenopathy, or mucosal symptoms such as photophobia or dysuria | |
| Examine the eruption | Examine for mucosal involvement, facial edema, and blistering; describe the symmetry and extent of involvement |
| Look for systemic findings such as fever, chills, lymphadenopathy, and organomegaly | |
| Photograph the eruption for the dermatologist if possible | |
| Order laboratory tests | Order liver function tests and complete blood count with differentials; assess for eosinophilia |
| Closely monitor patient | Stop anticonvulsant if history, physical findings suggest a drug-induced eruption |
| Refer patient to a dermatologist |
STEP 1: Take a thorough history
Ask the patient:
What medications are you taking? Because more than 100 medications could cause SJS or TEN, a detailed drug history is critical to determining whether a medication has induced the eruption.
When did you start taking the potentially offending medication? True lamotrigine-induced eruptions usually occur 5 days to 8 weeks after the first dose.10 SJS and TEN generally take 1 to 2 weeks to develop.
What is your current dosage? Has it increased or decreased recently? Rapid lamotrigine dosage escalations or use of lamotrigine with valproic acid can cause severe rash.9,10,18 Valproic acid increases serum lamotrigine by inhibiting its hepatic metabolism, thereby raising side-effect risk. In clinical trials, 30% of patients who received both anticonvulsants developed a rash.10
Have any family members had rashes after taking an anticonvulsant? Compared with the general population, siblings and first-degree relatives of patients with anticonvulsant-related eruptions are at higher risk for this complication.19 Decreased epoxide hydrolase activity might negate these patients’ ability to detoxify the arene oxide metabolite, which can cause adverse effects if it accumulates.
Do you have other medical problems? Hepatitis C, for example, can theoretically increase lamotrigine’s half-life, thereby elevating side-effect risk.11
Watch for anticonvulsant-related adverse events in patients with hepatic insufficiency because hepatitis might hinder anticonvulsant metabolism.20 Other medical comorbidities—such as HIV infection and systemic lupus erythematosus—also could increase the risk of antiepileptic-induced rash.10
Have you had fever, chills, or other symptoms? Patients with SJS and TEN usually present with systemic symptoms such as malaise, rash, lymphadenopathy, mucosal lesions, and/or symptoms such as photophobia, difficulty swallowing, rectal erosions, or dysuria. Patients with anticonvulsant hypersensitivity syndrome typically have fever and associated arthralgias, skin pain, lymphadenopathy, or a burning sensation on their skin. These symptoms generally are absent in localized cutaneous infections.
STEP 2: Examine the eruption
Cutaneous SJS and TEN findings usually include abrupt onset of erythematous macules—which progress to targetoid lesions containing central bullae—followed by extensive epidermal necrosis. Superficial lip and mouth necrosis occur early, leading to severe stomatitis.
TEN and SJS can appear similar clinically, but TEN
covers >30% of body surface area, whereas SJS covers <10%.
Rashes that cover 10% to 30% of body surface suggest SJS-TEN overlap syndrome.
Anticonvulsant hypersensitivity syndrome usually manifests as
a morbilliform eruption on the face, arms, and/or torso. The
lesions might become edematous and progress to exfoliation or vesiculobullae. Facial edema is a hallmark of anticonvulsant hypersensitivity,15,16 and pustules and/or erythroderma might also appear. Other warning signs include symmetrical widespread eruption and organomegaly.
STEP 3: Order laboratory tests
Check liver function and order a CBC with differential to measure eosinophils. Eosinophilia and abnormal LFT results can signal anticonvulsant hypersensitivity.
Eosinophils. A normal eosinophil count ranges between 0% and 5% of peripheral blood leukocytes in adults, at a count of 350 to 650/cm. Although upper limits of normal vary, values >500/cm suggest hypereosinophilia.21
LFTs. Normal aspartate aminotransferase and alanine aminotransferase levels are 0 to 42 U/L and 0 to 48 U/L, respectively. Any LFT elevation could signal anticonvulsant hypersensitivity syndrome.
STEP 4: Closely monitor the patient
Discontinue the anticonvulsant if findings suggest a cutaneous drug reaction, and contact the patient’s primary care physician or dermatologist immediately. Early consultation with a dermatologist can help determine the eruption’s cause and reveal therapeutic options.
Dr. Pejic is chief resident in the adult psychiatry residency program, Louisiana State University Health Sciences Center and Ochsner Clinic Foundation, New Orleans.
Dr. Klinger is a third-year dermatology resident, Dr. Conrad is assistant professor of clinical psychiatry, and Dr. Nesbitt is chairman, department of dermatology, Louisiana State University Health Sciences Center.
Related Resources
- High WA. Stevens-Johnson syndrome and toxic epidermal necrolysis in adults. UpToDate Online (version 15.1); June 9, 2007. www.uptodate.com.
- Martin KA, Krahn LE, Rosati MJ, Balan V. Hepatitis C: How to manage mood during interferon treatment. Current Psychiatry 2006;5(11):69-80. http://www.currentpsychiatry.com/article_pages.asp?AID=4553.
Drug Brand Names
- Bupropion • Wellbutrin
- Buspirone • BuSpar
- Carbamazepine • Tegretol, Equetro, others
- Ciprofloxacin • Cipro, Proquin
- Escitalopram • Lexapro
- Hydrochlorothiazide • various
- Lamotrigine • Lamictal
- Lithium • Eskalith, others
- Olanzapine • Zyprexa
- Peginterferon alfa-2B • PEG-Intron
- Phenytoin • Dilantin
- Ribavirin • Copegus, Rebetol, others
- Valproic acid • Depakote
Disclosure
Dr. Conrad receives research/grant support from AstraZeneca, Bristol-Myers Squibb, Forest Pharmaceuticals, GlaxoSmithKline, and Wyeth.
Drs. Pejic, Nesbitt, and Klinger report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
HISTORY: Depressed and sick
Mr. T, age 53, was diagnosed last year with hepatitis C and for 20 years has battled recurrent major depression with euthymia between episodes. His hepatologist asks us to evaluate his recent depressed mood and erratic behavior.
Less than 2 months ago, the hepatologist prescribed ribavirin, 1,000 mg bid, and peginterferon alfa-2B, 10 million IU/1.0 mL weekly, for hepatitis C. Soon afterward, Mr. T became irritable, especially toward his wife. He now refuses to leave his house most days because of overwhelming sadness and hopelessness. Once an avid motorcycle enthusiast, Mr. T has stopped riding and complains of fatigue, “fuzzy” thinking, and diminished concentration, but he denies suicidal thoughts or intent. He weighs 232 lb but has lost 15 lb in the last 6 weeks.
Five weeks ago, the hepatologist added bupropion XL, 150 mg/d, for Mr. T’s depressive symptoms, but the patient complained that the antidepressant “amped me up” and “made my mind race.” After 3 weeks, the hepatologist switched to escitalopram, 10 mg/d, but Mr. T’s agitation continued.
Several days after starting escitalopram, Mr. T experienced what he calls a “pink cloud” period—intensely pleasurable episodes that he says began in late childhood, usually last about 4 days, and occur 6 times annually. During these episodes, his thoughts race, his speech is mildly pressured, and he sleeps 5 hours or less nightly. While euphoric, he drives his motorcycle at 100 mph, starts several projects at once, and is distractible.
Once the “pink clouds” clear, Mr. T feels fatigued and “let down” as he does now. He says he has never reported these euphoric periods because he usually enjoys them.
Mr. T also has longstanding anxiety. Most days he is “on edge” and restless, feels muscle tension in his neck, and has trouble falling and staying sleep. After changing jobs last year, he began having panic attacks triggered by excessive worry. He denies anticipatory fear or avoidance, so we rule out panic disorder.
Additionally, Mr. T has been engaging in weekly binge-eating episodes during which he consumes nearly 50 large-sized cookies and 2 to 3 2-ounce bags of potato chips in 2 hours. He is wracked with guilt after bingeing and often feels embarrassed about being overweight (body mass index, 31 kg/m2). He does not purge but moderately restricts his diet between binges. He says he started bingeing at age 20, and at one point was bingeing 3 times a week.
Mr. T also complains that ribavirin and peginterferon are causing headaches, fatigue, and myalgias. He also takes hydrochlorothiazide, 25 mg/d, for hypertension, and is allergic to sulfonamides. He denies using alcohol and drugs but smokes 2 packs of cigarettes per day.
We diagnose bipolar II disorder based on Mr. T’s extreme mood shifts, history of major depressive episodes, recent hypomania, lack of manic or mixed episodes, and significant distress. His hypomania episodes last <1 week; episodes that last ≥1 week or require hospitalization would signal bipolar I disorder.
We rule out interferon-induced depression and hypomania1 because Mr. T showed signs of mood dysfunction long before he contracted hepatitis C. We also diagnose generalized anxiety disorder and eating disorder, not otherwise specified.
The authors’ observations
Diagnosing and managing bipolar disorder is challenging, especially when hypomania is not readily apparent.2
After we discuss treatment options with Mr. T, he chooses lamotrigine because it causes relatively few side effects and is less likely than valproic acid and other mood-stabilizing anticonvulsants to cause hepatotoxicity or pancreatitis.3 Lamotrigine also might reduce Mr. T’s anxiety.4
We do not try lithium because Mr. T is taking a diuretic (hydrochlorothiazide), which can cause lithium toxicity when used concomitantly. Also, lithium requires close laboratory monitoring, interacts with many medications, and can cause drowsiness, dry mouth, blurry vision, and fatigue.5 These
factors contraindicate lithium for Mr. T, who is taking several medications and suffers side effects from ribavirin and interferon.
Olanzapine might control Mr. T’s mood swings, but the neuroleptic can cause weight gain and metabolic syndrome6 and might complicate his eating disorder.
TREATMENT: A ‘rash’ reaction
We add lamotrigine, 25 mg/d, for 2 weeks and then increase to 50 mg/d.
Two days after the lamotrigine increase, Mr. T reports a rash on the left side of his trunk and left hip, buttock, and elbow (Figure). He also complains of mild chills and night sweats, although these symptoms emerged several weeks ago. He denies blistering, fevers, dysuria, nausea, or vomiting. We see no signs of lymphadenopathy, and mucosae are unaffected. Since he started lamotrigine, he says, he has not tried unfamiliar brands of shampoos, laundry detergents, or shower gels that might irritate his skin.
Figure: Did lamotrigine cause Mr. T’s rash?
Folliculocentric pustules around patient’s left elbow and throughout his left side.We have Mr. T come in that day for an emergency physical examination. At presentation, the rash appears infectious with isolated pustules throughout. We refer him to a dermatologist for same-day evaluation.
The authors’ observations
A rash is an immunologic reaction to an offending agent. If lamotrigine were causing the rash, lowering the dosage would not mitigate it.
We continued lamotrigine because the dermatologist could examine the rash within 24 hours of Mr. T’s complaint. Also, the agent was decreasing the patient’s mood, irritability, and anger. If we believed lamotrigine was causing the rash and could not obtain an immediate dermatology consult, we would have stopped the medication.
FOLLOW-UP: ‘Hot’ findings
During the patient history interview, the dermatologist discovers that Mr. T recently installed a whirlpool bath, and that the eruption occurred 3 to 5 days after the patient first used it. Physical examination shows groups of discrete folliculocentric pustules with surrounding erythema mainly on his extensor surfaces and left buttock. These findings and Mr. T’s history suggest a skin infection.
The dermatologist diagnoses hot tub folliculitis, an infection caused by exposure to contaminated whirlpools, hot tubs, or water slides. Cultures obtained that day grow Pseudomonas aeruginosa, confirming the diagnosis. The dermatologist tells Mr. T to stop using his whirlpool bath and prescribes topical gentamicin and ciprofloxacin, 500 mg bid for 10 days. We continue lamotrigine based on the dermatologist’s recommendation.
Two weeks later, Mr. T’s eruption resolves, and we increase lamotrigine to 100 mg/d, which improves his mood and achieves steady-state effectiveness.7 We continue escitalopram, 10 mg/d, then increase to 20 mg/d to treat his generalized anxiety. Mr. T begins experiencing anorgasmia 1 week after the escitalopram increase, so we switch to buspirone, 15 mg bid. After another 4 weeks, his anger, irritability, panic attacks, anxiety, and depression have diminished.
After 3 months, Mr. T’s hepatologist stops ribavirin and peginterferon because they are not helping his hepatitis C infection. Days later, Mr. T’s chills, sweats, and fatigue remit.
The hepatologist considers an experimental hepatitis C
medication.
We see Mr. T once monthly for supportive psychotherapy and medication management. Despite divorce proceedings and persistent mild depression he is optimistic, enjoys work, and rides his motorcycle safely twice a week.
The authors’ observations
Although Mr. T’s presentation and patient history clearly suggested an independent skin infection, distinguishing between an infection and anticonvulsant-induced rash can be difficult.
Lamotrigine and other antiepileptics (Table 1)8 have been associated with morbilliform eruptions, anticonvulsant hypersensitivity syndrome, erythema multiforme, Stevens-Johnson syndrome (SJS), and toxic epidermal necrolysis (TEN), a severe form of SJS with a 20% to 30% mortality rate.9,10
Table 1
Estimated risk of severe rash among first-time antiepileptic users*
| Drug | Total new users† | Total SJS/TEN cases | Risk per 10,000 new users |
|---|---|---|---|
| Carbamazepine | 286,360 | 39 | 1.4 |
| Lamotrigine | 55,154 | 14 | 2.5 |
| Phenobarbital | 8,659 | 7 | 8.1 |
| Phenytoin | 36,171 | 30 | 8.3 |
| Valproic acid | 103,150 | 4 | 0.4 |
| * Researchers reviewed records of patients hospitalized between 1998 and 2001 with SJS or TEN after using an anticonvulsant. | |||
| † Estimates based on number of dispensed prescriptions, average prescribed dosages, and duration of anticonvulsant use as recorded in Germany’s Mediplus database. | |||
| SJS: Stevens-Johnson syndrome; TEN: toxic epidermal necrolysis | |||
| Source: Adapted from reference 8 | |||
Although most lamotrigine-induced cutaneous eruptions are mild or self-limited, some are severe and potentially fatal. In clinical trials, approximately 10% of patients receiving lamotrigine for epilepsy developed cutaneous reactions.11 Among 3,348 patients with epilepsy who received lamotrigine, 11 (0.3%) required hospitalization for SJS or TEN.11
Anticonvulsant hypersensitivity syndrome, estimated to occur once per 1,000 to 10,000 exposures to anticonvulsants,12 can lead to fever, lymphadenopathy, hepatomegaly, and arthralgias. Although hypersensitivity to aromatic anticonvulsants such as phenytoin, carbamazepine, or phenobarbital is most common, hypersensitivity to lamotrigine also has been reported.13,14
Roughly 90% of patients with anticonvulsant hypersensitivity syndrome develop leukocytosis with eosinophilia, and some develop leukocytosis with agranulocytosis.15-17 Fulminant hepatitis can occur, which leads to most deaths associated with this syndrome.
4 steps to gauging rash
Taking a thorough history, examining the eruption, ordering liver function tests (LFTs) and a complete blood count (CBC), and referring the patient to a dermatologist are key to determining the seriousness of an eruption and planning treatment in patients taking anticonvulsants (Table 2). See the patient within 12 hours after he reports the rash, as SJS and TEN often progress rapidly.
Table 2
4 steps to determining rash severity and cause
| Take a thorough history | Find out when eruption occurred and when patient started the anticonvulsant |
| Ask about past rashes, other medicines, and family history of reactions to medications | |
| Find out if patient has had fever/chills, malaise, lymphadenopathy, or mucosal symptoms such as photophobia or dysuria | |
| Examine the eruption | Examine for mucosal involvement, facial edema, and blistering; describe the symmetry and extent of involvement |
| Look for systemic findings such as fever, chills, lymphadenopathy, and organomegaly | |
| Photograph the eruption for the dermatologist if possible | |
| Order laboratory tests | Order liver function tests and complete blood count with differentials; assess for eosinophilia |
| Closely monitor patient | Stop anticonvulsant if history, physical findings suggest a drug-induced eruption |
| Refer patient to a dermatologist |
STEP 1: Take a thorough history
Ask the patient:
What medications are you taking? Because more than 100 medications could cause SJS or TEN, a detailed drug history is critical to determining whether a medication has induced the eruption.
When did you start taking the potentially offending medication? True lamotrigine-induced eruptions usually occur 5 days to 8 weeks after the first dose.10 SJS and TEN generally take 1 to 2 weeks to develop.
What is your current dosage? Has it increased or decreased recently? Rapid lamotrigine dosage escalations or use of lamotrigine with valproic acid can cause severe rash.9,10,18 Valproic acid increases serum lamotrigine by inhibiting its hepatic metabolism, thereby raising side-effect risk. In clinical trials, 30% of patients who received both anticonvulsants developed a rash.10
Have any family members had rashes after taking an anticonvulsant? Compared with the general population, siblings and first-degree relatives of patients with anticonvulsant-related eruptions are at higher risk for this complication.19 Decreased epoxide hydrolase activity might negate these patients’ ability to detoxify the arene oxide metabolite, which can cause adverse effects if it accumulates.
Do you have other medical problems? Hepatitis C, for example, can theoretically increase lamotrigine’s half-life, thereby elevating side-effect risk.11
Watch for anticonvulsant-related adverse events in patients with hepatic insufficiency because hepatitis might hinder anticonvulsant metabolism.20 Other medical comorbidities—such as HIV infection and systemic lupus erythematosus—also could increase the risk of antiepileptic-induced rash.10
Have you had fever, chills, or other symptoms? Patients with SJS and TEN usually present with systemic symptoms such as malaise, rash, lymphadenopathy, mucosal lesions, and/or symptoms such as photophobia, difficulty swallowing, rectal erosions, or dysuria. Patients with anticonvulsant hypersensitivity syndrome typically have fever and associated arthralgias, skin pain, lymphadenopathy, or a burning sensation on their skin. These symptoms generally are absent in localized cutaneous infections.
STEP 2: Examine the eruption
Cutaneous SJS and TEN findings usually include abrupt onset of erythematous macules—which progress to targetoid lesions containing central bullae—followed by extensive epidermal necrosis. Superficial lip and mouth necrosis occur early, leading to severe stomatitis.
TEN and SJS can appear similar clinically, but TEN
covers >30% of body surface area, whereas SJS covers <10%.
Rashes that cover 10% to 30% of body surface suggest SJS-TEN overlap syndrome.
Anticonvulsant hypersensitivity syndrome usually manifests as
a morbilliform eruption on the face, arms, and/or torso. The
lesions might become edematous and progress to exfoliation or vesiculobullae. Facial edema is a hallmark of anticonvulsant hypersensitivity,15,16 and pustules and/or erythroderma might also appear. Other warning signs include symmetrical widespread eruption and organomegaly.
STEP 3: Order laboratory tests
Check liver function and order a CBC with differential to measure eosinophils. Eosinophilia and abnormal LFT results can signal anticonvulsant hypersensitivity.
Eosinophils. A normal eosinophil count ranges between 0% and 5% of peripheral blood leukocytes in adults, at a count of 350 to 650/cm. Although upper limits of normal vary, values >500/cm suggest hypereosinophilia.21
LFTs. Normal aspartate aminotransferase and alanine aminotransferase levels are 0 to 42 U/L and 0 to 48 U/L, respectively. Any LFT elevation could signal anticonvulsant hypersensitivity syndrome.
STEP 4: Closely monitor the patient
Discontinue the anticonvulsant if findings suggest a cutaneous drug reaction, and contact the patient’s primary care physician or dermatologist immediately. Early consultation with a dermatologist can help determine the eruption’s cause and reveal therapeutic options.
Dr. Pejic is chief resident in the adult psychiatry residency program, Louisiana State University Health Sciences Center and Ochsner Clinic Foundation, New Orleans.
Dr. Klinger is a third-year dermatology resident, Dr. Conrad is assistant professor of clinical psychiatry, and Dr. Nesbitt is chairman, department of dermatology, Louisiana State University Health Sciences Center.
Related Resources
- High WA. Stevens-Johnson syndrome and toxic epidermal necrolysis in adults. UpToDate Online (version 15.1); June 9, 2007. www.uptodate.com.
- Martin KA, Krahn LE, Rosati MJ, Balan V. Hepatitis C: How to manage mood during interferon treatment. Current Psychiatry 2006;5(11):69-80. http://www.currentpsychiatry.com/article_pages.asp?AID=4553.
Drug Brand Names
- Bupropion • Wellbutrin
- Buspirone • BuSpar
- Carbamazepine • Tegretol, Equetro, others
- Ciprofloxacin • Cipro, Proquin
- Escitalopram • Lexapro
- Hydrochlorothiazide • various
- Lamotrigine • Lamictal
- Lithium • Eskalith, others
- Olanzapine • Zyprexa
- Peginterferon alfa-2B • PEG-Intron
- Phenytoin • Dilantin
- Ribavirin • Copegus, Rebetol, others
- Valproic acid • Depakote
Disclosure
Dr. Conrad receives research/grant support from AstraZeneca, Bristol-Myers Squibb, Forest Pharmaceuticals, GlaxoSmithKline, and Wyeth.
Drs. Pejic, Nesbitt, and Klinger report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Crone CC, Gabriel GM, Wise TN. Managing the neuropsychiatric side effects of interferon-based therapy for hepatitis C. Cleve Clin J Med 2004;71(suppl 3):S27-S32.
2. Phelps JR, Ghaemi SN. Improving the diagnosis of bipolar disorder: predictive value of screening tests. J Affect Disord 2006;92:141-8.
3. Lacerda G, Krummel T, Sabourdy C, et al. Optimizing therapy of seizures in patients with renal or hepatic dysfunction. Neurology 2006;67(suppl 4):S28-S33.
4. Yumru M, Savas HA, Kurt E, et al. Atypical antipsychotics related metabolic syndrome in bipolar patients. J Affect Disord 2007;98:247-52.
5. Finley PR, Warner MD, Peabody CA. Clinical relevance of drug interactions with lithium. Clin Pharmacokinet 1995;3:172-91.
6. Keck PE, Jr, Strawn JR, McElroy SL. Pharmacologic treatment considerations in co-occurring bipolar and anxiety disorders. J Clin Psychiatry 2006;67(suppl 1):S8-S15.
7. Sachs GS, Printz DJ, Kahn DA, et al. The expert consensus guidelines: medication treatment of bipolar disorder 2000. A postgraduate medicine special report. New York: McGraw-Hill; 2000:24.
8. Mockenhaupt M, Messenheimer J, Tennis P, Schlingmann J. Risk of Stevens-Johnson syndrome and toxic epidermal necrolysis in new users of antiepileptics. Neurology 2005;64:1134-8.
9. Schlienger RG, Shapiro LE, Shear NH. Lamotrigine-induced severe cutaneous adverse reactions. Epilepsia 1998;29(suppl 7):S22-S26.
10. Calabrese JR, Sullivan JR, Bowden CL, et al. Rash in multicenter trials of lamotrigine in mood disorders: clinical relevance and management. J Clin Psychiatry 2002;63:1012-19.
11. Physicians’ desk reference. 61st ed. Montvale, NJ: Thomson PDR; 2007:1483-4, 1488.
12. Knowles SR, Shapiro LE, Shear NH. Anticonvulsant hypersensitivity syndrome: incidence, prevention and management. Drug Saf 1999;21:489-501.
13. Tennis P, Stern RS. Risk of serious cutaneous disorders after initiation of use of phenytoin, carbamazepine, or sodium valproate: a record linkage study. Neurology 1997;49:542-6.
14. Knowles SR, Shapiro LE, Shear NH. Anticonvulsant hypersensitivity syndrome: incidence, prevention and management. Drug Saf 1999;21:489-501.
15. Chang DK, Shear NH. Cutaneous reactions to anticonvulsants. Semin Neurol 1992;12:329-7.
16. Vittorio CC, Muglia JJ. Anticonvulsant hypersensitivity syndrome. Arch Intern Med 1995;155:2285-90.
17. Callot V, Roujeau JC, Bagot M, et al. Drug induced pseudo-lymphoma and hypersensitivity syndrome. Two different clinical entities. Arch Dermatol 1996;132:1315-21.
18. Yalcin B, Karaduman A. Stevens-Johnson syndrome associated with concomitant use of lamotrigine and valproic acid. J Am Acad Dermatol 2000;43:898-9.
19. Gennis MA, Vemuri R, Burns EA, et al. Familial occurrence of hypersensitivity to phenytoin. Am J Med 1991;91:631-4.
20. McLaren KD, Marangell LB. Special considerations in the treatment of patients with bipolar disorder and medical comorbidities. Ann Gen Hosp Psychiatry 2004;3:7.-
21. Valencak J, Ortiz-Urda S, Heere-Ress E, et al. Carbamazepine-induced DRESS syndrome with recurrent fever and exanthema. Int J Dermatol 2004;43:51-4.
1. Crone CC, Gabriel GM, Wise TN. Managing the neuropsychiatric side effects of interferon-based therapy for hepatitis C. Cleve Clin J Med 2004;71(suppl 3):S27-S32.
2. Phelps JR, Ghaemi SN. Improving the diagnosis of bipolar disorder: predictive value of screening tests. J Affect Disord 2006;92:141-8.
3. Lacerda G, Krummel T, Sabourdy C, et al. Optimizing therapy of seizures in patients with renal or hepatic dysfunction. Neurology 2006;67(suppl 4):S28-S33.
4. Yumru M, Savas HA, Kurt E, et al. Atypical antipsychotics related metabolic syndrome in bipolar patients. J Affect Disord 2007;98:247-52.
5. Finley PR, Warner MD, Peabody CA. Clinical relevance of drug interactions with lithium. Clin Pharmacokinet 1995;3:172-91.
6. Keck PE, Jr, Strawn JR, McElroy SL. Pharmacologic treatment considerations in co-occurring bipolar and anxiety disorders. J Clin Psychiatry 2006;67(suppl 1):S8-S15.
7. Sachs GS, Printz DJ, Kahn DA, et al. The expert consensus guidelines: medication treatment of bipolar disorder 2000. A postgraduate medicine special report. New York: McGraw-Hill; 2000:24.
8. Mockenhaupt M, Messenheimer J, Tennis P, Schlingmann J. Risk of Stevens-Johnson syndrome and toxic epidermal necrolysis in new users of antiepileptics. Neurology 2005;64:1134-8.
9. Schlienger RG, Shapiro LE, Shear NH. Lamotrigine-induced severe cutaneous adverse reactions. Epilepsia 1998;29(suppl 7):S22-S26.
10. Calabrese JR, Sullivan JR, Bowden CL, et al. Rash in multicenter trials of lamotrigine in mood disorders: clinical relevance and management. J Clin Psychiatry 2002;63:1012-19.
11. Physicians’ desk reference. 61st ed. Montvale, NJ: Thomson PDR; 2007:1483-4, 1488.
12. Knowles SR, Shapiro LE, Shear NH. Anticonvulsant hypersensitivity syndrome: incidence, prevention and management. Drug Saf 1999;21:489-501.
13. Tennis P, Stern RS. Risk of serious cutaneous disorders after initiation of use of phenytoin, carbamazepine, or sodium valproate: a record linkage study. Neurology 1997;49:542-6.
14. Knowles SR, Shapiro LE, Shear NH. Anticonvulsant hypersensitivity syndrome: incidence, prevention and management. Drug Saf 1999;21:489-501.
15. Chang DK, Shear NH. Cutaneous reactions to anticonvulsants. Semin Neurol 1992;12:329-7.
16. Vittorio CC, Muglia JJ. Anticonvulsant hypersensitivity syndrome. Arch Intern Med 1995;155:2285-90.
17. Callot V, Roujeau JC, Bagot M, et al. Drug induced pseudo-lymphoma and hypersensitivity syndrome. Two different clinical entities. Arch Dermatol 1996;132:1315-21.
18. Yalcin B, Karaduman A. Stevens-Johnson syndrome associated with concomitant use of lamotrigine and valproic acid. J Am Acad Dermatol 2000;43:898-9.
19. Gennis MA, Vemuri R, Burns EA, et al. Familial occurrence of hypersensitivity to phenytoin. Am J Med 1991;91:631-4.
20. McLaren KD, Marangell LB. Special considerations in the treatment of patients with bipolar disorder and medical comorbidities. Ann Gen Hosp Psychiatry 2004;3:7.-
21. Valencak J, Ortiz-Urda S, Heere-Ress E, et al. Carbamazepine-induced DRESS syndrome with recurrent fever and exanthema. Int J Dermatol 2004;43:51-4.
Modafinil: Not just for sleep disorders?
Ms. B, a middle-aged mother of 3, is being monitored for bipolar disorder. She has a history of stimulant abuse but has been in remission for 5 years. She complains of excessive daytime sleepiness. Most days she wakes at 7 AM, but sleeps on several occasions during the day. She also complains of fatigue and lack of motivation.
She is being treated with lithium, venlafaxine, and zolpidem and reports good adherence. Basic laboratory work and serum lithium levels are within acceptable ranges. Her symptoms do not improve when venlafaxine is titrated from 225 mg/d to 300 mg/d. She also reports previously failed trials with bupropion and fluoxetine.
We decide to try a psychostimulant as an augmenting agent. Because of her past stimulant abuse, we add modafinil, 100 mg/d and increase to 200 mg/d. Ms. B reports improvement in her daytime sleepiness and fatigue and—except for a mild headache—tolerates the medication well.
Modafinil is being investigated for potential roles in managing inattention, excess sleepiness, fatigue, and cognitive dysfunction associated with:
- mood disorders (major depression and bipolar depression)
- attention-deficit/hyperactivity disorder (ADHD)
- schizophrenia
- cocaine dependence.
This article discusses how the drug promotes wakefulness, how it might improve cognitive function, and what the evidence reveals about off-label indications.
How it works
Although modafinil’s precise mechanism of action is unknown, it is believed to promote wakefulness more selectively than conventional stimulants such as amphetamine and methylphenidate. Modafinil does not bind to norepinephrine, serotonin, dopamine, or benzodiazepine receptors.1,2 It might target specific hypothalamic regions such as the tuberomammillary nucleus and orexin neurons, which are peptide neurotransmitters that promote wakefulness.3,4
Clinical trials found that modafinil has beneficial effects on:
- working memory, recognition memory, and sustained attention in healthy humans
- prefrontal-dependent cognitive functions in schizophrenia, major depression, and adult ADHD.5
Evidence for approved indications
Modafinil is indicated to improve wakefulness in patients who have excessive sleepiness associated with narcolepsy, obstructive sleep apnea, or shift work sleep disorder. It was approved for reducing excessive sleepiness in narcoleptic patients after two 9-week placebo-controlled clinical trials. The drug significantly reduced sleepiness and improved overall disease status as measured by the Clinical Global Impression of Change (CGI-C) scale.6,7
In patients with shift work sleep disorder, a 12-week placebo-controlled clinical trial found that modafinil significantly improved sleep latency and CGI-C scores.10
Dosage and side effects. For patients with narcolepsy or obstructive sleep apnea, the recommended dose is 200 mg given in the morning.11 For patients prescribed modafinil for work-time wakefulness, the dose is 200 mg 1 hour before their work shift. Lower doses are recommended for patients who are elderly or have hepatic impairment. Those with severe hepatic impairment typically are prescribed 100 mg/d.11 Modafinil is rapidly absorbed and is metabolized primarily by the liver (Table 1). A summary of potential drug-drug interactions appears in Table 2.11
In pivotal trials, adverse events that occurred more frequently with modafinil than with placebo and in >5% of the study population included headache, nausea, nervousness, rhinitis, diarrhea, back pain, insomnia, dizziness, and dyspepsia. Headache was most commonly reported; in most patients, it resolved soon after they started taking modafinil. Post-marketing reports have included cases of psychosis, mania, and suspected serious skin reactions, including Stevens-Johnson syndrome.11 Modafinil lacks euphorigenic properties and has minimal potential for abuse.12
Table 1
Modafinil’s pharmacokinetics
| Absorbed rapidly, with peak plasma concentrations at 2 to 4 hours |
| Apparent steady states reached after 2 to 4 days of dosing |
| Half-life: 15 hours |
| Major route of elimination (~90%) is metabolism, primarily by the liver |
Selected drug-drug interactions with modafinil
| Action of modafinil | Potential drug interactions |
|---|---|
| Increases elimination of CYP 3A4 substrates | Carbamazepine, phenytoin may decrease modafinil levels Azole antifungals, protease inhibitors, and erythromycin may increase modafinil levels |
| Inhibits CYP 2C19 enzyme | Modafinil may increase levels of citalopram, diazepam, and sertraline |
| Decreases absorption of ethinyl estradiol | Modafinil can decrease effectiveness of oral contraceptives |
| CYP: cytochrome P-450 | |
| Source: Reference 11 | |
Evidence for off-label uses
Major depressive disorder (MDD). The fatigue and excessive sleepiness often seen with MDD often persist after other depressive symptoms have remitted with antidepressant treatment.13 Patients with these symptoms might benefit from modafinil’s stimulating properties. Conventional stimulants such as methylphenidate have been used to improve neurovegetative symptoms of depression, but modafinil offers several advantages:
- decreased adverse CNS effects
- fewer drug-drug interactions
- minimal risk for dependence or abuse.
A 6-week open-label study of 25 depressed patients with residual fatigue and sleepiness showed that adjunctive modafinil, 100 to 200 mg/d, significantly improved these symptoms, as well as Hamilton Rating Scale for Depression (HAM-D) score, as early as week 2. Seventy-six percent of patients responded to treatment, defined as a >50% reduction in HAM-D scores.16
Several open-label studies and case re-ports have evaluated adjunctive modafinil use in patients with:
- depression characterized by ongoing lethargy or apathy17
- depression with atypical features18
- seasonal affective disorder19
- partial response to antidepressants.20,21
Bipolar depression. A 6-week, double-blind, placebo-controlled trial randomly assigned 85 patients with bipolar depression to adjunctive modafinil, 100 to 200 mg/d, or placebo for 6 weeks (Table 3).22 The number of patients receiving an antidepressant or mood stabilizer was not significantly different between the modafinil and placebo groups.
The primary outcome measure was change in the Inventory for Depressive Symptoms (IDS) score from baseline to endpoint. Forty-four percent of patients receiving modafinil achieved a ≥50% reduction in IDS score, compared with 23% of the placebo group; this difference was statistically significant (P=0.03).
In this study, modafinil was well tolerated and did not induce mania or hypomania. Cases of modafinil-induced mania have been reported elsewhere.23,24
The mechanisms of modafinil’s antidepressant effects are unclear. The drug does not cause release of norepinephrine or dopamine. One study proposed that modafinil acts by releasing histamine and activating noradrenaline receptors.25 Activation of these receptors increases dopamine and norepinephrine in these areas, and excites histaminergic tuberomammillary neurons, increasing histamine levels. Another trial suggested that modafinil may improve mood by mechanisms similar to the antidepressant effects induced by sleep deprivation.26
Summary. Modafinil may have a role in managing residual fatigue and excessive sleepiness associated with MDD and bipolar depression. Evidence for a mood-elevating effect is minimal; additional studies are needed. Adjunctive modafinil and conventional stimulants have not been compared head-to-head in patients with mood disorders. Modafinil’s tolerability profile and lack of euphorigenic and reinforcing properties make it a potentially attractive alternative, however.
ADHD. Approximately 30% of ADHD patients do not respond to or are unable to tolerate conventional stimulant medications such as methylphenidate and dextroamphetamine.27 Several studies have evaluated modafinil as a potential treatment for ADHD based on the drug’s action on arousal and attention systems. Although modafinil’s precise mechanism of action in ADHD is unknown, proposed mechanisms include:
- hypothalamic and cerebral cortex neuronal activation
- action on histamine that results in internal vigilance.28
Can modafinil help patients with mood disorders?
| Author | Study design | Modafinil dose | Conclusion |
|---|---|---|---|
| Major depressive disorder | |||
| Fava et al, 200514 | 8-week, double-blind, placebo-controlled; 331 subjects with partial or no response to SSRI monotherapy | 200 mg/d | No significant difference between modafinil and placebo at final visit |
| DeBattista et al, 200315 | 6-week, double-blind, placebo-controlled; 136 subjects with partial response to antidepressant therapy | 100 to 400 mg/d | Significant improvement in sleepiness by week 1 and fatigue by week 2, but differences between modafinil and placebo were not statistically significant by end of study |
| Konuk et al, 200616 | 6-week, open-label; 25 subjects with residual sleepiness or fatigue after SSRI therapy | 100 to 200 mg/d | All patients showed significant improvement in sleepiness, fatigue, and HAM-D scores |
| Bipolar depression | |||
| Frye et al, 200722 | 6-week, double-blind, placebo-controlled trial; 85 subjects who did not respond to a mood stabilizer with or without concomitant antidepressant therapy | 100 to 200 mg/d (mean 177 mg/d) | 44% of modafinil patients achieved ≥50% reduction in IDS score compared with 23% in placebo group (P=0.03) |
| HAM-D: Hamilton Rating Scale for Depression; IDS: Inventory for Depressive Symptoms; SSRI: selective serotonin reuptake inhibitor | |||
CASE 2: Another Tx for ADHD
Matt, age 8, is referred to our outpatient child psychiatric clinic after his parents noted declining school performance associated with increased aggression and irritability. Our assessment strongly supports a diagnosis of ADHD without comorbid conditions. We start Matt on methylphenidate, 5 mg twice daily, which quickly improves his ADHD symptoms. However, the medication causes GI side effects and profound sleep and weight changes.
Matt’s parents request that their son be treated with a different type of agent. A trial of atomoxetine is not as effective as the initial methylphenidate dosage and produces similar side effects. We then consider modafinil because of its side effect profile. We start Matt on 100 mg once daily and titrate up to 200 mg/d 4 weeks later. Matt and his parents notice an immediate improvement in his ADHD symptoms with no side effects.
In children and adolescents. Wigal et al29 reviewed pooled data from 3 randomized, double-blind, placebo-controlled studies of modafinil in pediatric ADHD (Table 4). Modafinil was well tolerated and improved ADHD symptoms and behaviors regardless of patients’ stimulant use history.
In a recent open-label study, 220 children and young adolescents with ADHD who had completed 4 weeks of a double-blind, placebo-controlled trial were evaluated for an additional 8 weeks. Modafinil improved ADHD symptoms and overall clinical condition as determined by the parent- or clinician-completed ADHD Rating Scale-IV Home Version, the parent-completed Conners’ ADHD/DSM-IV Scale Parent Version, and the clinician-rated CGI scale.30 Insomnia, headache, and decreased appetite were the most commonly reported adverse events.
In adults. The results of 2 double-blind, placebo-controlled trials of modafinil in adults with ADHD have been positive:
- In 1 study, modafinil (mean 206.8 mg/d) was more effective than placebo and comparable to dextroamphetamine in improving ADHD symptoms.31
- In another, modafinil (a single 200-mg dose) increased cognitive performance during treatment.32
Schizophrenia. Double-blind, randomized placebo-controlled studies have evaluated modafinil for improving cognitive function and reducing negative symptoms in patients with schizophrenia. Results have been inconsistent.
One double-blind, randomized, placebo-controlled crossover study of 20 patients with chronic schizophrenia found that modafinil, 200 mg/d, significantly improved short-term verbal memory span and attentional set shifting—the ability to discriminate and selectively attend to various stimulus dimensions (Table 5).33 Two other controlled studies showed no differences between the effects of modafinil and placebo on schizophrenia’s fatigue, cognition, or positive or negative symptoms.34,35
Summary. Although open-label studies have shown modafinil has beneficial effects on cognitive symptoms, controlled data are scarce. Reports of modafinil-induced psychosis or mania11 may limit the drug’s usefulness in schizophrenia patients.
Cocaine dependence. No medications are FDA-approved for treating cocaine dependence. A placebo-controlled, double-blind trial found that modafinil blunts cocaine euphoria under controlled conditions.36 This effect is hypothesized to be secondary to modafinil’s glutamate-enhancing and gamma-aminobutyric acid inhibitory effects.37
Summary. A single study supports using modafinil to improve outcomes in cocaine-dependent patients receiving standardized psychosocial treatment. More research is needed.
Table 4
Modafinil and ADHD: What the evidence says
| Author | Study design | Modafinil dosage | Conclusion |
|---|---|---|---|
| Wigal et al, 200629 | Analysis of data from 3 double-blind, placebo-controlled trials; total 638 children/adolescents, some of whom had received prior stimulant therapy | 170 to 425 mg/d | Whether or not patients received prior stimulant treatment, modafinil significantly improved ADHD symptoms and was well tolerated |
| Boellner et al, 200630 | 8-week, open-label extension of a 4-week double-blind, placebo-controlled trial; 220 subjects ages 6-14 | 100 to 400 mg/d | Modafinil improved ADHD symptoms and overall clinical condition |
| Taylor et al, 200031 | 2-week, double-blind, placebo-controlled crossover comparing modafinil with dextroamphetamine; 22 adults | Mean 206.8 mg/d | Both modafinil and dextroamphetamine significantly improved ADHD symptoms compared with placebo |
| Turner et al, 200432 | Double-blind, placebo-controlled crossover; 20 adults | Single 200-mg dose | Modafinil improved results on cognitive tests, including short-term memory span, visual memory, spatial planning, and sustained attention |
| ADHD: attention-deficit/hyperactivity disorder | |||
Table 5
Modafinil for schizophrenia or cocaine dependence:
More research is needed
| Author | Study design | Modafinil dosage | Conclusion |
|---|---|---|---|
| Schizophrenia | |||
| Turner et al, 200433 | Double-blind, placebo-controlled crossover; 20 adults | 200 mg/d | Modafinil significantly improved attentional set shifting and short-term verbal memory span |
| Sevy et al, 200534 | 8-week, double-blind, placebo-controlled; 24 subjects | Up to 200 mg/d | No significant difference between modafinil and placebo in reducing fatigue or positive or negative symptoms or in improving cognition |
| Pierre et al, 200735 | 8-week, double-blind, placebo-controlled; 20 subjects | 100 to 200 mg/d | Modafinil did not significantly improve neurocognitive or negative symptoms |
| Cocaine dependence | |||
| Dackis et al, 200538 | 8-week, double-blind, placebo-controlled; 62 cocaine-dependent subjects | 400 mg/d | Patients receiving modafinil provided significantly more cocaine-negative urine samples and were significantly more likely to achieve =3 weeks cocaine abstinence than those receiving placebo |
Related resource
- Ballon JS, Feifel D. A systematic review of modafinil: potential clinical uses and mechanisms of action. J Clin Psychiatry 2006;67(4):554-66.
- Atomoxetine • Strattera
- Bupropion • Wellbutrin
- Carbamazepine • Carbatrol, Tegretol, others
- Citalopram • Celexa
- Dextroamphetamine • Dexedrine, DextroStat
- Diazepam • Valium
- Erythromycin • Ery-Tab, Eryc, others
- Fluoxetine • Prozac
- Lithium • Eskalith, Lithobid
- Methylphenidate • Ritalin, others
- Modafinil • Provigil
- Phenytoin • Dilantin
- Sertraline • Zoloft
- Venlafaxine • Effexor
- Zolpidem • Ambien
Dr. Ramaswamy receives research support from Bristol-Myers Squibb, Shire, and Forest Pharmaceuticals and is a consultant to Dainippon Sumitomo Pharma.
Dr. Mattai reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
Dr. Wilson receives research support from, is a consultant to, or is a speaker for the National Institute of Mental Health, the Substance Abuse and Mental Health Services Administration, the Veterans Administration, the State of Nebraska, the State of Ohio, Health Futures Foundation, Inc., Abbott Laboratories, Astra-Zeneca, Bristol-Myers Squibb, Elan, Eli Lilly and Company, GlaxoSmithKline, Janssen, Ortho-McNeil, Pfizer, and Wyeth.
1. Mignot E, Nishino S, Guilleminault C, et al. Modafinil binds to dopamine uptake carrier site with low affinity. Sleep 1994;17:436-7.
2. Lin J-S, Hou Y, Jouvet M. Potential brain neuronal targets for amphetamine, methylphenidate, and modafinil induced wakefulness, evidenced by c-fos immunocytochemistry in the cat. Proc Natl Acad Sci USA 1996;93:14128-33.
3. Scammell TE, Estabrooke IV, McCarthy MT, et al. Hypothalamic arousal regions are activated during modafinil-induced wakefulness. J Neurosci 2000;20(22):8620-8.
4. Stenberg D. Neuroanatomy and neurochemistry of sleep. Cell Mol Life Sci 2007;64(10):1187-204.
5. Minzenberg MJ, Carter CS. Modafinil: a review of neurochemical actions and effects on cognition. Neuropsychopharmacology In press.
6. U.S. Modafinil in Narcolepsy Multicenter Study Group. Randomized trial of modafinil for the treatment of pathological somnolence in narcolepsy. Ann Neurol 1998;43(1):88-97.
7. U.S. Modafinil in Narcolepsy Multicenter Study Group. Randomized trial of modafinil as a treatment for the excessive daytime somnolence of narcolepsy. Neurology 2000;54:1166-75.
8. Black JE, Hirshkowitz M. Modafinil for treatment of residual excessive sleepiness in nasal continuous positive airway pressure-treated obstructive sleep apnea/hypopnea syndrome. Sleep 2005;28(4):464-71.
9. Pack AI, Black JE, Schwartz JR, Matheson JK. Modafinil as adjunct therapy for daytime sleepiness in obstructive sleep apnea. Am J Respir Crit Care Med 2001;164(9):1675-81.
10. Czeisler CA, Walsh JK, Roth T, et al. and the U.S. Modafinil in Shift Work Sleep Disorder Study Group. Modafinil for excessive sleepiness associated with shift-work sleep disorder. N Engl J Med. 2005;353(5):476-86. Published correction appears in: N Engl J Med 2005;353(10):1078.-
11. Provigil [package insert] West Chester, PA: Cephalon Inc; 2004.
12. Myrick H, Malcolm R, Taylor B, et al. Modafinil: preclinical, clinical, and post-marketing surveillance—a review of abuse liability issues. Ann Clin Psychiatry 2004;16(2):101-9.
13. Baldwin DS, Papakostas GI. Symptoms of fatigue and sleepiness in major depressive disorder. J Clin Psychiatry 2006;67(suppl 6):9-15.
14. Fava M, Thase ME, DeBattista C. A multicenter, placebo-controlled study of modafinil augmentation in partial responders to selective serotonin reuptake inhibitors with persistent fatigue and sleepiness. J Clin Psychiatry 2005;66(1):85-93.
15. DeBattista C, Doghramji K, Menza MA, et al. and the Modafinil in Depression Study Group. Adjunct modafinil for the short-term treatment of fatigue and sleepiness in patients with major depressive disorder: a preliminary double-blind, placebo-controlled study. J Clin Psychiatry 2003;64(9):1057-64.
16. Konuk N, Atasoy N, Atik L, Akay O. Open-label study of adjunct modafinil for the treatment of patients with fatigue, sleepiness, and major depression treated with selective serotonin reuptake inhibitors. Adv Ther 2006;23(4):646-54.
17. Markovitz PJ, Wagner S. An open-label trial of modafinil augmentation in patients with partial response to antidepressant therapy. J Clin Psychopharmacol 2003;23(2):207-9.
18. Vaishnavi S, Gadde K, Alamy S, et al. Modafinil for atypical depression: effects of open-label and double-blind discontinuation treatment. J Clin Psychopharmacol 2006; 26(4): 373-8. Published correction appears in: J Clin Psychopharmacol. 2006;26(5):523.
19. Lundt L. Modafinil treatment in patients with seasonal affective disorder/winter depression: an open-label pilot study. J Affect Disord 2004;81(2):173-8.
20. DeBattista C, Lembke A, Solvason HB, et al. A prospective trial of modafinil as an adjunctive treatment of major depression. J Clin Psychopharmacol 2004;24(1):87-90.
21. Rasmussen NA, Schroder P, Olsen LR, et al. Modafinil augmentation in depressed patients with partial response to antidepressants: a pilot study on self-reported symptoms covered by the Major Depression Inventory (MDI) and the Symptom Checklist (SCL-92). Nord J Psychiatry 2005;59(3):173-8.
22. Frye MA, Grunze H, Suppes T, et al. A placebo-controlled evaluation of adjunctive modafinil in the treatment of bipolar depression. Am J Psychiatry 2007;164:1242-9.
23. Wolf J, Fiedler U, Anghelescu I, Schwertfeger N. Manic switch in a patient with treatment-resistant bipolar depression treated with modafinil. J Clin Psychiatry 2006;67(11):1817.-
24. Vorspan F, Warot D, Consoli A, et al. Mania in a boy treated with modafinil for narcolepsy. Am J Psychiatry 2005;162(4):813-4.
25. McClellan KJ, Spencer CM. Modafinil: a review of its pharmacology and clinical efficacy in the management of narcolepsy. CNS Drugs 1998;9(4):311-24.
26. Ballon JS, Feifel D. A systematic review of modafinil: potential clinical uses and mechanisms of action. J Clin Psychiatry 2006;67(4):554-66.
27. Dulcan M. Practice parameters for the assessment and treatment of children, adolescents and adults with attentiondeficit hyperactivity disorder. American Academy of Child and Adolescent Psychiatry. J Am Acad Child Adolesc Psychiatry 1997;36(suppl 10):85S-121S.
28. Lindsay SE, Gudelsky GA, Heaton PC. Use of modafinil for the treatment of attention deficit/hyperactivity disorder. Ann Pharmacother 2006;40(10):1829-33.
29. Wigal SB, Biederman J, Swanson JM, et al. Efficacy and safety of modafinil film-coated tablets in children and adolescents with or without prior stimulant treatment for attention-deficit/hyperactivity disorder: pooled analysis of 3 randomized, double-blind, placebo-controlled studies. Prim Care Companion J Clin Psychiatry 2006;8(6):352-60.
30. Boellner SW, Earl CQ, Arora S. Modafinil in children and adolescents with attention-deficit/hyperactivity disorder: a preliminary 8-week, open-label study. Curr Med Res Opin 2006;22(12):2457-65.
31. Taylor FB, Russo J. Efficacy of modafinil compared to dextroamphetamine for the treatment of attention deficit hyperactivity disorder in adults. J Child Adolesc Psychopharmacol 2000;10(4):311-20.
32. Turner DC, Clark L, Dowson J, et al. Modafinil improves cognition and response inhibition in adult attention-deficit/ hyperactivity disorder. Biol Psychiatry 2004;55(10):1031-40.
33. Turner DC, Clark L, Pomarol-Clotet E, et al. Modafinil improves cognition and attentional set shifting in patients with chronic schizophrenia. Neuropsychopharmacology 2004;29(7):1363-73.
34. Sevy S, Rosenthal MH, Alvir J, et al. Double-blind, placebo-controlled study of modafinil for fatigue and cognition in schizophrenia patients treated with psychotropic medications. J Clin Psychiatry 2005;66(7):839-43.
35. Pierre JM, Peloian JH, Wirshing DA, et al. A randomized, double-blind, placebo-controlled trial of modafinil for negative symptoms in schizophrenia. J Clin Psychiatry 2007;68(5):705-10.
36. Dackis CA, Lynch KG, Yu E, et al. Modafinil and cocaine: a double-blind, placebo-controlled drug interaction study. Drug Alcohol Depend 2003;70(1):29-37.
37. Perez de la Mora M, Aguilar-Garcia A, Ramon-Frias T, et al. Effects of the vigilance promoting drug modafinil on the synthesis of GABA and glutamate in slices of rat hypothalamus. Neurosci Lett 1999;259:181-5.
38. Dackis CA, Kampman KM, Lynch KG, et al. A double-blind, placebo-controlled trial of modafinil for cocaine dependence. Neuropsychopharmacology 2005;30(1):205-11.
Ms. B, a middle-aged mother of 3, is being monitored for bipolar disorder. She has a history of stimulant abuse but has been in remission for 5 years. She complains of excessive daytime sleepiness. Most days she wakes at 7 AM, but sleeps on several occasions during the day. She also complains of fatigue and lack of motivation.
She is being treated with lithium, venlafaxine, and zolpidem and reports good adherence. Basic laboratory work and serum lithium levels are within acceptable ranges. Her symptoms do not improve when venlafaxine is titrated from 225 mg/d to 300 mg/d. She also reports previously failed trials with bupropion and fluoxetine.
We decide to try a psychostimulant as an augmenting agent. Because of her past stimulant abuse, we add modafinil, 100 mg/d and increase to 200 mg/d. Ms. B reports improvement in her daytime sleepiness and fatigue and—except for a mild headache—tolerates the medication well.
Modafinil is being investigated for potential roles in managing inattention, excess sleepiness, fatigue, and cognitive dysfunction associated with:
- mood disorders (major depression and bipolar depression)
- attention-deficit/hyperactivity disorder (ADHD)
- schizophrenia
- cocaine dependence.
This article discusses how the drug promotes wakefulness, how it might improve cognitive function, and what the evidence reveals about off-label indications.
How it works
Although modafinil’s precise mechanism of action is unknown, it is believed to promote wakefulness more selectively than conventional stimulants such as amphetamine and methylphenidate. Modafinil does not bind to norepinephrine, serotonin, dopamine, or benzodiazepine receptors.1,2 It might target specific hypothalamic regions such as the tuberomammillary nucleus and orexin neurons, which are peptide neurotransmitters that promote wakefulness.3,4
Clinical trials found that modafinil has beneficial effects on:
- working memory, recognition memory, and sustained attention in healthy humans
- prefrontal-dependent cognitive functions in schizophrenia, major depression, and adult ADHD.5
Evidence for approved indications
Modafinil is indicated to improve wakefulness in patients who have excessive sleepiness associated with narcolepsy, obstructive sleep apnea, or shift work sleep disorder. It was approved for reducing excessive sleepiness in narcoleptic patients after two 9-week placebo-controlled clinical trials. The drug significantly reduced sleepiness and improved overall disease status as measured by the Clinical Global Impression of Change (CGI-C) scale.6,7
In patients with shift work sleep disorder, a 12-week placebo-controlled clinical trial found that modafinil significantly improved sleep latency and CGI-C scores.10
Dosage and side effects. For patients with narcolepsy or obstructive sleep apnea, the recommended dose is 200 mg given in the morning.11 For patients prescribed modafinil for work-time wakefulness, the dose is 200 mg 1 hour before their work shift. Lower doses are recommended for patients who are elderly or have hepatic impairment. Those with severe hepatic impairment typically are prescribed 100 mg/d.11 Modafinil is rapidly absorbed and is metabolized primarily by the liver (Table 1). A summary of potential drug-drug interactions appears in Table 2.11
In pivotal trials, adverse events that occurred more frequently with modafinil than with placebo and in >5% of the study population included headache, nausea, nervousness, rhinitis, diarrhea, back pain, insomnia, dizziness, and dyspepsia. Headache was most commonly reported; in most patients, it resolved soon after they started taking modafinil. Post-marketing reports have included cases of psychosis, mania, and suspected serious skin reactions, including Stevens-Johnson syndrome.11 Modafinil lacks euphorigenic properties and has minimal potential for abuse.12
Table 1
Modafinil’s pharmacokinetics
| Absorbed rapidly, with peak plasma concentrations at 2 to 4 hours |
| Apparent steady states reached after 2 to 4 days of dosing |
| Half-life: 15 hours |
| Major route of elimination (~90%) is metabolism, primarily by the liver |
Selected drug-drug interactions with modafinil
| Action of modafinil | Potential drug interactions |
|---|---|
| Increases elimination of CYP 3A4 substrates | Carbamazepine, phenytoin may decrease modafinil levels Azole antifungals, protease inhibitors, and erythromycin may increase modafinil levels |
| Inhibits CYP 2C19 enzyme | Modafinil may increase levels of citalopram, diazepam, and sertraline |
| Decreases absorption of ethinyl estradiol | Modafinil can decrease effectiveness of oral contraceptives |
| CYP: cytochrome P-450 | |
| Source: Reference 11 | |
Evidence for off-label uses
Major depressive disorder (MDD). The fatigue and excessive sleepiness often seen with MDD often persist after other depressive symptoms have remitted with antidepressant treatment.13 Patients with these symptoms might benefit from modafinil’s stimulating properties. Conventional stimulants such as methylphenidate have been used to improve neurovegetative symptoms of depression, but modafinil offers several advantages:
- decreased adverse CNS effects
- fewer drug-drug interactions
- minimal risk for dependence or abuse.
A 6-week open-label study of 25 depressed patients with residual fatigue and sleepiness showed that adjunctive modafinil, 100 to 200 mg/d, significantly improved these symptoms, as well as Hamilton Rating Scale for Depression (HAM-D) score, as early as week 2. Seventy-six percent of patients responded to treatment, defined as a >50% reduction in HAM-D scores.16
Several open-label studies and case re-ports have evaluated adjunctive modafinil use in patients with:
- depression characterized by ongoing lethargy or apathy17
- depression with atypical features18
- seasonal affective disorder19
- partial response to antidepressants.20,21
Bipolar depression. A 6-week, double-blind, placebo-controlled trial randomly assigned 85 patients with bipolar depression to adjunctive modafinil, 100 to 200 mg/d, or placebo for 6 weeks (Table 3).22 The number of patients receiving an antidepressant or mood stabilizer was not significantly different between the modafinil and placebo groups.
The primary outcome measure was change in the Inventory for Depressive Symptoms (IDS) score from baseline to endpoint. Forty-four percent of patients receiving modafinil achieved a ≥50% reduction in IDS score, compared with 23% of the placebo group; this difference was statistically significant (P=0.03).
In this study, modafinil was well tolerated and did not induce mania or hypomania. Cases of modafinil-induced mania have been reported elsewhere.23,24
The mechanisms of modafinil’s antidepressant effects are unclear. The drug does not cause release of norepinephrine or dopamine. One study proposed that modafinil acts by releasing histamine and activating noradrenaline receptors.25 Activation of these receptors increases dopamine and norepinephrine in these areas, and excites histaminergic tuberomammillary neurons, increasing histamine levels. Another trial suggested that modafinil may improve mood by mechanisms similar to the antidepressant effects induced by sleep deprivation.26
Summary. Modafinil may have a role in managing residual fatigue and excessive sleepiness associated with MDD and bipolar depression. Evidence for a mood-elevating effect is minimal; additional studies are needed. Adjunctive modafinil and conventional stimulants have not been compared head-to-head in patients with mood disorders. Modafinil’s tolerability profile and lack of euphorigenic and reinforcing properties make it a potentially attractive alternative, however.
ADHD. Approximately 30% of ADHD patients do not respond to or are unable to tolerate conventional stimulant medications such as methylphenidate and dextroamphetamine.27 Several studies have evaluated modafinil as a potential treatment for ADHD based on the drug’s action on arousal and attention systems. Although modafinil’s precise mechanism of action in ADHD is unknown, proposed mechanisms include:
- hypothalamic and cerebral cortex neuronal activation
- action on histamine that results in internal vigilance.28
Can modafinil help patients with mood disorders?
| Author | Study design | Modafinil dose | Conclusion |
|---|---|---|---|
| Major depressive disorder | |||
| Fava et al, 200514 | 8-week, double-blind, placebo-controlled; 331 subjects with partial or no response to SSRI monotherapy | 200 mg/d | No significant difference between modafinil and placebo at final visit |
| DeBattista et al, 200315 | 6-week, double-blind, placebo-controlled; 136 subjects with partial response to antidepressant therapy | 100 to 400 mg/d | Significant improvement in sleepiness by week 1 and fatigue by week 2, but differences between modafinil and placebo were not statistically significant by end of study |
| Konuk et al, 200616 | 6-week, open-label; 25 subjects with residual sleepiness or fatigue after SSRI therapy | 100 to 200 mg/d | All patients showed significant improvement in sleepiness, fatigue, and HAM-D scores |
| Bipolar depression | |||
| Frye et al, 200722 | 6-week, double-blind, placebo-controlled trial; 85 subjects who did not respond to a mood stabilizer with or without concomitant antidepressant therapy | 100 to 200 mg/d (mean 177 mg/d) | 44% of modafinil patients achieved ≥50% reduction in IDS score compared with 23% in placebo group (P=0.03) |
| HAM-D: Hamilton Rating Scale for Depression; IDS: Inventory for Depressive Symptoms; SSRI: selective serotonin reuptake inhibitor | |||
CASE 2: Another Tx for ADHD
Matt, age 8, is referred to our outpatient child psychiatric clinic after his parents noted declining school performance associated with increased aggression and irritability. Our assessment strongly supports a diagnosis of ADHD without comorbid conditions. We start Matt on methylphenidate, 5 mg twice daily, which quickly improves his ADHD symptoms. However, the medication causes GI side effects and profound sleep and weight changes.
Matt’s parents request that their son be treated with a different type of agent. A trial of atomoxetine is not as effective as the initial methylphenidate dosage and produces similar side effects. We then consider modafinil because of its side effect profile. We start Matt on 100 mg once daily and titrate up to 200 mg/d 4 weeks later. Matt and his parents notice an immediate improvement in his ADHD symptoms with no side effects.
In children and adolescents. Wigal et al29 reviewed pooled data from 3 randomized, double-blind, placebo-controlled studies of modafinil in pediatric ADHD (Table 4). Modafinil was well tolerated and improved ADHD symptoms and behaviors regardless of patients’ stimulant use history.
In a recent open-label study, 220 children and young adolescents with ADHD who had completed 4 weeks of a double-blind, placebo-controlled trial were evaluated for an additional 8 weeks. Modafinil improved ADHD symptoms and overall clinical condition as determined by the parent- or clinician-completed ADHD Rating Scale-IV Home Version, the parent-completed Conners’ ADHD/DSM-IV Scale Parent Version, and the clinician-rated CGI scale.30 Insomnia, headache, and decreased appetite were the most commonly reported adverse events.
In adults. The results of 2 double-blind, placebo-controlled trials of modafinil in adults with ADHD have been positive:
- In 1 study, modafinil (mean 206.8 mg/d) was more effective than placebo and comparable to dextroamphetamine in improving ADHD symptoms.31
- In another, modafinil (a single 200-mg dose) increased cognitive performance during treatment.32
Schizophrenia. Double-blind, randomized placebo-controlled studies have evaluated modafinil for improving cognitive function and reducing negative symptoms in patients with schizophrenia. Results have been inconsistent.
One double-blind, randomized, placebo-controlled crossover study of 20 patients with chronic schizophrenia found that modafinil, 200 mg/d, significantly improved short-term verbal memory span and attentional set shifting—the ability to discriminate and selectively attend to various stimulus dimensions (Table 5).33 Two other controlled studies showed no differences between the effects of modafinil and placebo on schizophrenia’s fatigue, cognition, or positive or negative symptoms.34,35
Summary. Although open-label studies have shown modafinil has beneficial effects on cognitive symptoms, controlled data are scarce. Reports of modafinil-induced psychosis or mania11 may limit the drug’s usefulness in schizophrenia patients.
Cocaine dependence. No medications are FDA-approved for treating cocaine dependence. A placebo-controlled, double-blind trial found that modafinil blunts cocaine euphoria under controlled conditions.36 This effect is hypothesized to be secondary to modafinil’s glutamate-enhancing and gamma-aminobutyric acid inhibitory effects.37
Summary. A single study supports using modafinil to improve outcomes in cocaine-dependent patients receiving standardized psychosocial treatment. More research is needed.
Table 4
Modafinil and ADHD: What the evidence says
| Author | Study design | Modafinil dosage | Conclusion |
|---|---|---|---|
| Wigal et al, 200629 | Analysis of data from 3 double-blind, placebo-controlled trials; total 638 children/adolescents, some of whom had received prior stimulant therapy | 170 to 425 mg/d | Whether or not patients received prior stimulant treatment, modafinil significantly improved ADHD symptoms and was well tolerated |
| Boellner et al, 200630 | 8-week, open-label extension of a 4-week double-blind, placebo-controlled trial; 220 subjects ages 6-14 | 100 to 400 mg/d | Modafinil improved ADHD symptoms and overall clinical condition |
| Taylor et al, 200031 | 2-week, double-blind, placebo-controlled crossover comparing modafinil with dextroamphetamine; 22 adults | Mean 206.8 mg/d | Both modafinil and dextroamphetamine significantly improved ADHD symptoms compared with placebo |
| Turner et al, 200432 | Double-blind, placebo-controlled crossover; 20 adults | Single 200-mg dose | Modafinil improved results on cognitive tests, including short-term memory span, visual memory, spatial planning, and sustained attention |
| ADHD: attention-deficit/hyperactivity disorder | |||
Table 5
Modafinil for schizophrenia or cocaine dependence:
More research is needed
| Author | Study design | Modafinil dosage | Conclusion |
|---|---|---|---|
| Schizophrenia | |||
| Turner et al, 200433 | Double-blind, placebo-controlled crossover; 20 adults | 200 mg/d | Modafinil significantly improved attentional set shifting and short-term verbal memory span |
| Sevy et al, 200534 | 8-week, double-blind, placebo-controlled; 24 subjects | Up to 200 mg/d | No significant difference between modafinil and placebo in reducing fatigue or positive or negative symptoms or in improving cognition |
| Pierre et al, 200735 | 8-week, double-blind, placebo-controlled; 20 subjects | 100 to 200 mg/d | Modafinil did not significantly improve neurocognitive or negative symptoms |
| Cocaine dependence | |||
| Dackis et al, 200538 | 8-week, double-blind, placebo-controlled; 62 cocaine-dependent subjects | 400 mg/d | Patients receiving modafinil provided significantly more cocaine-negative urine samples and were significantly more likely to achieve =3 weeks cocaine abstinence than those receiving placebo |
Related resource
- Ballon JS, Feifel D. A systematic review of modafinil: potential clinical uses and mechanisms of action. J Clin Psychiatry 2006;67(4):554-66.
- Atomoxetine • Strattera
- Bupropion • Wellbutrin
- Carbamazepine • Carbatrol, Tegretol, others
- Citalopram • Celexa
- Dextroamphetamine • Dexedrine, DextroStat
- Diazepam • Valium
- Erythromycin • Ery-Tab, Eryc, others
- Fluoxetine • Prozac
- Lithium • Eskalith, Lithobid
- Methylphenidate • Ritalin, others
- Modafinil • Provigil
- Phenytoin • Dilantin
- Sertraline • Zoloft
- Venlafaxine • Effexor
- Zolpidem • Ambien
Dr. Ramaswamy receives research support from Bristol-Myers Squibb, Shire, and Forest Pharmaceuticals and is a consultant to Dainippon Sumitomo Pharma.
Dr. Mattai reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
Dr. Wilson receives research support from, is a consultant to, or is a speaker for the National Institute of Mental Health, the Substance Abuse and Mental Health Services Administration, the Veterans Administration, the State of Nebraska, the State of Ohio, Health Futures Foundation, Inc., Abbott Laboratories, Astra-Zeneca, Bristol-Myers Squibb, Elan, Eli Lilly and Company, GlaxoSmithKline, Janssen, Ortho-McNeil, Pfizer, and Wyeth.
Ms. B, a middle-aged mother of 3, is being monitored for bipolar disorder. She has a history of stimulant abuse but has been in remission for 5 years. She complains of excessive daytime sleepiness. Most days she wakes at 7 AM, but sleeps on several occasions during the day. She also complains of fatigue and lack of motivation.
She is being treated with lithium, venlafaxine, and zolpidem and reports good adherence. Basic laboratory work and serum lithium levels are within acceptable ranges. Her symptoms do not improve when venlafaxine is titrated from 225 mg/d to 300 mg/d. She also reports previously failed trials with bupropion and fluoxetine.
We decide to try a psychostimulant as an augmenting agent. Because of her past stimulant abuse, we add modafinil, 100 mg/d and increase to 200 mg/d. Ms. B reports improvement in her daytime sleepiness and fatigue and—except for a mild headache—tolerates the medication well.
Modafinil is being investigated for potential roles in managing inattention, excess sleepiness, fatigue, and cognitive dysfunction associated with:
- mood disorders (major depression and bipolar depression)
- attention-deficit/hyperactivity disorder (ADHD)
- schizophrenia
- cocaine dependence.
This article discusses how the drug promotes wakefulness, how it might improve cognitive function, and what the evidence reveals about off-label indications.
How it works
Although modafinil’s precise mechanism of action is unknown, it is believed to promote wakefulness more selectively than conventional stimulants such as amphetamine and methylphenidate. Modafinil does not bind to norepinephrine, serotonin, dopamine, or benzodiazepine receptors.1,2 It might target specific hypothalamic regions such as the tuberomammillary nucleus and orexin neurons, which are peptide neurotransmitters that promote wakefulness.3,4
Clinical trials found that modafinil has beneficial effects on:
- working memory, recognition memory, and sustained attention in healthy humans
- prefrontal-dependent cognitive functions in schizophrenia, major depression, and adult ADHD.5
Evidence for approved indications
Modafinil is indicated to improve wakefulness in patients who have excessive sleepiness associated with narcolepsy, obstructive sleep apnea, or shift work sleep disorder. It was approved for reducing excessive sleepiness in narcoleptic patients after two 9-week placebo-controlled clinical trials. The drug significantly reduced sleepiness and improved overall disease status as measured by the Clinical Global Impression of Change (CGI-C) scale.6,7
In patients with shift work sleep disorder, a 12-week placebo-controlled clinical trial found that modafinil significantly improved sleep latency and CGI-C scores.10
Dosage and side effects. For patients with narcolepsy or obstructive sleep apnea, the recommended dose is 200 mg given in the morning.11 For patients prescribed modafinil for work-time wakefulness, the dose is 200 mg 1 hour before their work shift. Lower doses are recommended for patients who are elderly or have hepatic impairment. Those with severe hepatic impairment typically are prescribed 100 mg/d.11 Modafinil is rapidly absorbed and is metabolized primarily by the liver (Table 1). A summary of potential drug-drug interactions appears in Table 2.11
In pivotal trials, adverse events that occurred more frequently with modafinil than with placebo and in >5% of the study population included headache, nausea, nervousness, rhinitis, diarrhea, back pain, insomnia, dizziness, and dyspepsia. Headache was most commonly reported; in most patients, it resolved soon after they started taking modafinil. Post-marketing reports have included cases of psychosis, mania, and suspected serious skin reactions, including Stevens-Johnson syndrome.11 Modafinil lacks euphorigenic properties and has minimal potential for abuse.12
Table 1
Modafinil’s pharmacokinetics
| Absorbed rapidly, with peak plasma concentrations at 2 to 4 hours |
| Apparent steady states reached after 2 to 4 days of dosing |
| Half-life: 15 hours |
| Major route of elimination (~90%) is metabolism, primarily by the liver |
Selected drug-drug interactions with modafinil
| Action of modafinil | Potential drug interactions |
|---|---|
| Increases elimination of CYP 3A4 substrates | Carbamazepine, phenytoin may decrease modafinil levels Azole antifungals, protease inhibitors, and erythromycin may increase modafinil levels |
| Inhibits CYP 2C19 enzyme | Modafinil may increase levels of citalopram, diazepam, and sertraline |
| Decreases absorption of ethinyl estradiol | Modafinil can decrease effectiveness of oral contraceptives |
| CYP: cytochrome P-450 | |
| Source: Reference 11 | |
Evidence for off-label uses
Major depressive disorder (MDD). The fatigue and excessive sleepiness often seen with MDD often persist after other depressive symptoms have remitted with antidepressant treatment.13 Patients with these symptoms might benefit from modafinil’s stimulating properties. Conventional stimulants such as methylphenidate have been used to improve neurovegetative symptoms of depression, but modafinil offers several advantages:
- decreased adverse CNS effects
- fewer drug-drug interactions
- minimal risk for dependence or abuse.
A 6-week open-label study of 25 depressed patients with residual fatigue and sleepiness showed that adjunctive modafinil, 100 to 200 mg/d, significantly improved these symptoms, as well as Hamilton Rating Scale for Depression (HAM-D) score, as early as week 2. Seventy-six percent of patients responded to treatment, defined as a >50% reduction in HAM-D scores.16
Several open-label studies and case re-ports have evaluated adjunctive modafinil use in patients with:
- depression characterized by ongoing lethargy or apathy17
- depression with atypical features18
- seasonal affective disorder19
- partial response to antidepressants.20,21
Bipolar depression. A 6-week, double-blind, placebo-controlled trial randomly assigned 85 patients with bipolar depression to adjunctive modafinil, 100 to 200 mg/d, or placebo for 6 weeks (Table 3).22 The number of patients receiving an antidepressant or mood stabilizer was not significantly different between the modafinil and placebo groups.
The primary outcome measure was change in the Inventory for Depressive Symptoms (IDS) score from baseline to endpoint. Forty-four percent of patients receiving modafinil achieved a ≥50% reduction in IDS score, compared with 23% of the placebo group; this difference was statistically significant (P=0.03).
In this study, modafinil was well tolerated and did not induce mania or hypomania. Cases of modafinil-induced mania have been reported elsewhere.23,24
The mechanisms of modafinil’s antidepressant effects are unclear. The drug does not cause release of norepinephrine or dopamine. One study proposed that modafinil acts by releasing histamine and activating noradrenaline receptors.25 Activation of these receptors increases dopamine and norepinephrine in these areas, and excites histaminergic tuberomammillary neurons, increasing histamine levels. Another trial suggested that modafinil may improve mood by mechanisms similar to the antidepressant effects induced by sleep deprivation.26
Summary. Modafinil may have a role in managing residual fatigue and excessive sleepiness associated with MDD and bipolar depression. Evidence for a mood-elevating effect is minimal; additional studies are needed. Adjunctive modafinil and conventional stimulants have not been compared head-to-head in patients with mood disorders. Modafinil’s tolerability profile and lack of euphorigenic and reinforcing properties make it a potentially attractive alternative, however.
ADHD. Approximately 30% of ADHD patients do not respond to or are unable to tolerate conventional stimulant medications such as methylphenidate and dextroamphetamine.27 Several studies have evaluated modafinil as a potential treatment for ADHD based on the drug’s action on arousal and attention systems. Although modafinil’s precise mechanism of action in ADHD is unknown, proposed mechanisms include:
- hypothalamic and cerebral cortex neuronal activation
- action on histamine that results in internal vigilance.28
Can modafinil help patients with mood disorders?
| Author | Study design | Modafinil dose | Conclusion |
|---|---|---|---|
| Major depressive disorder | |||
| Fava et al, 200514 | 8-week, double-blind, placebo-controlled; 331 subjects with partial or no response to SSRI monotherapy | 200 mg/d | No significant difference between modafinil and placebo at final visit |
| DeBattista et al, 200315 | 6-week, double-blind, placebo-controlled; 136 subjects with partial response to antidepressant therapy | 100 to 400 mg/d | Significant improvement in sleepiness by week 1 and fatigue by week 2, but differences between modafinil and placebo were not statistically significant by end of study |
| Konuk et al, 200616 | 6-week, open-label; 25 subjects with residual sleepiness or fatigue after SSRI therapy | 100 to 200 mg/d | All patients showed significant improvement in sleepiness, fatigue, and HAM-D scores |
| Bipolar depression | |||
| Frye et al, 200722 | 6-week, double-blind, placebo-controlled trial; 85 subjects who did not respond to a mood stabilizer with or without concomitant antidepressant therapy | 100 to 200 mg/d (mean 177 mg/d) | 44% of modafinil patients achieved ≥50% reduction in IDS score compared with 23% in placebo group (P=0.03) |
| HAM-D: Hamilton Rating Scale for Depression; IDS: Inventory for Depressive Symptoms; SSRI: selective serotonin reuptake inhibitor | |||
CASE 2: Another Tx for ADHD
Matt, age 8, is referred to our outpatient child psychiatric clinic after his parents noted declining school performance associated with increased aggression and irritability. Our assessment strongly supports a diagnosis of ADHD without comorbid conditions. We start Matt on methylphenidate, 5 mg twice daily, which quickly improves his ADHD symptoms. However, the medication causes GI side effects and profound sleep and weight changes.
Matt’s parents request that their son be treated with a different type of agent. A trial of atomoxetine is not as effective as the initial methylphenidate dosage and produces similar side effects. We then consider modafinil because of its side effect profile. We start Matt on 100 mg once daily and titrate up to 200 mg/d 4 weeks later. Matt and his parents notice an immediate improvement in his ADHD symptoms with no side effects.
In children and adolescents. Wigal et al29 reviewed pooled data from 3 randomized, double-blind, placebo-controlled studies of modafinil in pediatric ADHD (Table 4). Modafinil was well tolerated and improved ADHD symptoms and behaviors regardless of patients’ stimulant use history.
In a recent open-label study, 220 children and young adolescents with ADHD who had completed 4 weeks of a double-blind, placebo-controlled trial were evaluated for an additional 8 weeks. Modafinil improved ADHD symptoms and overall clinical condition as determined by the parent- or clinician-completed ADHD Rating Scale-IV Home Version, the parent-completed Conners’ ADHD/DSM-IV Scale Parent Version, and the clinician-rated CGI scale.30 Insomnia, headache, and decreased appetite were the most commonly reported adverse events.
In adults. The results of 2 double-blind, placebo-controlled trials of modafinil in adults with ADHD have been positive:
- In 1 study, modafinil (mean 206.8 mg/d) was more effective than placebo and comparable to dextroamphetamine in improving ADHD symptoms.31
- In another, modafinil (a single 200-mg dose) increased cognitive performance during treatment.32
Schizophrenia. Double-blind, randomized placebo-controlled studies have evaluated modafinil for improving cognitive function and reducing negative symptoms in patients with schizophrenia. Results have been inconsistent.
One double-blind, randomized, placebo-controlled crossover study of 20 patients with chronic schizophrenia found that modafinil, 200 mg/d, significantly improved short-term verbal memory span and attentional set shifting—the ability to discriminate and selectively attend to various stimulus dimensions (Table 5).33 Two other controlled studies showed no differences between the effects of modafinil and placebo on schizophrenia’s fatigue, cognition, or positive or negative symptoms.34,35
Summary. Although open-label studies have shown modafinil has beneficial effects on cognitive symptoms, controlled data are scarce. Reports of modafinil-induced psychosis or mania11 may limit the drug’s usefulness in schizophrenia patients.
Cocaine dependence. No medications are FDA-approved for treating cocaine dependence. A placebo-controlled, double-blind trial found that modafinil blunts cocaine euphoria under controlled conditions.36 This effect is hypothesized to be secondary to modafinil’s glutamate-enhancing and gamma-aminobutyric acid inhibitory effects.37
Summary. A single study supports using modafinil to improve outcomes in cocaine-dependent patients receiving standardized psychosocial treatment. More research is needed.
Table 4
Modafinil and ADHD: What the evidence says
| Author | Study design | Modafinil dosage | Conclusion |
|---|---|---|---|
| Wigal et al, 200629 | Analysis of data from 3 double-blind, placebo-controlled trials; total 638 children/adolescents, some of whom had received prior stimulant therapy | 170 to 425 mg/d | Whether or not patients received prior stimulant treatment, modafinil significantly improved ADHD symptoms and was well tolerated |
| Boellner et al, 200630 | 8-week, open-label extension of a 4-week double-blind, placebo-controlled trial; 220 subjects ages 6-14 | 100 to 400 mg/d | Modafinil improved ADHD symptoms and overall clinical condition |
| Taylor et al, 200031 | 2-week, double-blind, placebo-controlled crossover comparing modafinil with dextroamphetamine; 22 adults | Mean 206.8 mg/d | Both modafinil and dextroamphetamine significantly improved ADHD symptoms compared with placebo |
| Turner et al, 200432 | Double-blind, placebo-controlled crossover; 20 adults | Single 200-mg dose | Modafinil improved results on cognitive tests, including short-term memory span, visual memory, spatial planning, and sustained attention |
| ADHD: attention-deficit/hyperactivity disorder | |||
Table 5
Modafinil for schizophrenia or cocaine dependence:
More research is needed
| Author | Study design | Modafinil dosage | Conclusion |
|---|---|---|---|
| Schizophrenia | |||
| Turner et al, 200433 | Double-blind, placebo-controlled crossover; 20 adults | 200 mg/d | Modafinil significantly improved attentional set shifting and short-term verbal memory span |
| Sevy et al, 200534 | 8-week, double-blind, placebo-controlled; 24 subjects | Up to 200 mg/d | No significant difference between modafinil and placebo in reducing fatigue or positive or negative symptoms or in improving cognition |
| Pierre et al, 200735 | 8-week, double-blind, placebo-controlled; 20 subjects | 100 to 200 mg/d | Modafinil did not significantly improve neurocognitive or negative symptoms |
| Cocaine dependence | |||
| Dackis et al, 200538 | 8-week, double-blind, placebo-controlled; 62 cocaine-dependent subjects | 400 mg/d | Patients receiving modafinil provided significantly more cocaine-negative urine samples and were significantly more likely to achieve =3 weeks cocaine abstinence than those receiving placebo |
Related resource
- Ballon JS, Feifel D. A systematic review of modafinil: potential clinical uses and mechanisms of action. J Clin Psychiatry 2006;67(4):554-66.
- Atomoxetine • Strattera
- Bupropion • Wellbutrin
- Carbamazepine • Carbatrol, Tegretol, others
- Citalopram • Celexa
- Dextroamphetamine • Dexedrine, DextroStat
- Diazepam • Valium
- Erythromycin • Ery-Tab, Eryc, others
- Fluoxetine • Prozac
- Lithium • Eskalith, Lithobid
- Methylphenidate • Ritalin, others
- Modafinil • Provigil
- Phenytoin • Dilantin
- Sertraline • Zoloft
- Venlafaxine • Effexor
- Zolpidem • Ambien
Dr. Ramaswamy receives research support from Bristol-Myers Squibb, Shire, and Forest Pharmaceuticals and is a consultant to Dainippon Sumitomo Pharma.
Dr. Mattai reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
Dr. Wilson receives research support from, is a consultant to, or is a speaker for the National Institute of Mental Health, the Substance Abuse and Mental Health Services Administration, the Veterans Administration, the State of Nebraska, the State of Ohio, Health Futures Foundation, Inc., Abbott Laboratories, Astra-Zeneca, Bristol-Myers Squibb, Elan, Eli Lilly and Company, GlaxoSmithKline, Janssen, Ortho-McNeil, Pfizer, and Wyeth.
1. Mignot E, Nishino S, Guilleminault C, et al. Modafinil binds to dopamine uptake carrier site with low affinity. Sleep 1994;17:436-7.
2. Lin J-S, Hou Y, Jouvet M. Potential brain neuronal targets for amphetamine, methylphenidate, and modafinil induced wakefulness, evidenced by c-fos immunocytochemistry in the cat. Proc Natl Acad Sci USA 1996;93:14128-33.
3. Scammell TE, Estabrooke IV, McCarthy MT, et al. Hypothalamic arousal regions are activated during modafinil-induced wakefulness. J Neurosci 2000;20(22):8620-8.
4. Stenberg D. Neuroanatomy and neurochemistry of sleep. Cell Mol Life Sci 2007;64(10):1187-204.
5. Minzenberg MJ, Carter CS. Modafinil: a review of neurochemical actions and effects on cognition. Neuropsychopharmacology In press.
6. U.S. Modafinil in Narcolepsy Multicenter Study Group. Randomized trial of modafinil for the treatment of pathological somnolence in narcolepsy. Ann Neurol 1998;43(1):88-97.
7. U.S. Modafinil in Narcolepsy Multicenter Study Group. Randomized trial of modafinil as a treatment for the excessive daytime somnolence of narcolepsy. Neurology 2000;54:1166-75.
8. Black JE, Hirshkowitz M. Modafinil for treatment of residual excessive sleepiness in nasal continuous positive airway pressure-treated obstructive sleep apnea/hypopnea syndrome. Sleep 2005;28(4):464-71.
9. Pack AI, Black JE, Schwartz JR, Matheson JK. Modafinil as adjunct therapy for daytime sleepiness in obstructive sleep apnea. Am J Respir Crit Care Med 2001;164(9):1675-81.
10. Czeisler CA, Walsh JK, Roth T, et al. and the U.S. Modafinil in Shift Work Sleep Disorder Study Group. Modafinil for excessive sleepiness associated with shift-work sleep disorder. N Engl J Med. 2005;353(5):476-86. Published correction appears in: N Engl J Med 2005;353(10):1078.-
11. Provigil [package insert] West Chester, PA: Cephalon Inc; 2004.
12. Myrick H, Malcolm R, Taylor B, et al. Modafinil: preclinical, clinical, and post-marketing surveillance—a review of abuse liability issues. Ann Clin Psychiatry 2004;16(2):101-9.
13. Baldwin DS, Papakostas GI. Symptoms of fatigue and sleepiness in major depressive disorder. J Clin Psychiatry 2006;67(suppl 6):9-15.
14. Fava M, Thase ME, DeBattista C. A multicenter, placebo-controlled study of modafinil augmentation in partial responders to selective serotonin reuptake inhibitors with persistent fatigue and sleepiness. J Clin Psychiatry 2005;66(1):85-93.
15. DeBattista C, Doghramji K, Menza MA, et al. and the Modafinil in Depression Study Group. Adjunct modafinil for the short-term treatment of fatigue and sleepiness in patients with major depressive disorder: a preliminary double-blind, placebo-controlled study. J Clin Psychiatry 2003;64(9):1057-64.
16. Konuk N, Atasoy N, Atik L, Akay O. Open-label study of adjunct modafinil for the treatment of patients with fatigue, sleepiness, and major depression treated with selective serotonin reuptake inhibitors. Adv Ther 2006;23(4):646-54.
17. Markovitz PJ, Wagner S. An open-label trial of modafinil augmentation in patients with partial response to antidepressant therapy. J Clin Psychopharmacol 2003;23(2):207-9.
18. Vaishnavi S, Gadde K, Alamy S, et al. Modafinil for atypical depression: effects of open-label and double-blind discontinuation treatment. J Clin Psychopharmacol 2006; 26(4): 373-8. Published correction appears in: J Clin Psychopharmacol. 2006;26(5):523.
19. Lundt L. Modafinil treatment in patients with seasonal affective disorder/winter depression: an open-label pilot study. J Affect Disord 2004;81(2):173-8.
20. DeBattista C, Lembke A, Solvason HB, et al. A prospective trial of modafinil as an adjunctive treatment of major depression. J Clin Psychopharmacol 2004;24(1):87-90.
21. Rasmussen NA, Schroder P, Olsen LR, et al. Modafinil augmentation in depressed patients with partial response to antidepressants: a pilot study on self-reported symptoms covered by the Major Depression Inventory (MDI) and the Symptom Checklist (SCL-92). Nord J Psychiatry 2005;59(3):173-8.
22. Frye MA, Grunze H, Suppes T, et al. A placebo-controlled evaluation of adjunctive modafinil in the treatment of bipolar depression. Am J Psychiatry 2007;164:1242-9.
23. Wolf J, Fiedler U, Anghelescu I, Schwertfeger N. Manic switch in a patient with treatment-resistant bipolar depression treated with modafinil. J Clin Psychiatry 2006;67(11):1817.-
24. Vorspan F, Warot D, Consoli A, et al. Mania in a boy treated with modafinil for narcolepsy. Am J Psychiatry 2005;162(4):813-4.
25. McClellan KJ, Spencer CM. Modafinil: a review of its pharmacology and clinical efficacy in the management of narcolepsy. CNS Drugs 1998;9(4):311-24.
26. Ballon JS, Feifel D. A systematic review of modafinil: potential clinical uses and mechanisms of action. J Clin Psychiatry 2006;67(4):554-66.
27. Dulcan M. Practice parameters for the assessment and treatment of children, adolescents and adults with attentiondeficit hyperactivity disorder. American Academy of Child and Adolescent Psychiatry. J Am Acad Child Adolesc Psychiatry 1997;36(suppl 10):85S-121S.
28. Lindsay SE, Gudelsky GA, Heaton PC. Use of modafinil for the treatment of attention deficit/hyperactivity disorder. Ann Pharmacother 2006;40(10):1829-33.
29. Wigal SB, Biederman J, Swanson JM, et al. Efficacy and safety of modafinil film-coated tablets in children and adolescents with or without prior stimulant treatment for attention-deficit/hyperactivity disorder: pooled analysis of 3 randomized, double-blind, placebo-controlled studies. Prim Care Companion J Clin Psychiatry 2006;8(6):352-60.
30. Boellner SW, Earl CQ, Arora S. Modafinil in children and adolescents with attention-deficit/hyperactivity disorder: a preliminary 8-week, open-label study. Curr Med Res Opin 2006;22(12):2457-65.
31. Taylor FB, Russo J. Efficacy of modafinil compared to dextroamphetamine for the treatment of attention deficit hyperactivity disorder in adults. J Child Adolesc Psychopharmacol 2000;10(4):311-20.
32. Turner DC, Clark L, Dowson J, et al. Modafinil improves cognition and response inhibition in adult attention-deficit/ hyperactivity disorder. Biol Psychiatry 2004;55(10):1031-40.
33. Turner DC, Clark L, Pomarol-Clotet E, et al. Modafinil improves cognition and attentional set shifting in patients with chronic schizophrenia. Neuropsychopharmacology 2004;29(7):1363-73.
34. Sevy S, Rosenthal MH, Alvir J, et al. Double-blind, placebo-controlled study of modafinil for fatigue and cognition in schizophrenia patients treated with psychotropic medications. J Clin Psychiatry 2005;66(7):839-43.
35. Pierre JM, Peloian JH, Wirshing DA, et al. A randomized, double-blind, placebo-controlled trial of modafinil for negative symptoms in schizophrenia. J Clin Psychiatry 2007;68(5):705-10.
36. Dackis CA, Lynch KG, Yu E, et al. Modafinil and cocaine: a double-blind, placebo-controlled drug interaction study. Drug Alcohol Depend 2003;70(1):29-37.
37. Perez de la Mora M, Aguilar-Garcia A, Ramon-Frias T, et al. Effects of the vigilance promoting drug modafinil on the synthesis of GABA and glutamate in slices of rat hypothalamus. Neurosci Lett 1999;259:181-5.
38. Dackis CA, Kampman KM, Lynch KG, et al. A double-blind, placebo-controlled trial of modafinil for cocaine dependence. Neuropsychopharmacology 2005;30(1):205-11.
1. Mignot E, Nishino S, Guilleminault C, et al. Modafinil binds to dopamine uptake carrier site with low affinity. Sleep 1994;17:436-7.
2. Lin J-S, Hou Y, Jouvet M. Potential brain neuronal targets for amphetamine, methylphenidate, and modafinil induced wakefulness, evidenced by c-fos immunocytochemistry in the cat. Proc Natl Acad Sci USA 1996;93:14128-33.
3. Scammell TE, Estabrooke IV, McCarthy MT, et al. Hypothalamic arousal regions are activated during modafinil-induced wakefulness. J Neurosci 2000;20(22):8620-8.
4. Stenberg D. Neuroanatomy and neurochemistry of sleep. Cell Mol Life Sci 2007;64(10):1187-204.
5. Minzenberg MJ, Carter CS. Modafinil: a review of neurochemical actions and effects on cognition. Neuropsychopharmacology In press.
6. U.S. Modafinil in Narcolepsy Multicenter Study Group. Randomized trial of modafinil for the treatment of pathological somnolence in narcolepsy. Ann Neurol 1998;43(1):88-97.
7. U.S. Modafinil in Narcolepsy Multicenter Study Group. Randomized trial of modafinil as a treatment for the excessive daytime somnolence of narcolepsy. Neurology 2000;54:1166-75.
8. Black JE, Hirshkowitz M. Modafinil for treatment of residual excessive sleepiness in nasal continuous positive airway pressure-treated obstructive sleep apnea/hypopnea syndrome. Sleep 2005;28(4):464-71.
9. Pack AI, Black JE, Schwartz JR, Matheson JK. Modafinil as adjunct therapy for daytime sleepiness in obstructive sleep apnea. Am J Respir Crit Care Med 2001;164(9):1675-81.
10. Czeisler CA, Walsh JK, Roth T, et al. and the U.S. Modafinil in Shift Work Sleep Disorder Study Group. Modafinil for excessive sleepiness associated with shift-work sleep disorder. N Engl J Med. 2005;353(5):476-86. Published correction appears in: N Engl J Med 2005;353(10):1078.-
11. Provigil [package insert] West Chester, PA: Cephalon Inc; 2004.
12. Myrick H, Malcolm R, Taylor B, et al. Modafinil: preclinical, clinical, and post-marketing surveillance—a review of abuse liability issues. Ann Clin Psychiatry 2004;16(2):101-9.
13. Baldwin DS, Papakostas GI. Symptoms of fatigue and sleepiness in major depressive disorder. J Clin Psychiatry 2006;67(suppl 6):9-15.
14. Fava M, Thase ME, DeBattista C. A multicenter, placebo-controlled study of modafinil augmentation in partial responders to selective serotonin reuptake inhibitors with persistent fatigue and sleepiness. J Clin Psychiatry 2005;66(1):85-93.
15. DeBattista C, Doghramji K, Menza MA, et al. and the Modafinil in Depression Study Group. Adjunct modafinil for the short-term treatment of fatigue and sleepiness in patients with major depressive disorder: a preliminary double-blind, placebo-controlled study. J Clin Psychiatry 2003;64(9):1057-64.
16. Konuk N, Atasoy N, Atik L, Akay O. Open-label study of adjunct modafinil for the treatment of patients with fatigue, sleepiness, and major depression treated with selective serotonin reuptake inhibitors. Adv Ther 2006;23(4):646-54.
17. Markovitz PJ, Wagner S. An open-label trial of modafinil augmentation in patients with partial response to antidepressant therapy. J Clin Psychopharmacol 2003;23(2):207-9.
18. Vaishnavi S, Gadde K, Alamy S, et al. Modafinil for atypical depression: effects of open-label and double-blind discontinuation treatment. J Clin Psychopharmacol 2006; 26(4): 373-8. Published correction appears in: J Clin Psychopharmacol. 2006;26(5):523.
19. Lundt L. Modafinil treatment in patients with seasonal affective disorder/winter depression: an open-label pilot study. J Affect Disord 2004;81(2):173-8.
20. DeBattista C, Lembke A, Solvason HB, et al. A prospective trial of modafinil as an adjunctive treatment of major depression. J Clin Psychopharmacol 2004;24(1):87-90.
21. Rasmussen NA, Schroder P, Olsen LR, et al. Modafinil augmentation in depressed patients with partial response to antidepressants: a pilot study on self-reported symptoms covered by the Major Depression Inventory (MDI) and the Symptom Checklist (SCL-92). Nord J Psychiatry 2005;59(3):173-8.
22. Frye MA, Grunze H, Suppes T, et al. A placebo-controlled evaluation of adjunctive modafinil in the treatment of bipolar depression. Am J Psychiatry 2007;164:1242-9.
23. Wolf J, Fiedler U, Anghelescu I, Schwertfeger N. Manic switch in a patient with treatment-resistant bipolar depression treated with modafinil. J Clin Psychiatry 2006;67(11):1817.-
24. Vorspan F, Warot D, Consoli A, et al. Mania in a boy treated with modafinil for narcolepsy. Am J Psychiatry 2005;162(4):813-4.
25. McClellan KJ, Spencer CM. Modafinil: a review of its pharmacology and clinical efficacy in the management of narcolepsy. CNS Drugs 1998;9(4):311-24.
26. Ballon JS, Feifel D. A systematic review of modafinil: potential clinical uses and mechanisms of action. J Clin Psychiatry 2006;67(4):554-66.
27. Dulcan M. Practice parameters for the assessment and treatment of children, adolescents and adults with attentiondeficit hyperactivity disorder. American Academy of Child and Adolescent Psychiatry. J Am Acad Child Adolesc Psychiatry 1997;36(suppl 10):85S-121S.
28. Lindsay SE, Gudelsky GA, Heaton PC. Use of modafinil for the treatment of attention deficit/hyperactivity disorder. Ann Pharmacother 2006;40(10):1829-33.
29. Wigal SB, Biederman J, Swanson JM, et al. Efficacy and safety of modafinil film-coated tablets in children and adolescents with or without prior stimulant treatment for attention-deficit/hyperactivity disorder: pooled analysis of 3 randomized, double-blind, placebo-controlled studies. Prim Care Companion J Clin Psychiatry 2006;8(6):352-60.
30. Boellner SW, Earl CQ, Arora S. Modafinil in children and adolescents with attention-deficit/hyperactivity disorder: a preliminary 8-week, open-label study. Curr Med Res Opin 2006;22(12):2457-65.
31. Taylor FB, Russo J. Efficacy of modafinil compared to dextroamphetamine for the treatment of attention deficit hyperactivity disorder in adults. J Child Adolesc Psychopharmacol 2000;10(4):311-20.
32. Turner DC, Clark L, Dowson J, et al. Modafinil improves cognition and response inhibition in adult attention-deficit/ hyperactivity disorder. Biol Psychiatry 2004;55(10):1031-40.
33. Turner DC, Clark L, Pomarol-Clotet E, et al. Modafinil improves cognition and attentional set shifting in patients with chronic schizophrenia. Neuropsychopharmacology 2004;29(7):1363-73.
34. Sevy S, Rosenthal MH, Alvir J, et al. Double-blind, placebo-controlled study of modafinil for fatigue and cognition in schizophrenia patients treated with psychotropic medications. J Clin Psychiatry 2005;66(7):839-43.
35. Pierre JM, Peloian JH, Wirshing DA, et al. A randomized, double-blind, placebo-controlled trial of modafinil for negative symptoms in schizophrenia. J Clin Psychiatry 2007;68(5):705-10.
36. Dackis CA, Lynch KG, Yu E, et al. Modafinil and cocaine: a double-blind, placebo-controlled drug interaction study. Drug Alcohol Depend 2003;70(1):29-37.
37. Perez de la Mora M, Aguilar-Garcia A, Ramon-Frias T, et al. Effects of the vigilance promoting drug modafinil on the synthesis of GABA and glutamate in slices of rat hypothalamus. Neurosci Lett 1999;259:181-5.
38. Dackis CA, Kampman KM, Lynch KG, et al. A double-blind, placebo-controlled trial of modafinil for cocaine dependence. Neuropsychopharmacology 2005;30(1):205-11.

