Neuroleptic malignant syndrome: Answers to 6 tough questions

Article Type
Changed
Tue, 12/11/2018 - 15:07
Display Headline
Neuroleptic malignant syndrome: Answers to 6 tough questions

Diagnosis and treatment of neuroleptic malignant syndrome (NMS) are controversial because this potentially life-threatening syndrome is rare and its presentation varies. These factors make it difficult to evaluate treatments in controlled clinical trials, and data about the relative efficacy of specific interventions are scarce. It may be possible, however, to develop rational treatment guidelines using empiric clinical data.1,2

This article examines the evidence related to 6 controversial aspects of NMS diagnosis and treatment:

  • most-reliable risk factors
  • NMS as a spectrum disorder
  • what causes NMS
  • NMS triggered by first-generation vs second-generation antipsychotics
  • first-line interventions
  • restarting antipsychotics after an NMS episode.

1. Are there reliable risk factors for NMS?

In small case-controlled studies, agitation, dehydration, and exhaustion were the most consistently found systemic factors believed to predispose patients taking antipsychotics to NMS (Table 1).3-5 Catatonia and organic brain syndromes may be separate risk factors.1,6

Preliminary studies also have implicated dopamine receptor abnormalities caused by genetic polymorphisms or effects of low serum iron.1,7,8 Pharmacologic studies have suggested that higher doses, rapid titration, and IM injections of antipsychotics are associated with increased NMS risk.3,5 Some studies suggest that 15% to 20% of NMS patients have a history of NMS episodes.1,2 In addition, high-potency first-generation antipsychotics (FGAs)—especially haloperidol—are assumed to carry higher risk than low-potency drugs and second-generation antipsychotics (SGAs), although this hypothesis remains difficult to prove.9-11

These risk factors, however, are not practical for estimating NMS risk in a given patient because they are relatively common compared with the low risk of NMS occurrence. For the vast majority of patients with psychotic symptoms, the benefits of properly indicated antipsychotic pharmacotherapy will outweigh the risks.

Table 1

Systemic
Agitation
Dehydration
Exhaustion
Low serum iron concentrations (normal: 60 to 170 mcg/dL)
Diagnoses
History of NMS
Catatonia
Organic brain syndromes
Central nervous system
Dopamine receptor dysfunction
Basal ganglia dysfunction
Sympathetic nervous system dysfunction
Pharmacologic treatment*
Intramuscular or intravenous injections
High-potency dopamine antagonists
Rapid dose titration
High doses
FGAs compared with SGAs (?)

*For individual patients, these common risk factors must be weighted again the benefits of antipsychotic therapy FGAs: first-generation antipsychotics; SGAs:second-generation antipsychotics; NMS: neuroleptic malignant syndromeSource: References 1-5

2. Is NMS related to parkinsonism, catatonia, or malignant hyperthermia?

Parkinsonsim. Some researchers have described NMS as an extreme parkinsonian crisis resulting from overwhelming blockade of dopamine pathways in the brain.1,2,12 In this view, NMS resembles the parkinsonian-hyperthermia syndrome that can occur in Parkinson's disease patients following abrupt discontinuation or loss of efficacy of dopaminergic therapy, which can be treated by reinstituting dopaminergic agents.13 Evidence to support this view includes:

  • Parkinsonian signs are a cardinal feature of NMS.
  • Withdrawal of dopamine agonists precipitates the syndrome.
  • All triggering drugs are dopamine receptor antagonists.
  • Risks of NMS correlates with drugs' dopamine receptor affinity.
  • Dopaminergic agonists may be an effective treatment.
  • Lesions in dopaminergic pathways produce a similar syndrome.
  • Patients with NMS have demonstrated low cerebrospinal fluid concentrations of the dopamine metabolite homovanillic acid.14

Catatonia. Fink et al15 and others16-18 have persuasively argued that NMS represents a form of drug-induced malignant catatonia. Evidence supporting this includes:

  • The 2 disorders share neuropsychiatric symptoms.
  • Catalonic signs are common in NMS.19
  • Malignant catatonia and NMS share physiologic and labratory signs.20
  • Reintroduction of antipsychotics can acutely worsen both conditions.
  • Benzodiazepines and electroconvulsive therapy (ECT) are effective treatments for both disorders.15-18

Lee21 examined the relationship between catatonic features and treatment response of 14 NMS patients. Most patients with catatonic symptoms responded to benzodiazepines, whereas none of those did who had an extrapyramidal-hyperthermic presentation without catatonia. Lee concluded that NMS is heterogeneous and may occur in catatonic and noncatatonic forms that differ in treatment response.

Malignant hyperthermia. Some clinicians have compared NMS with malignant hyperthermia caused by inhalational anesthetics and succinylcholine.1,2 Evidence includes

  • similar clinical signs of rigidity, hyperthermia, and hypermetabolism
  • similar psychologic and labratory signs, such as rhabdomyolysis
  • hyperthermia in both responding to dantrolene.

Although the 2 are similar in presentation, malignant hyperthermia occurs intraoperatively and reflects a pharmacogenetic disorder of calcium regulation in skeletal muscle. Additionally, rigidity in malignant hyperthermia does not respond to peripheral-acting muscle relaxants.1,22 Evidence suggests that patients who have previously experienced an NMS episodes are not at risk for malignant hyperthermia.22

3. What is the pathophysiology of NMS?

NMS pathophysiology is complex and likely involves interplay between multiple central and systemic pathways and neurotransmitters. As described above, compelling evidence suggests that dopamine blockade plays a central role.12

Dopamine blockade in the hypothalamus is believed to contribute to thermoregulatory failure, and blockade in the nigrostriatal system likely contributes to muscle rigidity and hypermetabolism. The loss of dopaminergic input to the anterior cingulate-medial orbitofrontal circuit and the lateral orbitofrontal circuit likely con-tributes to the mental status changes and catatonic features seen in NMS.12

 

 

Some researchers have proposed competing or complementary hypotheses, however. For example, Gurrera23 proposed that patients who are prone to developing NMS have a vulnerability to a hyperactive and dysregulated sympathetic nervous system, and this trait—together with dopamine system disruption induced by dopamine-blocking agents—produces NMS. Other investigators have implicated serotonin, norepinephrine, gamma-aminobutyric acid and glutaminergic mechanisms.1,12,24,25

4. Are FGAs or SGAs more likely to cause NMS?

NMS is assumed to occur less frequently in patients treated with SGAs than in those receiving FGAs, although this hypothesisis unproven. Isolated reports of NMS have been associated with nearly every SGA.9-11 It is difficult to prove FGA vs SGA liabilities because:

  • NMS is rare.
  • Dosing practices may be more conser-vative now than in the past.
  • Most clinicians are aware of the earlysigns of NMS.

In an epidemiological study of a large database, Stubner et al26 found that patients receiving SGAs had a lower risk of NMS than those treated with haloperidol.26 In this study, the overall rate of NMS was 0.02%.

NMS hotline data. We recently examined which medication classes were implicated in 111 NMS cases reported to the Neuroleptic Malignant Syndrome Information Service hotline (1-888-NMS-TEMP) between 1997 and 2006 (Figure). We included only cases of definite or probable NMS (as diagnosed by hotline consultants) in which a single antipsychotic was administered. Slightly more cases were attributed to FGAs (51%) than SGAs (45%). The remaining cases were attributed to neuroleptics used in medical settings (such as promethazineor prochlorperazine). Because they are now prescribed less often, FGAs accounted for a disproportionate number of NMS cases reported to the hotline. Haloperidol accounted for the majority of FGA cases and 44% of all cases. If we had excluded haloperidol and compared the NMS risk of SGAs to only intermediate- or low-potency FGAs, the relative advantage of SGAs would have been lost. On the other hand, it is clear that SGAs still carry a risk for NMS. Analyses suggest that the SGA-associated classic features of NMS—fever, muscle rigidity, and autonomic and mental status changes—are retained in patients receiving SGAs, although some may not develop the severe rigidity and extreme temperatures common in patients receiving FGAs.9-11 The milder clinical characteristics associated with SGAs may reflect more conservative prescribing patterns or increased awareness and earlier recognition of NMS, which would prevent fulminant presentations.

5. What is the evidence for specific NMS treatments?

NMS is rare, its presentation varies, and its progression is unpredictable. These factors make it difficult to evaluate treatments in controlled clinical trials, and data about the relative efficacy of specific interventions are scarce.

Even so, the notion that NMS represents an extreme variant of drug-induced parkinsonism or catatonia suggests that specific NMS treatments could be based on symptom severity or stage of presentation. We propose a treatment guideline basedon theoretical mechanisms and anecdotal data (Algorithm).2,27-29

Support. After immediate withdrawal of the offending medication, supportive therapy is the cornerstone of NMS treatment.1,2,27

For patients presenting with mild signs and symptoms, supportive care and careful clinical monitoring may be sufficient. Extreme hyperthermia demands volume resuscitation and cooling measures, intensive medical care, and careful monitoring for complications.

Treatment. Despite a lack of consensus on drug treatments for uncomplicated NMS, approximately 40% of patients with acute NMS receive pharmacologic treatments.2

Lorazepam, 1 to 2 mg parenterally, is a reasonable first-line therapy for NMS, especially in individuals with catatonic features.4,15-18,21,30,31 Some investigators recommend higher doses.15 Benzodiazepines are preferred if sedation is required in agitated NMS patients.4,15-18

Dopaminergic agents such as bromocriptine and amantadine enhance dopaminergic transmission to reverse parkinsonian symptoms and have been reported to reduce time to recovery and halve mortality rates when used alone or in conjunction with other treatments.13,27,32,33 Rapid discontinuation of these agents can result in rebound symptoms, although this may be true for any specific drug treatment of NMS.1,31,32

Dantrolene uncouples excitation-contraction coupling by enhancing calcium sequestration in sarcoplasmic reticulumin skeletal muscle and has been used to treat NMS hypermetabolic symptoms. Some reviews found improvement in up to 80% of NMS patients treated with dantrolene monotherapy.27,32-35 Compared with supportive care, time to recovery may be reduced—and mortality decreased by almost one-half—when dantrolene is used alone or in combination with other medications.

Not all case reports have shown that dantrolene, benzodiazepines, ordopaminergic agonists are effective in treating NMS.31,36 In our opinion, only advanced NMS cases—with extreme temperature elevations, severe rigidity, and evidence of systemic hypermetabolism—benefit from dantrolene treatment.1,2

ECT has been used successfully to reduce mortality from NMS and other catatonic-spectrum disorders. It is usually employed after supportive therapy and psychopharmacologic interventions fail.2,15,16,27,37 ECT for acute NMS typically consists of a series of 6 to 10 treatments with bilateral electrode placement. Daily ECT may be needed initially.15

 

 

6. Are antipsychotics contraindicated following an NMS episode?

The rate of NMS recurrence on retreatment with an antipsychotic has varied.38 We estimate that up to 30% of patients may be at risk of NMS recurrence when rechallenged with an antipsychotic.1 By following proper precautions (Table 2), however, you can safely treat most patients who require continued antipsychotic therapy.1,2 When you restart treatment, a lower-potency antipsychotic from a different chemical class may be a safer option than retrying the triggering agent, according to retrospective analyses of limited available data. A patient who develops NMS on a FGA might benefit from an SGA trial, although some risk of recurrence remains.1,10

Current Psychiatry 2007;6(8):89-95.
Drug brand names

  • Amantadine • Symmetrel
  • Bromocriptine • Parlodel
  • Chlorpromazine • Thorazine
  • Dantrolene • Dantrium
  • Fluphenazine • Prolixin
  • Haloperidol • Haldol
  • Lorazepam • Ativan
  • Loxapine • Loxitane
  • Perphenazine • Trilafon
  • Prochlorperazine • Compazine, Compro
  • Promethazine • Phenergan
  • Thioridazine • Mellaril

Disclosure

Dr. Strawn is an American Psychiatric Institute for Research and Education (APIRE)/Janssen Scholar.

Dr. Keck has received research support from or served as a consultant to Abbott Laboratories, American Diabetes Association, AstraZeneca Pharmaceuticals, Bristol-Myers Squibb, GlaxoSmithKline, Eli Lilly and Company, Janssen Pharmaceutica, National Institute of Mental Health, National Institute of Drug Abuse, Pfizer, Stanley Medical Research Institute, and UCB Pharma.

Dr. Caroff has received research support from Bristol-Myers Squibb, Ortho-McNeil Neurologics, and Pfizer.

References

1. Caroff SN. Neuroleptic malignant syndrome. In: Mann SC, Caroff SN, Keck PE Jr, Lazarus A, eds. Neuroleptic malignant syndrome and related conditions 2nd ed. Washington, DC: American Psychiatric Publishing Inc; 2003; 1-44.

2. Strawn JR, Keck PE, Jr, Caroff SN. Neuroleptic malignant syndrome Am J Psychiatry 2007;164:870-6.

3. Keck PE, Jr, Pope HG, Jr, Cohen BM, et al. Risk factors for neuroleptic malignant syndrome Arch Gen Psychiatry 1989;46:914-18.

4. Rosebush PI, Stewart TD. A prospective analysis of 24 episodes of neuroleptic malignant syndrome Am J Psychiatry 1989;146:717-25.

5. Berardi D, Amore M, Keck PE, Jr, et al. Clinical and pharmacologic risk factors for neuroleptic malignant syndrome: a case-control study. Biol Psychiatry 1998;44:748-54.

6. White DA, Robins AH. Catatonia: harbinger of the neuroleptic malignant syndrome Br J Psychiatry 1991;158:419-21.

7. Rosebush PI, Mazurek MF. Serum iron and neuroleptic malignant syndrome. Lancet 1991;338:149-51.

8. Lee JW. Serum iron in catatonia and neuroleptic malignant syndrome Biol Psychiatry 1998;44:499-507.

9. Ananth J, Parameswaran S, Gunatilake S, et al. Neuroleptic malignant syndrome and atypical antipsychotic drugs J Clin Psychiatry 2004;65:464-70.

10. Caroff SN, Mann SC, Campbell EC. Atypical antipsychotics and neuroleptic malignant syndrome Psychiatr Ann 2000;30:314-21.

11. Hasan S, Buckley P. Novel antipsychotics and the neuroleptic malignant syndrome Am J Psychiatry 1998;155:1113-16.

12. Mann SC, Caroff SN, Fricchione G, Campbell EC. Central dopamine hypoactivity and the pathogenesis of neuroleptic malignant syndrome Psychiatr Ann 2000;30:363-74.

13. Factor SA, Santiago A. Parkinsonism-hyperpyrexia syndrome in Parkinson’s disease. In: Frucht SJ, Fahn S, eds. Movement disorder emergencies: diagnosis and treatment. Totowa, NJ: Humana Press; 2005; 29-40.

14. Nisijima K, Ishiguro T. Cerebrospinal fluid levels of monoamine metabolites and gamma-aminobutyric acid in neuroleptic malignant syndrome. J Psychiatr Res 1995;27:233-44.

15. Fink M, Taylor MA. Neuroleptic malignant syndrome is malignant catatonia, warranting treatments efficacious for catatonia. Prog Neuropsychopharmacol Biol Psychiatry 2006;30:1182-3.

16. Fricchione G, Bush G, Fozdar M, et al. Recognition and treatment of the catatonic syndrome. J Intensive Care Med 1997;12:135-47.

17. Philbrick KL, Rummans TA. Malignant catatonia. J Neuropsychiatry Clin Neurosci 1994;6:1-13.

18. Mann SC, Caroff SN, Bleier HR, et al. Lethal catatonia. Am J Psychiatry 1986;143:1374-81.

19. Koch M, Chandragiri S, Rizvi S, et al. Catatonic signs in neuroleptic malignant syndrome. Compr Psychiatry 2000;41:73-5.

20. Lee JW. Laboratory findings. In: Caroff SN, Mann SC, Francis A, Fricchoine GL, eds. Catatonia: from psychopathology to neurobiology Washington, DC: American Psychiatric Press, Inc; 2004; 65-75.

21. Lee JW. Catatonic variants, hyperthermic extrapyramidal reactions, and subtypes of neuroleptic malignant syndrome. Ann Clin Psychiatry 2007;19:9-16.

22. Caroff SN, Rosenberg H, Mann SC, et al. Neuroleptic malignant syndrome in the perioperative setting. Am J Anesthesiol 2001;28:387-93.

23. Gurrera RJ. Sympathoadrenal hyperactivity and the etiology of neuroleptic malignant syndrome. Am J Psychiatry 1999;156:169-80.

24. Carroll BT. The universal field hypothesis of catatonia and neuroleptic malignant syndrome. CNS Spectr 2000;5:26-33.

25. Weller M, Kornhuber J. A rationale for NMDA receptor antagonist therapy of the neuroleptic malignant syndrome. Med Hypotheses 1992;38:329-33.

26. Stubner S, Rustenbeck E, Grohmann R, et al. Severe and uncommon involuntary movement disorders due to psychotropic drugs. Pharmacopsychiatry 2004;37(suppl 1):S54-S64.

27. Davis JM, Caroff SN, Mann SC. Treatment of neuroleptic malignant syndrome. Psychiatr Ann 2000;30:325-31.

28. Adityanjee PA, Singh S, Singh G, Ong S. Spectrum concept of neuroleptic malignant syndrome. Br J Psychiatry 1988;153:107-11.

29. Woodbury MM, Woodbury MA. Neuroleptic-induced catatonia as a stage in the progression toward neuroleptic malignant syndrome. J Am Acad Child Adolesc Psychiatry 1992;31:1161-4.

30. Francis A, Chondragivi S, Rizvi S, et al. Is lorazepam a treatment for neuroleptic malignant syndrome? CNS Spectr 2000;5:54-7.

31. Rosebush PI, Stewart T, Mazurek MF. The treatment of neuroleptic malignant syndrome. Are dantrolene and bromocriptine useful adjuncts to supportive care? Br J Psychiatry 1991;159:709-12.

32. Sakkas P, Davis JM, Janicak PG, Wang ZY. Drug treatment of the neuroleptic malignant syndrome. Psychopharmacol Bull 1991;27:381-4.

33. Rosenberg MR, Green M. Neuroleptic malignant syndrome: review of response to therapy. Arch Intern Med 1989;149:1927-31.

34. Yamawaki S, Morio M, Kazamutsuri G, et al. Clinical evaluation and effective usage of dantrolene sodium in neuroleptic malignant syndrome. Kiso to Rinsyou (Clinical Reports) 1993;27:1045-66.

35. Tsutsumi Y, Yamamoto K, Matsuura S, et al. The treatment of neuroleptic malignant syndrome using dantrolene sodium. Psychiatry Clin Neurosci 1998;52:433-8.

36. Reulbach U, Dutsch C, Biermann T, et al. Managing an effective treatment for neuroleptic malignant syndrome. Crit Care 2007;11:R4.-

37. Troller JN, Sachdev PS. Electroconvulsive treatment of neuroleptic malignant syndrome: a review and report of cases. Aust N Z J Psychiatry 1999;33:650-9.

38. Pope HG, Aizley HG, Keck PE, Jr, McElroy SL. Neuroleptic malignant syndrome: long term follow-up of 20 cases. J Clin Psychiatry 1991;52:208-12.

Article PDF
Author and Disclosure Information

Jeffrey R. Strawn, MD
Clinical instructor in psychiatry, department of psychiatry, University of Cincinnati College of Medicine

Paul E. Keck, Jr, MD
Professor of psychiatry, department of psychiatry, University of Cincinnati College of Medicine, president and CEO, Lindner Center of HOPE, Cincinnati, OH

Stanley N. Caroff, MD
Professor of psychiatry, department of psychiatry, University of Pennsylvania School of Medicine, chief of inpatient psychiatry, Psychiatry service, Philadelphia VA Medical Center

Issue
Current Psychiatry - 07(01)
Publications
Page Number
95-101
Legacy Keywords
neuroleptic malignant syndrome; NMS diagnosis; NMS risk factors; first-generation antipsychotics; second-generation antipsychotics; parkinsonism; catatonia; hyperthermia; NMS pathophysiology; Jeffrey R Strawn MD; Paul E Keck Jr MD; Stanley N Caroff MD
Sections
Author and Disclosure Information

Jeffrey R. Strawn, MD
Clinical instructor in psychiatry, department of psychiatry, University of Cincinnati College of Medicine

Paul E. Keck, Jr, MD
Professor of psychiatry, department of psychiatry, University of Cincinnati College of Medicine, president and CEO, Lindner Center of HOPE, Cincinnati, OH

Stanley N. Caroff, MD
Professor of psychiatry, department of psychiatry, University of Pennsylvania School of Medicine, chief of inpatient psychiatry, Psychiatry service, Philadelphia VA Medical Center

Author and Disclosure Information

Jeffrey R. Strawn, MD
Clinical instructor in psychiatry, department of psychiatry, University of Cincinnati College of Medicine

Paul E. Keck, Jr, MD
Professor of psychiatry, department of psychiatry, University of Cincinnati College of Medicine, president and CEO, Lindner Center of HOPE, Cincinnati, OH

Stanley N. Caroff, MD
Professor of psychiatry, department of psychiatry, University of Pennsylvania School of Medicine, chief of inpatient psychiatry, Psychiatry service, Philadelphia VA Medical Center

Article PDF
Article PDF

Diagnosis and treatment of neuroleptic malignant syndrome (NMS) are controversial because this potentially life-threatening syndrome is rare and its presentation varies. These factors make it difficult to evaluate treatments in controlled clinical trials, and data about the relative efficacy of specific interventions are scarce. It may be possible, however, to develop rational treatment guidelines using empiric clinical data.1,2

This article examines the evidence related to 6 controversial aspects of NMS diagnosis and treatment:

  • most-reliable risk factors
  • NMS as a spectrum disorder
  • what causes NMS
  • NMS triggered by first-generation vs second-generation antipsychotics
  • first-line interventions
  • restarting antipsychotics after an NMS episode.

1. Are there reliable risk factors for NMS?

In small case-controlled studies, agitation, dehydration, and exhaustion were the most consistently found systemic factors believed to predispose patients taking antipsychotics to NMS (Table 1).3-5 Catatonia and organic brain syndromes may be separate risk factors.1,6

Preliminary studies also have implicated dopamine receptor abnormalities caused by genetic polymorphisms or effects of low serum iron.1,7,8 Pharmacologic studies have suggested that higher doses, rapid titration, and IM injections of antipsychotics are associated with increased NMS risk.3,5 Some studies suggest that 15% to 20% of NMS patients have a history of NMS episodes.1,2 In addition, high-potency first-generation antipsychotics (FGAs)—especially haloperidol—are assumed to carry higher risk than low-potency drugs and second-generation antipsychotics (SGAs), although this hypothesis remains difficult to prove.9-11

These risk factors, however, are not practical for estimating NMS risk in a given patient because they are relatively common compared with the low risk of NMS occurrence. For the vast majority of patients with psychotic symptoms, the benefits of properly indicated antipsychotic pharmacotherapy will outweigh the risks.

Table 1

Systemic
Agitation
Dehydration
Exhaustion
Low serum iron concentrations (normal: 60 to 170 mcg/dL)
Diagnoses
History of NMS
Catatonia
Organic brain syndromes
Central nervous system
Dopamine receptor dysfunction
Basal ganglia dysfunction
Sympathetic nervous system dysfunction
Pharmacologic treatment*
Intramuscular or intravenous injections
High-potency dopamine antagonists
Rapid dose titration
High doses
FGAs compared with SGAs (?)

*For individual patients, these common risk factors must be weighted again the benefits of antipsychotic therapy FGAs: first-generation antipsychotics; SGAs:second-generation antipsychotics; NMS: neuroleptic malignant syndromeSource: References 1-5

2. Is NMS related to parkinsonism, catatonia, or malignant hyperthermia?

Parkinsonsim. Some researchers have described NMS as an extreme parkinsonian crisis resulting from overwhelming blockade of dopamine pathways in the brain.1,2,12 In this view, NMS resembles the parkinsonian-hyperthermia syndrome that can occur in Parkinson's disease patients following abrupt discontinuation or loss of efficacy of dopaminergic therapy, which can be treated by reinstituting dopaminergic agents.13 Evidence to support this view includes:

  • Parkinsonian signs are a cardinal feature of NMS.
  • Withdrawal of dopamine agonists precipitates the syndrome.
  • All triggering drugs are dopamine receptor antagonists.
  • Risks of NMS correlates with drugs' dopamine receptor affinity.
  • Dopaminergic agonists may be an effective treatment.
  • Lesions in dopaminergic pathways produce a similar syndrome.
  • Patients with NMS have demonstrated low cerebrospinal fluid concentrations of the dopamine metabolite homovanillic acid.14

Catatonia. Fink et al15 and others16-18 have persuasively argued that NMS represents a form of drug-induced malignant catatonia. Evidence supporting this includes:

  • The 2 disorders share neuropsychiatric symptoms.
  • Catalonic signs are common in NMS.19
  • Malignant catatonia and NMS share physiologic and labratory signs.20
  • Reintroduction of antipsychotics can acutely worsen both conditions.
  • Benzodiazepines and electroconvulsive therapy (ECT) are effective treatments for both disorders.15-18

Lee21 examined the relationship between catatonic features and treatment response of 14 NMS patients. Most patients with catatonic symptoms responded to benzodiazepines, whereas none of those did who had an extrapyramidal-hyperthermic presentation without catatonia. Lee concluded that NMS is heterogeneous and may occur in catatonic and noncatatonic forms that differ in treatment response.

Malignant hyperthermia. Some clinicians have compared NMS with malignant hyperthermia caused by inhalational anesthetics and succinylcholine.1,2 Evidence includes

  • similar clinical signs of rigidity, hyperthermia, and hypermetabolism
  • similar psychologic and labratory signs, such as rhabdomyolysis
  • hyperthermia in both responding to dantrolene.

Although the 2 are similar in presentation, malignant hyperthermia occurs intraoperatively and reflects a pharmacogenetic disorder of calcium regulation in skeletal muscle. Additionally, rigidity in malignant hyperthermia does not respond to peripheral-acting muscle relaxants.1,22 Evidence suggests that patients who have previously experienced an NMS episodes are not at risk for malignant hyperthermia.22

3. What is the pathophysiology of NMS?

NMS pathophysiology is complex and likely involves interplay between multiple central and systemic pathways and neurotransmitters. As described above, compelling evidence suggests that dopamine blockade plays a central role.12

Dopamine blockade in the hypothalamus is believed to contribute to thermoregulatory failure, and blockade in the nigrostriatal system likely contributes to muscle rigidity and hypermetabolism. The loss of dopaminergic input to the anterior cingulate-medial orbitofrontal circuit and the lateral orbitofrontal circuit likely con-tributes to the mental status changes and catatonic features seen in NMS.12

 

 

Some researchers have proposed competing or complementary hypotheses, however. For example, Gurrera23 proposed that patients who are prone to developing NMS have a vulnerability to a hyperactive and dysregulated sympathetic nervous system, and this trait—together with dopamine system disruption induced by dopamine-blocking agents—produces NMS. Other investigators have implicated serotonin, norepinephrine, gamma-aminobutyric acid and glutaminergic mechanisms.1,12,24,25

4. Are FGAs or SGAs more likely to cause NMS?

NMS is assumed to occur less frequently in patients treated with SGAs than in those receiving FGAs, although this hypothesisis unproven. Isolated reports of NMS have been associated with nearly every SGA.9-11 It is difficult to prove FGA vs SGA liabilities because:

  • NMS is rare.
  • Dosing practices may be more conser-vative now than in the past.
  • Most clinicians are aware of the earlysigns of NMS.

In an epidemiological study of a large database, Stubner et al26 found that patients receiving SGAs had a lower risk of NMS than those treated with haloperidol.26 In this study, the overall rate of NMS was 0.02%.

NMS hotline data. We recently examined which medication classes were implicated in 111 NMS cases reported to the Neuroleptic Malignant Syndrome Information Service hotline (1-888-NMS-TEMP) between 1997 and 2006 (Figure). We included only cases of definite or probable NMS (as diagnosed by hotline consultants) in which a single antipsychotic was administered. Slightly more cases were attributed to FGAs (51%) than SGAs (45%). The remaining cases were attributed to neuroleptics used in medical settings (such as promethazineor prochlorperazine). Because they are now prescribed less often, FGAs accounted for a disproportionate number of NMS cases reported to the hotline. Haloperidol accounted for the majority of FGA cases and 44% of all cases. If we had excluded haloperidol and compared the NMS risk of SGAs to only intermediate- or low-potency FGAs, the relative advantage of SGAs would have been lost. On the other hand, it is clear that SGAs still carry a risk for NMS. Analyses suggest that the SGA-associated classic features of NMS—fever, muscle rigidity, and autonomic and mental status changes—are retained in patients receiving SGAs, although some may not develop the severe rigidity and extreme temperatures common in patients receiving FGAs.9-11 The milder clinical characteristics associated with SGAs may reflect more conservative prescribing patterns or increased awareness and earlier recognition of NMS, which would prevent fulminant presentations.

5. What is the evidence for specific NMS treatments?

NMS is rare, its presentation varies, and its progression is unpredictable. These factors make it difficult to evaluate treatments in controlled clinical trials, and data about the relative efficacy of specific interventions are scarce.

Even so, the notion that NMS represents an extreme variant of drug-induced parkinsonism or catatonia suggests that specific NMS treatments could be based on symptom severity or stage of presentation. We propose a treatment guideline basedon theoretical mechanisms and anecdotal data (Algorithm).2,27-29

Support. After immediate withdrawal of the offending medication, supportive therapy is the cornerstone of NMS treatment.1,2,27

For patients presenting with mild signs and symptoms, supportive care and careful clinical monitoring may be sufficient. Extreme hyperthermia demands volume resuscitation and cooling measures, intensive medical care, and careful monitoring for complications.

Treatment. Despite a lack of consensus on drug treatments for uncomplicated NMS, approximately 40% of patients with acute NMS receive pharmacologic treatments.2

Lorazepam, 1 to 2 mg parenterally, is a reasonable first-line therapy for NMS, especially in individuals with catatonic features.4,15-18,21,30,31 Some investigators recommend higher doses.15 Benzodiazepines are preferred if sedation is required in agitated NMS patients.4,15-18

Dopaminergic agents such as bromocriptine and amantadine enhance dopaminergic transmission to reverse parkinsonian symptoms and have been reported to reduce time to recovery and halve mortality rates when used alone or in conjunction with other treatments.13,27,32,33 Rapid discontinuation of these agents can result in rebound symptoms, although this may be true for any specific drug treatment of NMS.1,31,32

Dantrolene uncouples excitation-contraction coupling by enhancing calcium sequestration in sarcoplasmic reticulumin skeletal muscle and has been used to treat NMS hypermetabolic symptoms. Some reviews found improvement in up to 80% of NMS patients treated with dantrolene monotherapy.27,32-35 Compared with supportive care, time to recovery may be reduced—and mortality decreased by almost one-half—when dantrolene is used alone or in combination with other medications.

Not all case reports have shown that dantrolene, benzodiazepines, ordopaminergic agonists are effective in treating NMS.31,36 In our opinion, only advanced NMS cases—with extreme temperature elevations, severe rigidity, and evidence of systemic hypermetabolism—benefit from dantrolene treatment.1,2

ECT has been used successfully to reduce mortality from NMS and other catatonic-spectrum disorders. It is usually employed after supportive therapy and psychopharmacologic interventions fail.2,15,16,27,37 ECT for acute NMS typically consists of a series of 6 to 10 treatments with bilateral electrode placement. Daily ECT may be needed initially.15

 

 

6. Are antipsychotics contraindicated following an NMS episode?

The rate of NMS recurrence on retreatment with an antipsychotic has varied.38 We estimate that up to 30% of patients may be at risk of NMS recurrence when rechallenged with an antipsychotic.1 By following proper precautions (Table 2), however, you can safely treat most patients who require continued antipsychotic therapy.1,2 When you restart treatment, a lower-potency antipsychotic from a different chemical class may be a safer option than retrying the triggering agent, according to retrospective analyses of limited available data. A patient who develops NMS on a FGA might benefit from an SGA trial, although some risk of recurrence remains.1,10

Current Psychiatry 2007;6(8):89-95.
Drug brand names

  • Amantadine • Symmetrel
  • Bromocriptine • Parlodel
  • Chlorpromazine • Thorazine
  • Dantrolene • Dantrium
  • Fluphenazine • Prolixin
  • Haloperidol • Haldol
  • Lorazepam • Ativan
  • Loxapine • Loxitane
  • Perphenazine • Trilafon
  • Prochlorperazine • Compazine, Compro
  • Promethazine • Phenergan
  • Thioridazine • Mellaril

Disclosure

Dr. Strawn is an American Psychiatric Institute for Research and Education (APIRE)/Janssen Scholar.

Dr. Keck has received research support from or served as a consultant to Abbott Laboratories, American Diabetes Association, AstraZeneca Pharmaceuticals, Bristol-Myers Squibb, GlaxoSmithKline, Eli Lilly and Company, Janssen Pharmaceutica, National Institute of Mental Health, National Institute of Drug Abuse, Pfizer, Stanley Medical Research Institute, and UCB Pharma.

Dr. Caroff has received research support from Bristol-Myers Squibb, Ortho-McNeil Neurologics, and Pfizer.

Diagnosis and treatment of neuroleptic malignant syndrome (NMS) are controversial because this potentially life-threatening syndrome is rare and its presentation varies. These factors make it difficult to evaluate treatments in controlled clinical trials, and data about the relative efficacy of specific interventions are scarce. It may be possible, however, to develop rational treatment guidelines using empiric clinical data.1,2

This article examines the evidence related to 6 controversial aspects of NMS diagnosis and treatment:

  • most-reliable risk factors
  • NMS as a spectrum disorder
  • what causes NMS
  • NMS triggered by first-generation vs second-generation antipsychotics
  • first-line interventions
  • restarting antipsychotics after an NMS episode.

1. Are there reliable risk factors for NMS?

In small case-controlled studies, agitation, dehydration, and exhaustion were the most consistently found systemic factors believed to predispose patients taking antipsychotics to NMS (Table 1).3-5 Catatonia and organic brain syndromes may be separate risk factors.1,6

Preliminary studies also have implicated dopamine receptor abnormalities caused by genetic polymorphisms or effects of low serum iron.1,7,8 Pharmacologic studies have suggested that higher doses, rapid titration, and IM injections of antipsychotics are associated with increased NMS risk.3,5 Some studies suggest that 15% to 20% of NMS patients have a history of NMS episodes.1,2 In addition, high-potency first-generation antipsychotics (FGAs)—especially haloperidol—are assumed to carry higher risk than low-potency drugs and second-generation antipsychotics (SGAs), although this hypothesis remains difficult to prove.9-11

These risk factors, however, are not practical for estimating NMS risk in a given patient because they are relatively common compared with the low risk of NMS occurrence. For the vast majority of patients with psychotic symptoms, the benefits of properly indicated antipsychotic pharmacotherapy will outweigh the risks.

Table 1

Systemic
Agitation
Dehydration
Exhaustion
Low serum iron concentrations (normal: 60 to 170 mcg/dL)
Diagnoses
History of NMS
Catatonia
Organic brain syndromes
Central nervous system
Dopamine receptor dysfunction
Basal ganglia dysfunction
Sympathetic nervous system dysfunction
Pharmacologic treatment*
Intramuscular or intravenous injections
High-potency dopamine antagonists
Rapid dose titration
High doses
FGAs compared with SGAs (?)

*For individual patients, these common risk factors must be weighted again the benefits of antipsychotic therapy FGAs: first-generation antipsychotics; SGAs:second-generation antipsychotics; NMS: neuroleptic malignant syndromeSource: References 1-5

2. Is NMS related to parkinsonism, catatonia, or malignant hyperthermia?

Parkinsonsim. Some researchers have described NMS as an extreme parkinsonian crisis resulting from overwhelming blockade of dopamine pathways in the brain.1,2,12 In this view, NMS resembles the parkinsonian-hyperthermia syndrome that can occur in Parkinson's disease patients following abrupt discontinuation or loss of efficacy of dopaminergic therapy, which can be treated by reinstituting dopaminergic agents.13 Evidence to support this view includes:

  • Parkinsonian signs are a cardinal feature of NMS.
  • Withdrawal of dopamine agonists precipitates the syndrome.
  • All triggering drugs are dopamine receptor antagonists.
  • Risks of NMS correlates with drugs' dopamine receptor affinity.
  • Dopaminergic agonists may be an effective treatment.
  • Lesions in dopaminergic pathways produce a similar syndrome.
  • Patients with NMS have demonstrated low cerebrospinal fluid concentrations of the dopamine metabolite homovanillic acid.14

Catatonia. Fink et al15 and others16-18 have persuasively argued that NMS represents a form of drug-induced malignant catatonia. Evidence supporting this includes:

  • The 2 disorders share neuropsychiatric symptoms.
  • Catalonic signs are common in NMS.19
  • Malignant catatonia and NMS share physiologic and labratory signs.20
  • Reintroduction of antipsychotics can acutely worsen both conditions.
  • Benzodiazepines and electroconvulsive therapy (ECT) are effective treatments for both disorders.15-18

Lee21 examined the relationship between catatonic features and treatment response of 14 NMS patients. Most patients with catatonic symptoms responded to benzodiazepines, whereas none of those did who had an extrapyramidal-hyperthermic presentation without catatonia. Lee concluded that NMS is heterogeneous and may occur in catatonic and noncatatonic forms that differ in treatment response.

Malignant hyperthermia. Some clinicians have compared NMS with malignant hyperthermia caused by inhalational anesthetics and succinylcholine.1,2 Evidence includes

  • similar clinical signs of rigidity, hyperthermia, and hypermetabolism
  • similar psychologic and labratory signs, such as rhabdomyolysis
  • hyperthermia in both responding to dantrolene.

Although the 2 are similar in presentation, malignant hyperthermia occurs intraoperatively and reflects a pharmacogenetic disorder of calcium regulation in skeletal muscle. Additionally, rigidity in malignant hyperthermia does not respond to peripheral-acting muscle relaxants.1,22 Evidence suggests that patients who have previously experienced an NMS episodes are not at risk for malignant hyperthermia.22

3. What is the pathophysiology of NMS?

NMS pathophysiology is complex and likely involves interplay between multiple central and systemic pathways and neurotransmitters. As described above, compelling evidence suggests that dopamine blockade plays a central role.12

Dopamine blockade in the hypothalamus is believed to contribute to thermoregulatory failure, and blockade in the nigrostriatal system likely contributes to muscle rigidity and hypermetabolism. The loss of dopaminergic input to the anterior cingulate-medial orbitofrontal circuit and the lateral orbitofrontal circuit likely con-tributes to the mental status changes and catatonic features seen in NMS.12

 

 

Some researchers have proposed competing or complementary hypotheses, however. For example, Gurrera23 proposed that patients who are prone to developing NMS have a vulnerability to a hyperactive and dysregulated sympathetic nervous system, and this trait—together with dopamine system disruption induced by dopamine-blocking agents—produces NMS. Other investigators have implicated serotonin, norepinephrine, gamma-aminobutyric acid and glutaminergic mechanisms.1,12,24,25

4. Are FGAs or SGAs more likely to cause NMS?

NMS is assumed to occur less frequently in patients treated with SGAs than in those receiving FGAs, although this hypothesisis unproven. Isolated reports of NMS have been associated with nearly every SGA.9-11 It is difficult to prove FGA vs SGA liabilities because:

  • NMS is rare.
  • Dosing practices may be more conser-vative now than in the past.
  • Most clinicians are aware of the earlysigns of NMS.

In an epidemiological study of a large database, Stubner et al26 found that patients receiving SGAs had a lower risk of NMS than those treated with haloperidol.26 In this study, the overall rate of NMS was 0.02%.

NMS hotline data. We recently examined which medication classes were implicated in 111 NMS cases reported to the Neuroleptic Malignant Syndrome Information Service hotline (1-888-NMS-TEMP) between 1997 and 2006 (Figure). We included only cases of definite or probable NMS (as diagnosed by hotline consultants) in which a single antipsychotic was administered. Slightly more cases were attributed to FGAs (51%) than SGAs (45%). The remaining cases were attributed to neuroleptics used in medical settings (such as promethazineor prochlorperazine). Because they are now prescribed less often, FGAs accounted for a disproportionate number of NMS cases reported to the hotline. Haloperidol accounted for the majority of FGA cases and 44% of all cases. If we had excluded haloperidol and compared the NMS risk of SGAs to only intermediate- or low-potency FGAs, the relative advantage of SGAs would have been lost. On the other hand, it is clear that SGAs still carry a risk for NMS. Analyses suggest that the SGA-associated classic features of NMS—fever, muscle rigidity, and autonomic and mental status changes—are retained in patients receiving SGAs, although some may not develop the severe rigidity and extreme temperatures common in patients receiving FGAs.9-11 The milder clinical characteristics associated with SGAs may reflect more conservative prescribing patterns or increased awareness and earlier recognition of NMS, which would prevent fulminant presentations.

5. What is the evidence for specific NMS treatments?

NMS is rare, its presentation varies, and its progression is unpredictable. These factors make it difficult to evaluate treatments in controlled clinical trials, and data about the relative efficacy of specific interventions are scarce.

Even so, the notion that NMS represents an extreme variant of drug-induced parkinsonism or catatonia suggests that specific NMS treatments could be based on symptom severity or stage of presentation. We propose a treatment guideline basedon theoretical mechanisms and anecdotal data (Algorithm).2,27-29

Support. After immediate withdrawal of the offending medication, supportive therapy is the cornerstone of NMS treatment.1,2,27

For patients presenting with mild signs and symptoms, supportive care and careful clinical monitoring may be sufficient. Extreme hyperthermia demands volume resuscitation and cooling measures, intensive medical care, and careful monitoring for complications.

Treatment. Despite a lack of consensus on drug treatments for uncomplicated NMS, approximately 40% of patients with acute NMS receive pharmacologic treatments.2

Lorazepam, 1 to 2 mg parenterally, is a reasonable first-line therapy for NMS, especially in individuals with catatonic features.4,15-18,21,30,31 Some investigators recommend higher doses.15 Benzodiazepines are preferred if sedation is required in agitated NMS patients.4,15-18

Dopaminergic agents such as bromocriptine and amantadine enhance dopaminergic transmission to reverse parkinsonian symptoms and have been reported to reduce time to recovery and halve mortality rates when used alone or in conjunction with other treatments.13,27,32,33 Rapid discontinuation of these agents can result in rebound symptoms, although this may be true for any specific drug treatment of NMS.1,31,32

Dantrolene uncouples excitation-contraction coupling by enhancing calcium sequestration in sarcoplasmic reticulumin skeletal muscle and has been used to treat NMS hypermetabolic symptoms. Some reviews found improvement in up to 80% of NMS patients treated with dantrolene monotherapy.27,32-35 Compared with supportive care, time to recovery may be reduced—and mortality decreased by almost one-half—when dantrolene is used alone or in combination with other medications.

Not all case reports have shown that dantrolene, benzodiazepines, ordopaminergic agonists are effective in treating NMS.31,36 In our opinion, only advanced NMS cases—with extreme temperature elevations, severe rigidity, and evidence of systemic hypermetabolism—benefit from dantrolene treatment.1,2

ECT has been used successfully to reduce mortality from NMS and other catatonic-spectrum disorders. It is usually employed after supportive therapy and psychopharmacologic interventions fail.2,15,16,27,37 ECT for acute NMS typically consists of a series of 6 to 10 treatments with bilateral electrode placement. Daily ECT may be needed initially.15

 

 

6. Are antipsychotics contraindicated following an NMS episode?

The rate of NMS recurrence on retreatment with an antipsychotic has varied.38 We estimate that up to 30% of patients may be at risk of NMS recurrence when rechallenged with an antipsychotic.1 By following proper precautions (Table 2), however, you can safely treat most patients who require continued antipsychotic therapy.1,2 When you restart treatment, a lower-potency antipsychotic from a different chemical class may be a safer option than retrying the triggering agent, according to retrospective analyses of limited available data. A patient who develops NMS on a FGA might benefit from an SGA trial, although some risk of recurrence remains.1,10

Current Psychiatry 2007;6(8):89-95.
Drug brand names

  • Amantadine • Symmetrel
  • Bromocriptine • Parlodel
  • Chlorpromazine • Thorazine
  • Dantrolene • Dantrium
  • Fluphenazine • Prolixin
  • Haloperidol • Haldol
  • Lorazepam • Ativan
  • Loxapine • Loxitane
  • Perphenazine • Trilafon
  • Prochlorperazine • Compazine, Compro
  • Promethazine • Phenergan
  • Thioridazine • Mellaril

Disclosure

Dr. Strawn is an American Psychiatric Institute for Research and Education (APIRE)/Janssen Scholar.

Dr. Keck has received research support from or served as a consultant to Abbott Laboratories, American Diabetes Association, AstraZeneca Pharmaceuticals, Bristol-Myers Squibb, GlaxoSmithKline, Eli Lilly and Company, Janssen Pharmaceutica, National Institute of Mental Health, National Institute of Drug Abuse, Pfizer, Stanley Medical Research Institute, and UCB Pharma.

Dr. Caroff has received research support from Bristol-Myers Squibb, Ortho-McNeil Neurologics, and Pfizer.

References

1. Caroff SN. Neuroleptic malignant syndrome. In: Mann SC, Caroff SN, Keck PE Jr, Lazarus A, eds. Neuroleptic malignant syndrome and related conditions 2nd ed. Washington, DC: American Psychiatric Publishing Inc; 2003; 1-44.

2. Strawn JR, Keck PE, Jr, Caroff SN. Neuroleptic malignant syndrome Am J Psychiatry 2007;164:870-6.

3. Keck PE, Jr, Pope HG, Jr, Cohen BM, et al. Risk factors for neuroleptic malignant syndrome Arch Gen Psychiatry 1989;46:914-18.

4. Rosebush PI, Stewart TD. A prospective analysis of 24 episodes of neuroleptic malignant syndrome Am J Psychiatry 1989;146:717-25.

5. Berardi D, Amore M, Keck PE, Jr, et al. Clinical and pharmacologic risk factors for neuroleptic malignant syndrome: a case-control study. Biol Psychiatry 1998;44:748-54.

6. White DA, Robins AH. Catatonia: harbinger of the neuroleptic malignant syndrome Br J Psychiatry 1991;158:419-21.

7. Rosebush PI, Mazurek MF. Serum iron and neuroleptic malignant syndrome. Lancet 1991;338:149-51.

8. Lee JW. Serum iron in catatonia and neuroleptic malignant syndrome Biol Psychiatry 1998;44:499-507.

9. Ananth J, Parameswaran S, Gunatilake S, et al. Neuroleptic malignant syndrome and atypical antipsychotic drugs J Clin Psychiatry 2004;65:464-70.

10. Caroff SN, Mann SC, Campbell EC. Atypical antipsychotics and neuroleptic malignant syndrome Psychiatr Ann 2000;30:314-21.

11. Hasan S, Buckley P. Novel antipsychotics and the neuroleptic malignant syndrome Am J Psychiatry 1998;155:1113-16.

12. Mann SC, Caroff SN, Fricchione G, Campbell EC. Central dopamine hypoactivity and the pathogenesis of neuroleptic malignant syndrome Psychiatr Ann 2000;30:363-74.

13. Factor SA, Santiago A. Parkinsonism-hyperpyrexia syndrome in Parkinson’s disease. In: Frucht SJ, Fahn S, eds. Movement disorder emergencies: diagnosis and treatment. Totowa, NJ: Humana Press; 2005; 29-40.

14. Nisijima K, Ishiguro T. Cerebrospinal fluid levels of monoamine metabolites and gamma-aminobutyric acid in neuroleptic malignant syndrome. J Psychiatr Res 1995;27:233-44.

15. Fink M, Taylor MA. Neuroleptic malignant syndrome is malignant catatonia, warranting treatments efficacious for catatonia. Prog Neuropsychopharmacol Biol Psychiatry 2006;30:1182-3.

16. Fricchione G, Bush G, Fozdar M, et al. Recognition and treatment of the catatonic syndrome. J Intensive Care Med 1997;12:135-47.

17. Philbrick KL, Rummans TA. Malignant catatonia. J Neuropsychiatry Clin Neurosci 1994;6:1-13.

18. Mann SC, Caroff SN, Bleier HR, et al. Lethal catatonia. Am J Psychiatry 1986;143:1374-81.

19. Koch M, Chandragiri S, Rizvi S, et al. Catatonic signs in neuroleptic malignant syndrome. Compr Psychiatry 2000;41:73-5.

20. Lee JW. Laboratory findings. In: Caroff SN, Mann SC, Francis A, Fricchoine GL, eds. Catatonia: from psychopathology to neurobiology Washington, DC: American Psychiatric Press, Inc; 2004; 65-75.

21. Lee JW. Catatonic variants, hyperthermic extrapyramidal reactions, and subtypes of neuroleptic malignant syndrome. Ann Clin Psychiatry 2007;19:9-16.

22. Caroff SN, Rosenberg H, Mann SC, et al. Neuroleptic malignant syndrome in the perioperative setting. Am J Anesthesiol 2001;28:387-93.

23. Gurrera RJ. Sympathoadrenal hyperactivity and the etiology of neuroleptic malignant syndrome. Am J Psychiatry 1999;156:169-80.

24. Carroll BT. The universal field hypothesis of catatonia and neuroleptic malignant syndrome. CNS Spectr 2000;5:26-33.

25. Weller M, Kornhuber J. A rationale for NMDA receptor antagonist therapy of the neuroleptic malignant syndrome. Med Hypotheses 1992;38:329-33.

26. Stubner S, Rustenbeck E, Grohmann R, et al. Severe and uncommon involuntary movement disorders due to psychotropic drugs. Pharmacopsychiatry 2004;37(suppl 1):S54-S64.

27. Davis JM, Caroff SN, Mann SC. Treatment of neuroleptic malignant syndrome. Psychiatr Ann 2000;30:325-31.

28. Adityanjee PA, Singh S, Singh G, Ong S. Spectrum concept of neuroleptic malignant syndrome. Br J Psychiatry 1988;153:107-11.

29. Woodbury MM, Woodbury MA. Neuroleptic-induced catatonia as a stage in the progression toward neuroleptic malignant syndrome. J Am Acad Child Adolesc Psychiatry 1992;31:1161-4.

30. Francis A, Chondragivi S, Rizvi S, et al. Is lorazepam a treatment for neuroleptic malignant syndrome? CNS Spectr 2000;5:54-7.

31. Rosebush PI, Stewart T, Mazurek MF. The treatment of neuroleptic malignant syndrome. Are dantrolene and bromocriptine useful adjuncts to supportive care? Br J Psychiatry 1991;159:709-12.

32. Sakkas P, Davis JM, Janicak PG, Wang ZY. Drug treatment of the neuroleptic malignant syndrome. Psychopharmacol Bull 1991;27:381-4.

33. Rosenberg MR, Green M. Neuroleptic malignant syndrome: review of response to therapy. Arch Intern Med 1989;149:1927-31.

34. Yamawaki S, Morio M, Kazamutsuri G, et al. Clinical evaluation and effective usage of dantrolene sodium in neuroleptic malignant syndrome. Kiso to Rinsyou (Clinical Reports) 1993;27:1045-66.

35. Tsutsumi Y, Yamamoto K, Matsuura S, et al. The treatment of neuroleptic malignant syndrome using dantrolene sodium. Psychiatry Clin Neurosci 1998;52:433-8.

36. Reulbach U, Dutsch C, Biermann T, et al. Managing an effective treatment for neuroleptic malignant syndrome. Crit Care 2007;11:R4.-

37. Troller JN, Sachdev PS. Electroconvulsive treatment of neuroleptic malignant syndrome: a review and report of cases. Aust N Z J Psychiatry 1999;33:650-9.

38. Pope HG, Aizley HG, Keck PE, Jr, McElroy SL. Neuroleptic malignant syndrome: long term follow-up of 20 cases. J Clin Psychiatry 1991;52:208-12.

References

1. Caroff SN. Neuroleptic malignant syndrome. In: Mann SC, Caroff SN, Keck PE Jr, Lazarus A, eds. Neuroleptic malignant syndrome and related conditions 2nd ed. Washington, DC: American Psychiatric Publishing Inc; 2003; 1-44.

2. Strawn JR, Keck PE, Jr, Caroff SN. Neuroleptic malignant syndrome Am J Psychiatry 2007;164:870-6.

3. Keck PE, Jr, Pope HG, Jr, Cohen BM, et al. Risk factors for neuroleptic malignant syndrome Arch Gen Psychiatry 1989;46:914-18.

4. Rosebush PI, Stewart TD. A prospective analysis of 24 episodes of neuroleptic malignant syndrome Am J Psychiatry 1989;146:717-25.

5. Berardi D, Amore M, Keck PE, Jr, et al. Clinical and pharmacologic risk factors for neuroleptic malignant syndrome: a case-control study. Biol Psychiatry 1998;44:748-54.

6. White DA, Robins AH. Catatonia: harbinger of the neuroleptic malignant syndrome Br J Psychiatry 1991;158:419-21.

7. Rosebush PI, Mazurek MF. Serum iron and neuroleptic malignant syndrome. Lancet 1991;338:149-51.

8. Lee JW. Serum iron in catatonia and neuroleptic malignant syndrome Biol Psychiatry 1998;44:499-507.

9. Ananth J, Parameswaran S, Gunatilake S, et al. Neuroleptic malignant syndrome and atypical antipsychotic drugs J Clin Psychiatry 2004;65:464-70.

10. Caroff SN, Mann SC, Campbell EC. Atypical antipsychotics and neuroleptic malignant syndrome Psychiatr Ann 2000;30:314-21.

11. Hasan S, Buckley P. Novel antipsychotics and the neuroleptic malignant syndrome Am J Psychiatry 1998;155:1113-16.

12. Mann SC, Caroff SN, Fricchione G, Campbell EC. Central dopamine hypoactivity and the pathogenesis of neuroleptic malignant syndrome Psychiatr Ann 2000;30:363-74.

13. Factor SA, Santiago A. Parkinsonism-hyperpyrexia syndrome in Parkinson’s disease. In: Frucht SJ, Fahn S, eds. Movement disorder emergencies: diagnosis and treatment. Totowa, NJ: Humana Press; 2005; 29-40.

14. Nisijima K, Ishiguro T. Cerebrospinal fluid levels of monoamine metabolites and gamma-aminobutyric acid in neuroleptic malignant syndrome. J Psychiatr Res 1995;27:233-44.

15. Fink M, Taylor MA. Neuroleptic malignant syndrome is malignant catatonia, warranting treatments efficacious for catatonia. Prog Neuropsychopharmacol Biol Psychiatry 2006;30:1182-3.

16. Fricchione G, Bush G, Fozdar M, et al. Recognition and treatment of the catatonic syndrome. J Intensive Care Med 1997;12:135-47.

17. Philbrick KL, Rummans TA. Malignant catatonia. J Neuropsychiatry Clin Neurosci 1994;6:1-13.

18. Mann SC, Caroff SN, Bleier HR, et al. Lethal catatonia. Am J Psychiatry 1986;143:1374-81.

19. Koch M, Chandragiri S, Rizvi S, et al. Catatonic signs in neuroleptic malignant syndrome. Compr Psychiatry 2000;41:73-5.

20. Lee JW. Laboratory findings. In: Caroff SN, Mann SC, Francis A, Fricchoine GL, eds. Catatonia: from psychopathology to neurobiology Washington, DC: American Psychiatric Press, Inc; 2004; 65-75.

21. Lee JW. Catatonic variants, hyperthermic extrapyramidal reactions, and subtypes of neuroleptic malignant syndrome. Ann Clin Psychiatry 2007;19:9-16.

22. Caroff SN, Rosenberg H, Mann SC, et al. Neuroleptic malignant syndrome in the perioperative setting. Am J Anesthesiol 2001;28:387-93.

23. Gurrera RJ. Sympathoadrenal hyperactivity and the etiology of neuroleptic malignant syndrome. Am J Psychiatry 1999;156:169-80.

24. Carroll BT. The universal field hypothesis of catatonia and neuroleptic malignant syndrome. CNS Spectr 2000;5:26-33.

25. Weller M, Kornhuber J. A rationale for NMDA receptor antagonist therapy of the neuroleptic malignant syndrome. Med Hypotheses 1992;38:329-33.

26. Stubner S, Rustenbeck E, Grohmann R, et al. Severe and uncommon involuntary movement disorders due to psychotropic drugs. Pharmacopsychiatry 2004;37(suppl 1):S54-S64.

27. Davis JM, Caroff SN, Mann SC. Treatment of neuroleptic malignant syndrome. Psychiatr Ann 2000;30:325-31.

28. Adityanjee PA, Singh S, Singh G, Ong S. Spectrum concept of neuroleptic malignant syndrome. Br J Psychiatry 1988;153:107-11.

29. Woodbury MM, Woodbury MA. Neuroleptic-induced catatonia as a stage in the progression toward neuroleptic malignant syndrome. J Am Acad Child Adolesc Psychiatry 1992;31:1161-4.

30. Francis A, Chondragivi S, Rizvi S, et al. Is lorazepam a treatment for neuroleptic malignant syndrome? CNS Spectr 2000;5:54-7.

31. Rosebush PI, Stewart T, Mazurek MF. The treatment of neuroleptic malignant syndrome. Are dantrolene and bromocriptine useful adjuncts to supportive care? Br J Psychiatry 1991;159:709-12.

32. Sakkas P, Davis JM, Janicak PG, Wang ZY. Drug treatment of the neuroleptic malignant syndrome. Psychopharmacol Bull 1991;27:381-4.

33. Rosenberg MR, Green M. Neuroleptic malignant syndrome: review of response to therapy. Arch Intern Med 1989;149:1927-31.

34. Yamawaki S, Morio M, Kazamutsuri G, et al. Clinical evaluation and effective usage of dantrolene sodium in neuroleptic malignant syndrome. Kiso to Rinsyou (Clinical Reports) 1993;27:1045-66.

35. Tsutsumi Y, Yamamoto K, Matsuura S, et al. The treatment of neuroleptic malignant syndrome using dantrolene sodium. Psychiatry Clin Neurosci 1998;52:433-8.

36. Reulbach U, Dutsch C, Biermann T, et al. Managing an effective treatment for neuroleptic malignant syndrome. Crit Care 2007;11:R4.-

37. Troller JN, Sachdev PS. Electroconvulsive treatment of neuroleptic malignant syndrome: a review and report of cases. Aust N Z J Psychiatry 1999;33:650-9.

38. Pope HG, Aizley HG, Keck PE, Jr, McElroy SL. Neuroleptic malignant syndrome: long term follow-up of 20 cases. J Clin Psychiatry 1991;52:208-12.

Issue
Current Psychiatry - 07(01)
Issue
Current Psychiatry - 07(01)
Page Number
95-101
Page Number
95-101
Publications
Publications
Article Type
Display Headline
Neuroleptic malignant syndrome: Answers to 6 tough questions
Display Headline
Neuroleptic malignant syndrome: Answers to 6 tough questions
Legacy Keywords
neuroleptic malignant syndrome; NMS diagnosis; NMS risk factors; first-generation antipsychotics; second-generation antipsychotics; parkinsonism; catatonia; hyperthermia; NMS pathophysiology; Jeffrey R Strawn MD; Paul E Keck Jr MD; Stanley N Caroff MD
Legacy Keywords
neuroleptic malignant syndrome; NMS diagnosis; NMS risk factors; first-generation antipsychotics; second-generation antipsychotics; parkinsonism; catatonia; hyperthermia; NMS pathophysiology; Jeffrey R Strawn MD; Paul E Keck Jr MD; Stanley N Caroff MD
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Commentary: When medical illness complicates schizophrenia and bipolar disorder

Article Type
Changed
Mon, 04/16/2018 - 14:16
Display Headline
Commentary: When medical illness complicates schizophrenia and bipolar disorder

Patients with schizophrenia or bipolar disorder face a higher risk of premature death, compared with the general population. Their overall mortality rate is elevated not only by higher suicide rates but also by higher rates of medical comorbidities, including obesity, diabetes mellitus, cardiovascular and pulmonary diseases, HIV infection, and cancer.1,2

Schizophrenia and bipolar disorder are also frequently complicated by psychiatric comorbidities including alcohol and substance use disorders (such as nicotine dependence), anxiety disorders, and eating disorders (such as binge eating).3,4 Even so, the impact of psychiatric comorbidity on medical morbidity and mortality has not been adequately investigated and has—until rather recently—received little attention from clinical researchers.

Within the past 5 years or so, epidemiologic and population studies have shown increased morbidity and mortality from medical illnesses in patients with serious and persistent mental illness.5,6 These findings have sparked renewed interest in behavioral, biological, and psychosocial factors associated with schizophrenia and bipolar disorder that may contribute to comorbid medical illnesses. These factors include:

  • negative and depressive symptoms
  • physical inactivity and poor diet
  • hypothalamic-pituitary-adrenal axis dysregulation associated with acute psychotic and affective episodes
  • amotivation
  • social isolation
  • limited access to primary and preventive health care.7

The degree to which these factors may elevate medical comorbidity rates in patients with Commentary these serious psychiatric illnesses has not been well-studied or described.

TREATMENT IMPLICATIONS

These observations raise concerns about safe and effective use of medications by patients with schizophrenia or bipolar disorder as well as medical illness. Issues for clinicians to consider include:

  • possible pharmacokinetic and pharmacodynamic interactions in patients taking concomitant medications for psychiatric and medical illnesses
  • potential beneficial and adverse effects of psychotropics on medical illnesses
  • and—conversely—potential beneficial and adverse effects on mood and psychotic disorders of medications used to treat medical illnesses.

Being aware of metabolic effects when prescribing psychotropics is not a new idea. Lithium, for example, has long been known to cause weight gain, suppress thyroid hormone, and interact with thiazide diuretics when used to treat bipolar disorder. Thus, therapeutic blood monitoring is recommended for patients receiving acute and maintenance lithium therapy.

More recently, atypical antipsychotics such as olanzapine and clozapine (and risperidone and quetiapine to a lesser extent) have been shown to produce substantial weight gain and other metabolic effects that increase the risk of diabetes and cardiovascular disease.7 As a result, the American Diabetes Association—along with the American Psychiatric Association and other groups—now recommends that psychiatrists and other physicians monitor patients’ body weight and body mass index, vital signs, serum glucose, and lipids when prescribing these agents.7-11 Careful monitoring should improve the medical care of patients with schizophrenia and bipolar disorder and help protect them from the medical risks associated with overweight and obesity.

Similar precautions are needed when schizophrenia and bipolar disorder co-occur with other medical illnesses treated by complex pharmacologic regimens, such as pulmonary disease, cancer, and HIV. Three points to keep in mind are:

  • careful selection of treatments for co-occurring medical illnesses, considering potential effects on the primary psychiatric disorder
  • the impact of psychotropics on patients’ medical illnesses
  • and the potential for drug interactions.
References

1. Kilbourne AM, Cornelius JR, Han X, et al. Burden of general medical conditions among individuals with bipolar disorder. Bipolar Disord 2004;6:368-73.

2. Meyer JM, Nasrallah HA. Medical illness and schizophrenia Washington, DC: American Psychiatric Press, Inc, 2003.

3. McElroy SL, Altshuler LL, Suppes T, et al. Axis I psychiatric comorbidity and its relationship with historical illness variables in 288 patients with bipolar disorder. Am J Psychiatry 2001;158:420-6.

4. Green AI, Canuso CM, Brenner MJ, et al. Detection and management of comorbidity in patients with schizophrenia. Psychiatr Clin North Am 2003;26:115-39.

5. Buda M, Tsuang MT, Fleming JA. Causes of death in DSM-III schizophrenics and other psychotics (atypical group). Comparison with the general population. Arch Gen Psychiatry 1988;45:283-5.

6. Osby U, Brandt L, Correia N, et al. Excess mortality in bipolar and unipolar disorder in Sweden. Arch Gen Psychiatry 2001;58:844-50.

7. Keck PE, Jr, Buse JB, Dagago-Jack S, et al. Managing metabolic concerns in patients with severe mental illness: a special report (Postgrad Med). Minneapolis, MN: McGraw-Hill, 2003.

8. American Diabetes Association. Consensus Development Conference on Antipsychotic Drugs and Obesity and Diabetes. Diabetes Care 2004;27:596-601.

9. Marder SR, Essock SM, Miller AL, et al. Physical health monitoring of patients with schizophrenia. Am J Psychiatry 2004;161:1334-9.

10. Chue P, Kovacs CS. Safety and tolerability of atypical antipsychotics in patients with bipolar disorder: prevalence, monitoring and management. Bipolar Disord 2003;5(suppl 2):62-79.

11. Nasrallah HA, Newcomer JW. Atypical antipsychotics and metabolic dysregulation. Evaluating the risk/benefit equation and improving standard of care. J Clin Psychopharmacol 2004;24(suppl 2):S7-S14.

Author and Disclosure Information

Paul E. Keck, Jr, MD
Professor and vice chairman for research Department of psychiatry, University of Cincinnati Deputy Editor, CURRENTPSYCHIATRY

Henry A. Nasrallah, MD
Professor of psychiatry, neurology, and neuroscience Associate dean, department of psychiatry University of Cincinnati College of Medicine, Cincinnati, OH

Issue
Current Psychiatry - 03(12)
Publications
Topics
Page Number
41-42
Sections
Author and Disclosure Information

Paul E. Keck, Jr, MD
Professor and vice chairman for research Department of psychiatry, University of Cincinnati Deputy Editor, CURRENTPSYCHIATRY

Henry A. Nasrallah, MD
Professor of psychiatry, neurology, and neuroscience Associate dean, department of psychiatry University of Cincinnati College of Medicine, Cincinnati, OH

Author and Disclosure Information

Paul E. Keck, Jr, MD
Professor and vice chairman for research Department of psychiatry, University of Cincinnati Deputy Editor, CURRENTPSYCHIATRY

Henry A. Nasrallah, MD
Professor of psychiatry, neurology, and neuroscience Associate dean, department of psychiatry University of Cincinnati College of Medicine, Cincinnati, OH

Patients with schizophrenia or bipolar disorder face a higher risk of premature death, compared with the general population. Their overall mortality rate is elevated not only by higher suicide rates but also by higher rates of medical comorbidities, including obesity, diabetes mellitus, cardiovascular and pulmonary diseases, HIV infection, and cancer.1,2

Schizophrenia and bipolar disorder are also frequently complicated by psychiatric comorbidities including alcohol and substance use disorders (such as nicotine dependence), anxiety disorders, and eating disorders (such as binge eating).3,4 Even so, the impact of psychiatric comorbidity on medical morbidity and mortality has not been adequately investigated and has—until rather recently—received little attention from clinical researchers.

Within the past 5 years or so, epidemiologic and population studies have shown increased morbidity and mortality from medical illnesses in patients with serious and persistent mental illness.5,6 These findings have sparked renewed interest in behavioral, biological, and psychosocial factors associated with schizophrenia and bipolar disorder that may contribute to comorbid medical illnesses. These factors include:

  • negative and depressive symptoms
  • physical inactivity and poor diet
  • hypothalamic-pituitary-adrenal axis dysregulation associated with acute psychotic and affective episodes
  • amotivation
  • social isolation
  • limited access to primary and preventive health care.7

The degree to which these factors may elevate medical comorbidity rates in patients with Commentary these serious psychiatric illnesses has not been well-studied or described.

TREATMENT IMPLICATIONS

These observations raise concerns about safe and effective use of medications by patients with schizophrenia or bipolar disorder as well as medical illness. Issues for clinicians to consider include:

  • possible pharmacokinetic and pharmacodynamic interactions in patients taking concomitant medications for psychiatric and medical illnesses
  • potential beneficial and adverse effects of psychotropics on medical illnesses
  • and—conversely—potential beneficial and adverse effects on mood and psychotic disorders of medications used to treat medical illnesses.

Being aware of metabolic effects when prescribing psychotropics is not a new idea. Lithium, for example, has long been known to cause weight gain, suppress thyroid hormone, and interact with thiazide diuretics when used to treat bipolar disorder. Thus, therapeutic blood monitoring is recommended for patients receiving acute and maintenance lithium therapy.

More recently, atypical antipsychotics such as olanzapine and clozapine (and risperidone and quetiapine to a lesser extent) have been shown to produce substantial weight gain and other metabolic effects that increase the risk of diabetes and cardiovascular disease.7 As a result, the American Diabetes Association—along with the American Psychiatric Association and other groups—now recommends that psychiatrists and other physicians monitor patients’ body weight and body mass index, vital signs, serum glucose, and lipids when prescribing these agents.7-11 Careful monitoring should improve the medical care of patients with schizophrenia and bipolar disorder and help protect them from the medical risks associated with overweight and obesity.

Similar precautions are needed when schizophrenia and bipolar disorder co-occur with other medical illnesses treated by complex pharmacologic regimens, such as pulmonary disease, cancer, and HIV. Three points to keep in mind are:

  • careful selection of treatments for co-occurring medical illnesses, considering potential effects on the primary psychiatric disorder
  • the impact of psychotropics on patients’ medical illnesses
  • and the potential for drug interactions.

Patients with schizophrenia or bipolar disorder face a higher risk of premature death, compared with the general population. Their overall mortality rate is elevated not only by higher suicide rates but also by higher rates of medical comorbidities, including obesity, diabetes mellitus, cardiovascular and pulmonary diseases, HIV infection, and cancer.1,2

Schizophrenia and bipolar disorder are also frequently complicated by psychiatric comorbidities including alcohol and substance use disorders (such as nicotine dependence), anxiety disorders, and eating disorders (such as binge eating).3,4 Even so, the impact of psychiatric comorbidity on medical morbidity and mortality has not been adequately investigated and has—until rather recently—received little attention from clinical researchers.

Within the past 5 years or so, epidemiologic and population studies have shown increased morbidity and mortality from medical illnesses in patients with serious and persistent mental illness.5,6 These findings have sparked renewed interest in behavioral, biological, and psychosocial factors associated with schizophrenia and bipolar disorder that may contribute to comorbid medical illnesses. These factors include:

  • negative and depressive symptoms
  • physical inactivity and poor diet
  • hypothalamic-pituitary-adrenal axis dysregulation associated with acute psychotic and affective episodes
  • amotivation
  • social isolation
  • limited access to primary and preventive health care.7

The degree to which these factors may elevate medical comorbidity rates in patients with Commentary these serious psychiatric illnesses has not been well-studied or described.

TREATMENT IMPLICATIONS

These observations raise concerns about safe and effective use of medications by patients with schizophrenia or bipolar disorder as well as medical illness. Issues for clinicians to consider include:

  • possible pharmacokinetic and pharmacodynamic interactions in patients taking concomitant medications for psychiatric and medical illnesses
  • potential beneficial and adverse effects of psychotropics on medical illnesses
  • and—conversely—potential beneficial and adverse effects on mood and psychotic disorders of medications used to treat medical illnesses.

Being aware of metabolic effects when prescribing psychotropics is not a new idea. Lithium, for example, has long been known to cause weight gain, suppress thyroid hormone, and interact with thiazide diuretics when used to treat bipolar disorder. Thus, therapeutic blood monitoring is recommended for patients receiving acute and maintenance lithium therapy.

More recently, atypical antipsychotics such as olanzapine and clozapine (and risperidone and quetiapine to a lesser extent) have been shown to produce substantial weight gain and other metabolic effects that increase the risk of diabetes and cardiovascular disease.7 As a result, the American Diabetes Association—along with the American Psychiatric Association and other groups—now recommends that psychiatrists and other physicians monitor patients’ body weight and body mass index, vital signs, serum glucose, and lipids when prescribing these agents.7-11 Careful monitoring should improve the medical care of patients with schizophrenia and bipolar disorder and help protect them from the medical risks associated with overweight and obesity.

Similar precautions are needed when schizophrenia and bipolar disorder co-occur with other medical illnesses treated by complex pharmacologic regimens, such as pulmonary disease, cancer, and HIV. Three points to keep in mind are:

  • careful selection of treatments for co-occurring medical illnesses, considering potential effects on the primary psychiatric disorder
  • the impact of psychotropics on patients’ medical illnesses
  • and the potential for drug interactions.
References

1. Kilbourne AM, Cornelius JR, Han X, et al. Burden of general medical conditions among individuals with bipolar disorder. Bipolar Disord 2004;6:368-73.

2. Meyer JM, Nasrallah HA. Medical illness and schizophrenia Washington, DC: American Psychiatric Press, Inc, 2003.

3. McElroy SL, Altshuler LL, Suppes T, et al. Axis I psychiatric comorbidity and its relationship with historical illness variables in 288 patients with bipolar disorder. Am J Psychiatry 2001;158:420-6.

4. Green AI, Canuso CM, Brenner MJ, et al. Detection and management of comorbidity in patients with schizophrenia. Psychiatr Clin North Am 2003;26:115-39.

5. Buda M, Tsuang MT, Fleming JA. Causes of death in DSM-III schizophrenics and other psychotics (atypical group). Comparison with the general population. Arch Gen Psychiatry 1988;45:283-5.

6. Osby U, Brandt L, Correia N, et al. Excess mortality in bipolar and unipolar disorder in Sweden. Arch Gen Psychiatry 2001;58:844-50.

7. Keck PE, Jr, Buse JB, Dagago-Jack S, et al. Managing metabolic concerns in patients with severe mental illness: a special report (Postgrad Med). Minneapolis, MN: McGraw-Hill, 2003.

8. American Diabetes Association. Consensus Development Conference on Antipsychotic Drugs and Obesity and Diabetes. Diabetes Care 2004;27:596-601.

9. Marder SR, Essock SM, Miller AL, et al. Physical health monitoring of patients with schizophrenia. Am J Psychiatry 2004;161:1334-9.

10. Chue P, Kovacs CS. Safety and tolerability of atypical antipsychotics in patients with bipolar disorder: prevalence, monitoring and management. Bipolar Disord 2003;5(suppl 2):62-79.

11. Nasrallah HA, Newcomer JW. Atypical antipsychotics and metabolic dysregulation. Evaluating the risk/benefit equation and improving standard of care. J Clin Psychopharmacol 2004;24(suppl 2):S7-S14.

References

1. Kilbourne AM, Cornelius JR, Han X, et al. Burden of general medical conditions among individuals with bipolar disorder. Bipolar Disord 2004;6:368-73.

2. Meyer JM, Nasrallah HA. Medical illness and schizophrenia Washington, DC: American Psychiatric Press, Inc, 2003.

3. McElroy SL, Altshuler LL, Suppes T, et al. Axis I psychiatric comorbidity and its relationship with historical illness variables in 288 patients with bipolar disorder. Am J Psychiatry 2001;158:420-6.

4. Green AI, Canuso CM, Brenner MJ, et al. Detection and management of comorbidity in patients with schizophrenia. Psychiatr Clin North Am 2003;26:115-39.

5. Buda M, Tsuang MT, Fleming JA. Causes of death in DSM-III schizophrenics and other psychotics (atypical group). Comparison with the general population. Arch Gen Psychiatry 1988;45:283-5.

6. Osby U, Brandt L, Correia N, et al. Excess mortality in bipolar and unipolar disorder in Sweden. Arch Gen Psychiatry 2001;58:844-50.

7. Keck PE, Jr, Buse JB, Dagago-Jack S, et al. Managing metabolic concerns in patients with severe mental illness: a special report (Postgrad Med). Minneapolis, MN: McGraw-Hill, 2003.

8. American Diabetes Association. Consensus Development Conference on Antipsychotic Drugs and Obesity and Diabetes. Diabetes Care 2004;27:596-601.

9. Marder SR, Essock SM, Miller AL, et al. Physical health monitoring of patients with schizophrenia. Am J Psychiatry 2004;161:1334-9.

10. Chue P, Kovacs CS. Safety and tolerability of atypical antipsychotics in patients with bipolar disorder: prevalence, monitoring and management. Bipolar Disord 2003;5(suppl 2):62-79.

11. Nasrallah HA, Newcomer JW. Atypical antipsychotics and metabolic dysregulation. Evaluating the risk/benefit equation and improving standard of care. J Clin Psychopharmacol 2004;24(suppl 2):S7-S14.

Issue
Current Psychiatry - 03(12)
Issue
Current Psychiatry - 03(12)
Page Number
41-42
Page Number
41-42
Publications
Publications
Topics
Article Type
Display Headline
Commentary: When medical illness complicates schizophrenia and bipolar disorder
Display Headline
Commentary: When medical illness complicates schizophrenia and bipolar disorder
Sections
Article Source

PURLs Copyright

Inside the Article

Update on eating disorders: Binge-eating disorder

Article Type
Changed
Tue, 12/11/2018 - 15:10
Display Headline
Update on eating disorders: Binge-eating disorder

Clinical snapshot of BED

Managing patients with binge-eating disorder (BED) often requires behavioral, medical, and psychiatric interventions.

These patients suffer from recurrent episodes of distressing, uncontrollable overeating, but they do not purge or show other compensatory weight-loss behaviors common to bulimia nervosa1 and anorexia nervosa.2-10 As a result, they are often overweight or obese and may have obesity-related illnesses, such as hypertension or type 2 diabetes. Mild to severe depression—unipolar or bipolar—is a common psychopathology.

Because no one treatment fits all patients with binge eating disorder, their management usually requires an individualized program of:

  • behavioral weight control
  • psychotherapy
  • and sometimes medications.

In our weight management clinic, we consider medication options based on patient preference and whether BED is uncomplicated (Figure 1) or coexists with a mood disorder (Figure 2).

This article presents the evidence on which we base our comprehensive approach. General psychiatrists with knowledge of BED can treat patients with this eating disorder, although complicated cases may require referral for specialized treatment.

Figure 1 Medication options for uncomplicated BED


Clinical characteristics

Psychiatric comorbidity. BED often occurs in patients with mood, anxiety, substance-abuse, impulsecontrol, and personality disorders.4,6,10-12 Mood disorder—particularly depression—appears to be the most common comorbidity. BED can occur with bipolar disorder12—a comorbidity that in our experience is underrecognized both clinically and in the literature.

Patients with BED and bipolar disorder show increased impulsivity and mood lability. As bipolar II disorder and other “soft-spectrum” forms are more common than bipolar I disorder, BED is also more likely to occur with hypomania than mania.

Overweight. Not surprisingly, BED is associated with overweight and obesity.5,8,9,11 Not all patients with BED are overweight or obese, but most who participate in clinical trials of BED treatments are at least overweight. BED has been reported in up to:

  • 30% of participants in weight-loss programs7
  • 70% of participants in groups such as Overeaters Anonymous
  • 50% of patients who seek bariatric surgery.5

In our experience, patients are often more distressed by their weight than by their binge eating, depression, or anxiety. Indeed, overweight and obesity are the usual reasons patients with BED present for treatment at our center.

Diagnosis. BED’s validity as a clinical diagnosis has been controversial since the disorder was first included in DSM-IV (Table 1).3 Debate continues about some definitions in the DSM criteria, including what amount of food is “definitely larger” than most people would eat and what is “loss of control over eating.”

Nevertheless, screening for BED is relatively easy. Clinicians may use the eating disorder section of the Structured Clinical Interview for DSM-IV or the Eating Disorders Examination. Alternatively, simply ask patients if they have episodes of uncontrollable overeating, during which they eat unusually large amounts of food and their eating feels out of control.

Course. BED begins in adolescence or adulthood. Disease course is variable, with periods of remission, recurrence, and chronicity.6,7,10 Interestingly, one prospective study showed that even if the binge eating resolves, persons may still develop obesity.13

Prevalence. BED affects 1.5% to 3% of the U.S. population. It is more common in women than men, equally prevalent in whites and blacks, and more prevalent than anorexia nervosa and bulimia nervosa combined.11,14 Subthreshold BED—such as obesity with infrequent or nondistressing binge eating—appears to be much more common,10 although no data are available.

Theories of binge eating

BED’s cause is unknown, but biological, familial, and psychosocial factors have been implicated.

Biological factors. The neurotransmitters serotonin (5-HT) and dopamine—as well as various peptides—have been shown to help regulate feeding behavior.10

Table 1

Diagnostic criteria for binge-eating disorder*

  1. Recurrent episodes of binge eating are characterized by both of the following:
    • eating in a discrete period of time (as within any 2 hours) an amount of food that is definitely larger than what most people would eat in a similar period under similar circumstances
    • a sense of lack of control over eating during the episode (a feeling that one cannot stop eating or control what or how much one is eating)
  2. The binge-eating episodes are associated with three or more of the following:
    • eating much more rapidly than normal
    • eating until feeling uncomfortably full
    • eating large amounts of food when not feeling physically hungry
    • eating alone because of being embarrassed by how much one is eating
    • feeling disgusted with oneself, depressed, or very guilty after overeating
  3. Marked distress regarding binge eating is present.
  4. The binge eating occurs, on average, at least 2 days a week for 6 months.
  5. The binge eating is not associated with the regular use of inappropriate compensatory behaviors (purging, fasting, excessive exercise) and does not occur exclusively during the course of anorexia nervosa or bulimia nervosa.
* Research criteria, DSM-IV-TR appendix B.
Source: Reprinted with permission from the Diagnostic and statistical manual of mental disorders, 4th edition, text revision. Copyright 2000. American Psychiatric Association.
 

 

Serotonin. Reduced 5-HT transporter binding has been shown in obese women with BED.15 Their 5-HT binding improved and binge eating subsided with group psychotherapy and fluoxetine, although the women continued to gain weight.

Figure 2 Medication options for BED with obesity and a mood disorder*



Dopamine. Obese patients who compulsively overeat may have lower levels of dopamine D2 receptors than do normal-weight controls.16

Genetic factors. In severely obese patients (body mass index 44±2), those with a DSM-IV diagnosis of BED exhibited mutations of the melanocortin 4 receptor gene, which affects the anorectic properties of alpha melanocyte-stimulating hormone.17

Familial factors associated with BED include parental depression and obesity.18

Psychosocial correlates include physical and sexual abuse, bullying by peers, and discrimination because of being overweight.19

Treatment recommendations

Few systematic studies have examined BED treatment. Emerging research suggests that behavioral weight-loss treatment, specialized psychotherapies, and medications may be effective in some patients with BED.4,6,8

Behavioral weight-loss treatment’s main goal is to manage the patient’s weight with a lower-calorie, healthy diet and to increase exercise.20,21

Over the short term (<1 year), behavioral weight-loss treatment produces similar weight loss in obese patients with or without BED; long-term results in both groups, however, have not been satisfactory.20,21 No studies have examined the efficacy of specialized diets (such as low-carbohydrate regimens) in patients with BED.

Specialized psychotherapy’s goal is to modify bingeeating behavior with behavioral self-management strategies, reducing interpersonal dysfunction and stress, and/or managing affective dysregulation.

Cognitive-behavioral therapy (CBT) and interpersonal therapy (IPT) have been effective in reducing binge eating, both acutely and for up to 12 months4,20-24 but less effective in achieving and maintaining weight loss. Patients who achieve remission in binge eating after undergoing CBT or IPI often experience modest but stable weight loss.20-22 For example, in a comparison study of CBT and IPT:

  • After 20 weekly sessions, patients whose binge eating was in remission lost weight (mean body mass index [BMI] −0.5 ± 1.5 kg/m2), whereas those who continued to binge gained weight (mean BMI +0.4 ±2.0 kg/m2).
  • At 12 months’ follow-up, patients still in remission continued to lose weight (mean BMI −1.0 ± 3.0 kg/m2), whereas those no longer in remission gained weight (mean BMI +0.7 ±2.9 kg/m2[P = 0.01]).22

Self-help and dialectical behavioral therapy (DBT) may also help reduce binge eating in BED. As with CBT and IPT, they are less effective in weight loss. In the only controlled study of DBT,24 patients achieved an average 2.5-lb weight loss after 20 weeks of DBT, compared with an average 0.6-lb weight gain in the control group. This difference was not significant, and the report did not include data on weight loss maintenance.

In summary, CBT may be more effective than behavioral weight loss treatment for reducing binge eating, but behavioral weight loss is more effective for weight loss.

Medications for BED

Medications that have been tried for BED include antidepressants, appetite suppressants, and anticonvulsants.25,26 Antidepressants are used to treat BED because:

  • BED is often associated with depressive symptoms and disorders.
  • BED is related to bulimia nervosa, and placebo-controlled trials have shown that the binge eating of bulimia nervosa responds to several classes of antidepressants. The selective serotonin reuptake inhibitor (SSRI) fluoxetine is the only medication indicated for treating any eating disorder (bulimia nervosa).
  • Bupropion and venlafaxine—a serotonin-norepinephrine reuptake inhibitor (SNRI)—have weight-loss properties.

SSRIs are the most extensively studied antidepressants for treating BED. SSRIs have weightloss properties, but only short term.25-26 Citalopram, fluoxetine, fluvoxamine, and sertraline have reduced binge eating and body weight more effectively than placebo during 6 to 9 weeks of treatment (Table 2).25-26 However, one controlled study23 showed that fluoxetine was not significantly more effective than placebo in reducing binge frequency or body weight after 16 weeks.

TCAs. Studies of tricyclic antidepressants (TCAs) for BED are sparse, and results have been mixed. In one trial, imipramine was similar to placebo in reducing binge frequency and body weight. In a placebo-controlled study of patients with nonpurging bulimia nervosa, desipramine reduced binge eating but had no effect on body weight.25,26

Table 2

Drug therapies shown to be effective for BED*

MedicationBinge eatingWeightDepressionStudy sizeDuration (weeks)Dosage (mg/d)
Antidepressants
Citalopram++38620 to 60
Fluoxetine †+++60620 to 80
Fluvoxamine++85950 to 300
Sertraline++34650 to 200
Appetite suppressant
Sibutramine+++601215
Anticonvulsant
Topiramate++611450 to 600
+ Improvement
− No improvement
* Randomized, controlled trials. Antidepressants were studied in patients with BED; sibutramine and topiramate were studied in patients with BED and associated obesity.
† One 16-week trial of fluoxetine for BED (reference 23) did not show statistically significant differences in post-treatment binge frequency or body-mass index.

Venlafaxine. In a retrospective review of 35 consecutive obese women with BED, venlafaxine, mean 222 mg/d for 28 to 300 days (median 120 days), reduced binge eating, body weight, and depressive symptoms.27

 

 

Bupropion has been more effective than placebo for treating:

  • uncomplicated obesity (short- and long-term)
  • obesity associated with depressive symptoms
  • bulimia nervosa (although bupropion is contraindicated in these patients because of seizure risk).26,28,29

No controlled trials have studied bupropion for BED. When using dosages effective in depressive disorders, we find bupropion helpful in reducing binge eating, body weight, and depressive symptoms in BED patients.

Appetite suppressants decrease appetite and weight, may increase satiety, and may reduce depressive symptoms.

Sibutramine—a serotonin, norepinephrine, and dopamine reuptake inhibitor indicated for managing obesity—has been reported effective in BED in a 12-week, randomized, double-blind, placebo-controlled trial. A 15-mg/d dosage reduced binge frequency, body weight, and depressive symptoms more effectively than placebo in 60 obese patients with BED.30 Most-frequent adverse effects (dry mouth and constipation) were mild and benign, and no significant complications were observed.

Sibutramine’s mechanism of action in BED is unknown. However, it suppressed food intake during binge-eating episodes in patients with BED in a randomized, controlled, cross-over laboratory study.31

Orlistat. We know of no published controlled studies of the lipase inhibitor orlistat in treating BED. In our experience, some patients do well with this agent, though we have observed infrequent purging episodes with it in patients with BED.

With orlistat, 120 mg tid, our BED patients have experienced weight loss comparable to that seen in uncomplicated obesity at similar dosages. Orlistat seems most effective for:

  • patients whose binge eating is in remission
  • those who responded to behavioral weightloss treatment, a psychological treatment, or another medication.

Anticonvulsants such as topiramate and zonisamide have been shown effective in treating obesity32,33 and are sometimes used to treat BED. Obese BED patients with mood disorders often do best with psychotherapy plus medication

Topiramate at dosages of 50 to 600 mg/d (median 212 mg/d) reduced binge-eating frequency, obsessive-compulsive features of binge eating, and body weight more effectively than placebo in a 14-week study of 61 obese patients with BED. These effects were maintained across 48 weeks in an open-label extension trial.34

Zonisamide, mean 513 mg/d, produced similar results during a prospective, open-label, 12-week trial in 15 patients with BED.35 A controlled trial to replicate these findings is ongoing.

BED may respond to anticonvulsant therapy for several reasons:

  • Some anticonvulsants are effective in treating bipolar disorder, which may occur with BED.12
  • Some anticonvulsants have shown benefit in conditions associated with pathologic impulsivity, such as substance abuse, impulse-control, and cluster B disorders.10

Growing evidence shows that bulimia nervosa and BED may be associated with pathologic impulsivity.

Combination therapies are generally more effective than monotherapies in patients with mood disorders, uncomplicated obesity, and possibly bulimia nervosa. Even so, few trials have systematically studied combination therapy in managing patients with BED.

Two studies compared psychotherapy and antidepressants alone and in combination in treating BED.21,23 Both showed that CBT alone was more effective in decreasing binge frequency than desipramine alone,21 fluoxetine alone,23 and the combination of CBT and medication. On the other hand, patients who took desipramine either alone or in combination experienced a greater degree of weight loss than those who did not take desipramine.21

In another combination therapy, exercise has been shown to be an effective adjunct to CBT in maintenance treatment of obese women with BED.36

No studies have compared behavioral weight management or a specialized psychotherapy in combination with an antiobesity drug or a weight-loss anticonvulsant in treating BED.

Treatment recommendations

In our experience, BED patients—particularly those with obesity and psychopathology—often do best with some combination of psychological treatment and medication:

  • The psychological component may be behavioral weight-loss treatment, a specialized psychotherapy such as CBT or IPT, or some combination of behavioral weight-loss treatment and specialized psychotherapy.
  • The medication component may consist of an antidepressant, anticonvulsant, antiobesity drug, or multiple drugs (such as an SSRI or sibutramine with topiramate for BED with major depression, or topiramate with lithium for BED with bipolar disorder).

Although combination therapies may be optimal for some patients, this approach remains unproven in controlled trials.

Patient preference. In addition to comorbidities, patient preference is an important consideration when choosing BED treatments. We determine our patients’ preferences by educating them as much as possible about their options. We explain the benefits and weaknesses of all treatments and encourage them to participate in forming their individualized treatment plans.

Patients sometimes have strong treatment preferences. Some prefer psychological treatments, whereas others prefer medications. Working with patient preferences enhances treatment adherence. For example, patients who fail a preferred treatment are often more willing to adhere to another treatment modality about which they initially were skeptical.

 

 

Related resources

  • Bray GA, Bouchard C (eds). Handbook of obesity: clinical applications (2nd ed). New York, NY: Marcel Dekker, 2004.
  • Cooper Z, Zairburn CG, Hawker DM. Cognitive behavioral treatment of obesity. A clinician’s guide. New York: Guilford Press, 2003.
  • Carter WP, Hudson JI, Lalonde JK, et al. Pharmacologic treatment of binge eating disorder. Int J Eat Disord 2003;34(suppl):S74-88.

Drug brand names

  • Bupropion • Wellbutrin
  • Citalopram • Celexa
  • Desipramine • Norpramin
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Imipramine • Tofranil
  • Lamotrigine • Lamictal
  • Lithium • Eskalith, others
  • Orlistat • Xenical
  • Sertraline • Zoloft
  • Sibutramine • Meridia
  • Topiramate • Topamax
  • Venlafaxine • Effexor
  • Zonisamide • Zonegran

Disclosure

Dr. Kotwal receives grant support from Elan Corporation and is a speaker for Ortho-McNeil Pharmaceutical and Pfizer Inc.

Dr. Kaneria and Ms. Guerdjikova report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. McElroy is a consultant to Abbott Laboratories, Bristol-Myers Squibb Co., Elan Corporation, GlaxoSmithKline, Janssen Pharmaceutica, Eli Lilly and Co., and Ortho-McNeil Pharmaceutical. She receives grant/research support from Elan Pharmaceuticals, Forest Pharmaceuticals, Merck & Co., Ortho-McNeil Pharmaceutical, and Sanofi-Synthelabo and is a speaker for Eli Lilly and Co. and Ortho-McNeil Pharmaceutical.

References

1. Pope HG, Hudson JI. Bulimia nervosa: Persistent disorders requires equally persistent treatment. Current Psychiatry 2004;3(1):13-22.

2. Halmi KA. Anorexia nervosa: Dual therapy can bring patients back from the brink. Current Psychiatry 2004;3(3):39-56.

3. American Psychiatric Association. Diagnostic and statistical manual of mental disorders (4th ed). Washington, DC: American Psychiatric Association, 1994.

4. Agras WS. Treatment of binge eating disorder. In: Gabbard GO (ed). Treatments of psychiatric disorders (3rd ed). Washington, DC: American Psychiatric Press, 2001;2209-19.

5. de Zwaan M. Binge eating disorder and obesity. Int J Obes Relat Metab Disord 2001;25(suppl 1):S51-5.

6. Dingemans AE, Bruna MJ, van Furth EF. Binge eating disorder: a review. Int J Obes Relat Metab Disord 2002;26:299-307.

7. Fairburn CG, Harrison PJ. Eating disorders. Lancet 2003;361:407-16.

8. Walsh BT (ed). The current status of binge eating disorder. Int J Eat Disord 2003;34(suppl):S1-120.

9. Devlin MJ, Goldfein JA, Dobrow I. What is this thing called BED? Current status of binge eating disorder nosology. Int J Eat Disord 2003;34(suppl):S2-18.

10. McElroy SL, Kotwal R. Binge eating. In: Hollander E, Stein D (eds). Handbook of impulse control disorders Washington, DC: American Psychiatric Press (in press).

11. Smith DE, Marcus MD, Lewis CE, et al. Prevalence of binge eating disorder, obesity, and depression in a biracial cohort of young adults. Ann Behav Med 1998;20:227-32.

12. Kruger S, Shugar G, Cooke RG. Comorbidity of binge eating disorder and the partial binge eating syndrome with bipolar disorder. Int J Eat Disord 1996;19:45-52.

13. Fairburn CG, Cooper Z, Doll H, et al. The natural course of bulimia nervosa and binge eating disorder in young women. Arch Gen Psychiatry 2000;57:659-65.

14. Striegel-Moore RH, Franko DL. Epidemiology of binge eating disorder. Int J Eat Disord 2003;34(suppl):S19-29.

15. Tammela LI, Rissanen A, Kuikka JT, et al. Treatment improves serotonin transporter binding and reduces binge eating. Psychopharmacology (Berl) 2003;170:89-93.

16. Wang GJ, Volkow ND, Logan J, et al. Brain dopamine and obesity. Lancet 2001;357:354-7.

17. Branson R, Potoczna N, Kral JG, et al. Binge eating as a major phenotype of melanocortin 4 receptor gene mutations. N Engl J Med 2003;348:1096-103.

18. Fairburn CG, Doll HA, Welch SL, et al. Risk factors for binge eating disorder: a community-based, case-control study. Arch Gen Psychiatry 1998;55:425-32.

19. Striegel-Moore RH, Dohm FA, Pike KM, et al. Abuse, bullying, and discrimination as risk factors for binge eating disorder. Am J Psychiatry 2002;159:1902-7.

20. Wonderlich SA, de Zwaan M, Mitchell JE, et al. Psychological and dietary treatments of binge eating disorder: conceptual implications. Int J Eat Disord 2003;34(suppl):S58-78.

21. Agras WS, Telch DF, Arnow B, et al. Weight loss, cognitive-behavioral, and desipramine treatments in binge eating disorder. An additive design. Behav Ther 1994;25:225-38.

22. Wilfley DE, Welch RR, Stein RI, et al. A randomized comparison of group cognitive-behavioral therapy and group interpersonal psychotherapy for the treatment of overweight individuals with binge eating disorder. Arch Gen Psychiatry 2002;59:713-21.

23. Grilo CM. A controlled study of cognitive behavioral therapy and fluoxetine for binge eating disorder (presentation) Charleston, SC: Eating Disorders Research Society annual meeting, 2002.

24. Telch CF, Agras WS, Linehan MM. Dialectical behavior therapy for binge eating disorder. J Consult Clin Psychol 2001;69:1061-5.

25. Carter WP, Hudson JI, Lalonde JK, et al. Pharmacologic treatment of binge eating disorder. Int J Eat Disord 2003;34(suppl):S74-88.

26. Appolinario JC, McElroy SL. Pharmacologic approaches in the treatment of binge eating disorder. Curr Drug Targets (in press).

27. Malhotra S, King KH, Welge JA, et al. Venlafaxine treatment of binge-eating disorder associated with obesity: a series of 35 patients. J Clin Psychiatry 2002;63:802-6.

28. Anderson JW, Greenway FL, Fujioka K, et al. Bupropion SR enhances weight loss: a 48-week double-blind, placebo-controlled trial. Obes Res 2002;10:633-41.

29. McElroy SL, Kotwal R, Malhotra S, et al. Are mood disorders and obesity related? A review for the mental health professional. J Clin Psychiatry (in press).

30. Appolinario JC, Bacaltchuk J, Sichieri R, et al. A randomized, double-blind, placebo-controlled study of sibutramine in the treatment of binge-eating disorder. Arch Gen Psychiatry 2003;60:1109-16.

31. Mitchell JE, Gosnell BA, Roerig JL, et al. Effects of sibutramine on binge eating, hunger, and fullness in a laboratory human feeding paradigm. Obes Res 2003;11:599-602.

32. Bray GA, Hollander P, Klein S, et al. A 6-month randomized, placebo-controlled, dose-ranging trial of topiramate for weight loss in obesity. Obes Res 2003;11:722-33.

33. Gadde KM, Franciscy DM, Wagner HR, 2nd, Krishnan KR. Zonisamide for weight loss in obese adults: a randomized controlled trial. JAMA 2003;289:1820-5.

34. McElroy SL, Arnold LM, Shapira NA, et al. Topiramate in the treatment of binge eating disorder associated with obesity: a randomized, placebo-controlled trial. Am J Psychiatry 2003;160:255-61.

35. McElroy SL, Kotwal R, Hudson JI, et al. Zonisamide in the treatment of binge-eating disorder: an open-label, prospective trial. J Clin Psychiatry 2004;65:50-6.

36. Pendleton VR, Goodrick GK, Poston WSC, et al. Exercise augments the effects of cognitive-behavioral therapy in the treatment of binge eating. Int J Eat Disord 2002;31(2):172-84.

Author and Disclosure Information

Paul E. Keck, Jr, MD

Renu Kotwal, MD
Assistant professor of clinical psychiatry

Rakesh Kaneria, MD
Fourth-year resident in psychiatry

Anna Guerdjikova, MS
PhD candidate, neuroscience program

Susan L. McElroy, MD
Professor of psychiatry and neuroscience

Division of Psychopharmacology Research Department of Psychiatry University of Cincinnati College of Medicine

Issue
Current Psychiatry - 03(04)
Publications
Topics
Page Number
31-47
Sections
Author and Disclosure Information

Paul E. Keck, Jr, MD

Renu Kotwal, MD
Assistant professor of clinical psychiatry

Rakesh Kaneria, MD
Fourth-year resident in psychiatry

Anna Guerdjikova, MS
PhD candidate, neuroscience program

Susan L. McElroy, MD
Professor of psychiatry and neuroscience

Division of Psychopharmacology Research Department of Psychiatry University of Cincinnati College of Medicine

Author and Disclosure Information

Paul E. Keck, Jr, MD

Renu Kotwal, MD
Assistant professor of clinical psychiatry

Rakesh Kaneria, MD
Fourth-year resident in psychiatry

Anna Guerdjikova, MS
PhD candidate, neuroscience program

Susan L. McElroy, MD
Professor of psychiatry and neuroscience

Division of Psychopharmacology Research Department of Psychiatry University of Cincinnati College of Medicine

Clinical snapshot of BED

Managing patients with binge-eating disorder (BED) often requires behavioral, medical, and psychiatric interventions.

These patients suffer from recurrent episodes of distressing, uncontrollable overeating, but they do not purge or show other compensatory weight-loss behaviors common to bulimia nervosa1 and anorexia nervosa.2-10 As a result, they are often overweight or obese and may have obesity-related illnesses, such as hypertension or type 2 diabetes. Mild to severe depression—unipolar or bipolar—is a common psychopathology.

Because no one treatment fits all patients with binge eating disorder, their management usually requires an individualized program of:

  • behavioral weight control
  • psychotherapy
  • and sometimes medications.

In our weight management clinic, we consider medication options based on patient preference and whether BED is uncomplicated (Figure 1) or coexists with a mood disorder (Figure 2).

This article presents the evidence on which we base our comprehensive approach. General psychiatrists with knowledge of BED can treat patients with this eating disorder, although complicated cases may require referral for specialized treatment.

Figure 1 Medication options for uncomplicated BED


Clinical characteristics

Psychiatric comorbidity. BED often occurs in patients with mood, anxiety, substance-abuse, impulsecontrol, and personality disorders.4,6,10-12 Mood disorder—particularly depression—appears to be the most common comorbidity. BED can occur with bipolar disorder12—a comorbidity that in our experience is underrecognized both clinically and in the literature.

Patients with BED and bipolar disorder show increased impulsivity and mood lability. As bipolar II disorder and other “soft-spectrum” forms are more common than bipolar I disorder, BED is also more likely to occur with hypomania than mania.

Overweight. Not surprisingly, BED is associated with overweight and obesity.5,8,9,11 Not all patients with BED are overweight or obese, but most who participate in clinical trials of BED treatments are at least overweight. BED has been reported in up to:

  • 30% of participants in weight-loss programs7
  • 70% of participants in groups such as Overeaters Anonymous
  • 50% of patients who seek bariatric surgery.5

In our experience, patients are often more distressed by their weight than by their binge eating, depression, or anxiety. Indeed, overweight and obesity are the usual reasons patients with BED present for treatment at our center.

Diagnosis. BED’s validity as a clinical diagnosis has been controversial since the disorder was first included in DSM-IV (Table 1).3 Debate continues about some definitions in the DSM criteria, including what amount of food is “definitely larger” than most people would eat and what is “loss of control over eating.”

Nevertheless, screening for BED is relatively easy. Clinicians may use the eating disorder section of the Structured Clinical Interview for DSM-IV or the Eating Disorders Examination. Alternatively, simply ask patients if they have episodes of uncontrollable overeating, during which they eat unusually large amounts of food and their eating feels out of control.

Course. BED begins in adolescence or adulthood. Disease course is variable, with periods of remission, recurrence, and chronicity.6,7,10 Interestingly, one prospective study showed that even if the binge eating resolves, persons may still develop obesity.13

Prevalence. BED affects 1.5% to 3% of the U.S. population. It is more common in women than men, equally prevalent in whites and blacks, and more prevalent than anorexia nervosa and bulimia nervosa combined.11,14 Subthreshold BED—such as obesity with infrequent or nondistressing binge eating—appears to be much more common,10 although no data are available.

Theories of binge eating

BED’s cause is unknown, but biological, familial, and psychosocial factors have been implicated.

Biological factors. The neurotransmitters serotonin (5-HT) and dopamine—as well as various peptides—have been shown to help regulate feeding behavior.10

Table 1

Diagnostic criteria for binge-eating disorder*

  1. Recurrent episodes of binge eating are characterized by both of the following:
    • eating in a discrete period of time (as within any 2 hours) an amount of food that is definitely larger than what most people would eat in a similar period under similar circumstances
    • a sense of lack of control over eating during the episode (a feeling that one cannot stop eating or control what or how much one is eating)
  2. The binge-eating episodes are associated with three or more of the following:
    • eating much more rapidly than normal
    • eating until feeling uncomfortably full
    • eating large amounts of food when not feeling physically hungry
    • eating alone because of being embarrassed by how much one is eating
    • feeling disgusted with oneself, depressed, or very guilty after overeating
  3. Marked distress regarding binge eating is present.
  4. The binge eating occurs, on average, at least 2 days a week for 6 months.
  5. The binge eating is not associated with the regular use of inappropriate compensatory behaviors (purging, fasting, excessive exercise) and does not occur exclusively during the course of anorexia nervosa or bulimia nervosa.
* Research criteria, DSM-IV-TR appendix B.
Source: Reprinted with permission from the Diagnostic and statistical manual of mental disorders, 4th edition, text revision. Copyright 2000. American Psychiatric Association.
 

 

Serotonin. Reduced 5-HT transporter binding has been shown in obese women with BED.15 Their 5-HT binding improved and binge eating subsided with group psychotherapy and fluoxetine, although the women continued to gain weight.

Figure 2 Medication options for BED with obesity and a mood disorder*



Dopamine. Obese patients who compulsively overeat may have lower levels of dopamine D2 receptors than do normal-weight controls.16

Genetic factors. In severely obese patients (body mass index 44±2), those with a DSM-IV diagnosis of BED exhibited mutations of the melanocortin 4 receptor gene, which affects the anorectic properties of alpha melanocyte-stimulating hormone.17

Familial factors associated with BED include parental depression and obesity.18

Psychosocial correlates include physical and sexual abuse, bullying by peers, and discrimination because of being overweight.19

Treatment recommendations

Few systematic studies have examined BED treatment. Emerging research suggests that behavioral weight-loss treatment, specialized psychotherapies, and medications may be effective in some patients with BED.4,6,8

Behavioral weight-loss treatment’s main goal is to manage the patient’s weight with a lower-calorie, healthy diet and to increase exercise.20,21

Over the short term (<1 year), behavioral weight-loss treatment produces similar weight loss in obese patients with or without BED; long-term results in both groups, however, have not been satisfactory.20,21 No studies have examined the efficacy of specialized diets (such as low-carbohydrate regimens) in patients with BED.

Specialized psychotherapy’s goal is to modify bingeeating behavior with behavioral self-management strategies, reducing interpersonal dysfunction and stress, and/or managing affective dysregulation.

Cognitive-behavioral therapy (CBT) and interpersonal therapy (IPT) have been effective in reducing binge eating, both acutely and for up to 12 months4,20-24 but less effective in achieving and maintaining weight loss. Patients who achieve remission in binge eating after undergoing CBT or IPI often experience modest but stable weight loss.20-22 For example, in a comparison study of CBT and IPT:

  • After 20 weekly sessions, patients whose binge eating was in remission lost weight (mean body mass index [BMI] −0.5 ± 1.5 kg/m2), whereas those who continued to binge gained weight (mean BMI +0.4 ±2.0 kg/m2).
  • At 12 months’ follow-up, patients still in remission continued to lose weight (mean BMI −1.0 ± 3.0 kg/m2), whereas those no longer in remission gained weight (mean BMI +0.7 ±2.9 kg/m2[P = 0.01]).22

Self-help and dialectical behavioral therapy (DBT) may also help reduce binge eating in BED. As with CBT and IPT, they are less effective in weight loss. In the only controlled study of DBT,24 patients achieved an average 2.5-lb weight loss after 20 weeks of DBT, compared with an average 0.6-lb weight gain in the control group. This difference was not significant, and the report did not include data on weight loss maintenance.

In summary, CBT may be more effective than behavioral weight loss treatment for reducing binge eating, but behavioral weight loss is more effective for weight loss.

Medications for BED

Medications that have been tried for BED include antidepressants, appetite suppressants, and anticonvulsants.25,26 Antidepressants are used to treat BED because:

  • BED is often associated with depressive symptoms and disorders.
  • BED is related to bulimia nervosa, and placebo-controlled trials have shown that the binge eating of bulimia nervosa responds to several classes of antidepressants. The selective serotonin reuptake inhibitor (SSRI) fluoxetine is the only medication indicated for treating any eating disorder (bulimia nervosa).
  • Bupropion and venlafaxine—a serotonin-norepinephrine reuptake inhibitor (SNRI)—have weight-loss properties.

SSRIs are the most extensively studied antidepressants for treating BED. SSRIs have weightloss properties, but only short term.25-26 Citalopram, fluoxetine, fluvoxamine, and sertraline have reduced binge eating and body weight more effectively than placebo during 6 to 9 weeks of treatment (Table 2).25-26 However, one controlled study23 showed that fluoxetine was not significantly more effective than placebo in reducing binge frequency or body weight after 16 weeks.

TCAs. Studies of tricyclic antidepressants (TCAs) for BED are sparse, and results have been mixed. In one trial, imipramine was similar to placebo in reducing binge frequency and body weight. In a placebo-controlled study of patients with nonpurging bulimia nervosa, desipramine reduced binge eating but had no effect on body weight.25,26

Table 2

Drug therapies shown to be effective for BED*

MedicationBinge eatingWeightDepressionStudy sizeDuration (weeks)Dosage (mg/d)
Antidepressants
Citalopram++38620 to 60
Fluoxetine †+++60620 to 80
Fluvoxamine++85950 to 300
Sertraline++34650 to 200
Appetite suppressant
Sibutramine+++601215
Anticonvulsant
Topiramate++611450 to 600
+ Improvement
− No improvement
* Randomized, controlled trials. Antidepressants were studied in patients with BED; sibutramine and topiramate were studied in patients with BED and associated obesity.
† One 16-week trial of fluoxetine for BED (reference 23) did not show statistically significant differences in post-treatment binge frequency or body-mass index.

Venlafaxine. In a retrospective review of 35 consecutive obese women with BED, venlafaxine, mean 222 mg/d for 28 to 300 days (median 120 days), reduced binge eating, body weight, and depressive symptoms.27

 

 

Bupropion has been more effective than placebo for treating:

  • uncomplicated obesity (short- and long-term)
  • obesity associated with depressive symptoms
  • bulimia nervosa (although bupropion is contraindicated in these patients because of seizure risk).26,28,29

No controlled trials have studied bupropion for BED. When using dosages effective in depressive disorders, we find bupropion helpful in reducing binge eating, body weight, and depressive symptoms in BED patients.

Appetite suppressants decrease appetite and weight, may increase satiety, and may reduce depressive symptoms.

Sibutramine—a serotonin, norepinephrine, and dopamine reuptake inhibitor indicated for managing obesity—has been reported effective in BED in a 12-week, randomized, double-blind, placebo-controlled trial. A 15-mg/d dosage reduced binge frequency, body weight, and depressive symptoms more effectively than placebo in 60 obese patients with BED.30 Most-frequent adverse effects (dry mouth and constipation) were mild and benign, and no significant complications were observed.

Sibutramine’s mechanism of action in BED is unknown. However, it suppressed food intake during binge-eating episodes in patients with BED in a randomized, controlled, cross-over laboratory study.31

Orlistat. We know of no published controlled studies of the lipase inhibitor orlistat in treating BED. In our experience, some patients do well with this agent, though we have observed infrequent purging episodes with it in patients with BED.

With orlistat, 120 mg tid, our BED patients have experienced weight loss comparable to that seen in uncomplicated obesity at similar dosages. Orlistat seems most effective for:

  • patients whose binge eating is in remission
  • those who responded to behavioral weightloss treatment, a psychological treatment, or another medication.

Anticonvulsants such as topiramate and zonisamide have been shown effective in treating obesity32,33 and are sometimes used to treat BED. Obese BED patients with mood disorders often do best with psychotherapy plus medication

Topiramate at dosages of 50 to 600 mg/d (median 212 mg/d) reduced binge-eating frequency, obsessive-compulsive features of binge eating, and body weight more effectively than placebo in a 14-week study of 61 obese patients with BED. These effects were maintained across 48 weeks in an open-label extension trial.34

Zonisamide, mean 513 mg/d, produced similar results during a prospective, open-label, 12-week trial in 15 patients with BED.35 A controlled trial to replicate these findings is ongoing.

BED may respond to anticonvulsant therapy for several reasons:

  • Some anticonvulsants are effective in treating bipolar disorder, which may occur with BED.12
  • Some anticonvulsants have shown benefit in conditions associated with pathologic impulsivity, such as substance abuse, impulse-control, and cluster B disorders.10

Growing evidence shows that bulimia nervosa and BED may be associated with pathologic impulsivity.

Combination therapies are generally more effective than monotherapies in patients with mood disorders, uncomplicated obesity, and possibly bulimia nervosa. Even so, few trials have systematically studied combination therapy in managing patients with BED.

Two studies compared psychotherapy and antidepressants alone and in combination in treating BED.21,23 Both showed that CBT alone was more effective in decreasing binge frequency than desipramine alone,21 fluoxetine alone,23 and the combination of CBT and medication. On the other hand, patients who took desipramine either alone or in combination experienced a greater degree of weight loss than those who did not take desipramine.21

In another combination therapy, exercise has been shown to be an effective adjunct to CBT in maintenance treatment of obese women with BED.36

No studies have compared behavioral weight management or a specialized psychotherapy in combination with an antiobesity drug or a weight-loss anticonvulsant in treating BED.

Treatment recommendations

In our experience, BED patients—particularly those with obesity and psychopathology—often do best with some combination of psychological treatment and medication:

  • The psychological component may be behavioral weight-loss treatment, a specialized psychotherapy such as CBT or IPT, or some combination of behavioral weight-loss treatment and specialized psychotherapy.
  • The medication component may consist of an antidepressant, anticonvulsant, antiobesity drug, or multiple drugs (such as an SSRI or sibutramine with topiramate for BED with major depression, or topiramate with lithium for BED with bipolar disorder).

Although combination therapies may be optimal for some patients, this approach remains unproven in controlled trials.

Patient preference. In addition to comorbidities, patient preference is an important consideration when choosing BED treatments. We determine our patients’ preferences by educating them as much as possible about their options. We explain the benefits and weaknesses of all treatments and encourage them to participate in forming their individualized treatment plans.

Patients sometimes have strong treatment preferences. Some prefer psychological treatments, whereas others prefer medications. Working with patient preferences enhances treatment adherence. For example, patients who fail a preferred treatment are often more willing to adhere to another treatment modality about which they initially were skeptical.

 

 

Related resources

  • Bray GA, Bouchard C (eds). Handbook of obesity: clinical applications (2nd ed). New York, NY: Marcel Dekker, 2004.
  • Cooper Z, Zairburn CG, Hawker DM. Cognitive behavioral treatment of obesity. A clinician’s guide. New York: Guilford Press, 2003.
  • Carter WP, Hudson JI, Lalonde JK, et al. Pharmacologic treatment of binge eating disorder. Int J Eat Disord 2003;34(suppl):S74-88.

Drug brand names

  • Bupropion • Wellbutrin
  • Citalopram • Celexa
  • Desipramine • Norpramin
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Imipramine • Tofranil
  • Lamotrigine • Lamictal
  • Lithium • Eskalith, others
  • Orlistat • Xenical
  • Sertraline • Zoloft
  • Sibutramine • Meridia
  • Topiramate • Topamax
  • Venlafaxine • Effexor
  • Zonisamide • Zonegran

Disclosure

Dr. Kotwal receives grant support from Elan Corporation and is a speaker for Ortho-McNeil Pharmaceutical and Pfizer Inc.

Dr. Kaneria and Ms. Guerdjikova report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. McElroy is a consultant to Abbott Laboratories, Bristol-Myers Squibb Co., Elan Corporation, GlaxoSmithKline, Janssen Pharmaceutica, Eli Lilly and Co., and Ortho-McNeil Pharmaceutical. She receives grant/research support from Elan Pharmaceuticals, Forest Pharmaceuticals, Merck & Co., Ortho-McNeil Pharmaceutical, and Sanofi-Synthelabo and is a speaker for Eli Lilly and Co. and Ortho-McNeil Pharmaceutical.

Clinical snapshot of BED

Managing patients with binge-eating disorder (BED) often requires behavioral, medical, and psychiatric interventions.

These patients suffer from recurrent episodes of distressing, uncontrollable overeating, but they do not purge or show other compensatory weight-loss behaviors common to bulimia nervosa1 and anorexia nervosa.2-10 As a result, they are often overweight or obese and may have obesity-related illnesses, such as hypertension or type 2 diabetes. Mild to severe depression—unipolar or bipolar—is a common psychopathology.

Because no one treatment fits all patients with binge eating disorder, their management usually requires an individualized program of:

  • behavioral weight control
  • psychotherapy
  • and sometimes medications.

In our weight management clinic, we consider medication options based on patient preference and whether BED is uncomplicated (Figure 1) or coexists with a mood disorder (Figure 2).

This article presents the evidence on which we base our comprehensive approach. General psychiatrists with knowledge of BED can treat patients with this eating disorder, although complicated cases may require referral for specialized treatment.

Figure 1 Medication options for uncomplicated BED


Clinical characteristics

Psychiatric comorbidity. BED often occurs in patients with mood, anxiety, substance-abuse, impulsecontrol, and personality disorders.4,6,10-12 Mood disorder—particularly depression—appears to be the most common comorbidity. BED can occur with bipolar disorder12—a comorbidity that in our experience is underrecognized both clinically and in the literature.

Patients with BED and bipolar disorder show increased impulsivity and mood lability. As bipolar II disorder and other “soft-spectrum” forms are more common than bipolar I disorder, BED is also more likely to occur with hypomania than mania.

Overweight. Not surprisingly, BED is associated with overweight and obesity.5,8,9,11 Not all patients with BED are overweight or obese, but most who participate in clinical trials of BED treatments are at least overweight. BED has been reported in up to:

  • 30% of participants in weight-loss programs7
  • 70% of participants in groups such as Overeaters Anonymous
  • 50% of patients who seek bariatric surgery.5

In our experience, patients are often more distressed by their weight than by their binge eating, depression, or anxiety. Indeed, overweight and obesity are the usual reasons patients with BED present for treatment at our center.

Diagnosis. BED’s validity as a clinical diagnosis has been controversial since the disorder was first included in DSM-IV (Table 1).3 Debate continues about some definitions in the DSM criteria, including what amount of food is “definitely larger” than most people would eat and what is “loss of control over eating.”

Nevertheless, screening for BED is relatively easy. Clinicians may use the eating disorder section of the Structured Clinical Interview for DSM-IV or the Eating Disorders Examination. Alternatively, simply ask patients if they have episodes of uncontrollable overeating, during which they eat unusually large amounts of food and their eating feels out of control.

Course. BED begins in adolescence or adulthood. Disease course is variable, with periods of remission, recurrence, and chronicity.6,7,10 Interestingly, one prospective study showed that even if the binge eating resolves, persons may still develop obesity.13

Prevalence. BED affects 1.5% to 3% of the U.S. population. It is more common in women than men, equally prevalent in whites and blacks, and more prevalent than anorexia nervosa and bulimia nervosa combined.11,14 Subthreshold BED—such as obesity with infrequent or nondistressing binge eating—appears to be much more common,10 although no data are available.

Theories of binge eating

BED’s cause is unknown, but biological, familial, and psychosocial factors have been implicated.

Biological factors. The neurotransmitters serotonin (5-HT) and dopamine—as well as various peptides—have been shown to help regulate feeding behavior.10

Table 1

Diagnostic criteria for binge-eating disorder*

  1. Recurrent episodes of binge eating are characterized by both of the following:
    • eating in a discrete period of time (as within any 2 hours) an amount of food that is definitely larger than what most people would eat in a similar period under similar circumstances
    • a sense of lack of control over eating during the episode (a feeling that one cannot stop eating or control what or how much one is eating)
  2. The binge-eating episodes are associated with three or more of the following:
    • eating much more rapidly than normal
    • eating until feeling uncomfortably full
    • eating large amounts of food when not feeling physically hungry
    • eating alone because of being embarrassed by how much one is eating
    • feeling disgusted with oneself, depressed, or very guilty after overeating
  3. Marked distress regarding binge eating is present.
  4. The binge eating occurs, on average, at least 2 days a week for 6 months.
  5. The binge eating is not associated with the regular use of inappropriate compensatory behaviors (purging, fasting, excessive exercise) and does not occur exclusively during the course of anorexia nervosa or bulimia nervosa.
* Research criteria, DSM-IV-TR appendix B.
Source: Reprinted with permission from the Diagnostic and statistical manual of mental disorders, 4th edition, text revision. Copyright 2000. American Psychiatric Association.
 

 

Serotonin. Reduced 5-HT transporter binding has been shown in obese women with BED.15 Their 5-HT binding improved and binge eating subsided with group psychotherapy and fluoxetine, although the women continued to gain weight.

Figure 2 Medication options for BED with obesity and a mood disorder*



Dopamine. Obese patients who compulsively overeat may have lower levels of dopamine D2 receptors than do normal-weight controls.16

Genetic factors. In severely obese patients (body mass index 44±2), those with a DSM-IV diagnosis of BED exhibited mutations of the melanocortin 4 receptor gene, which affects the anorectic properties of alpha melanocyte-stimulating hormone.17

Familial factors associated with BED include parental depression and obesity.18

Psychosocial correlates include physical and sexual abuse, bullying by peers, and discrimination because of being overweight.19

Treatment recommendations

Few systematic studies have examined BED treatment. Emerging research suggests that behavioral weight-loss treatment, specialized psychotherapies, and medications may be effective in some patients with BED.4,6,8

Behavioral weight-loss treatment’s main goal is to manage the patient’s weight with a lower-calorie, healthy diet and to increase exercise.20,21

Over the short term (<1 year), behavioral weight-loss treatment produces similar weight loss in obese patients with or without BED; long-term results in both groups, however, have not been satisfactory.20,21 No studies have examined the efficacy of specialized diets (such as low-carbohydrate regimens) in patients with BED.

Specialized psychotherapy’s goal is to modify bingeeating behavior with behavioral self-management strategies, reducing interpersonal dysfunction and stress, and/or managing affective dysregulation.

Cognitive-behavioral therapy (CBT) and interpersonal therapy (IPT) have been effective in reducing binge eating, both acutely and for up to 12 months4,20-24 but less effective in achieving and maintaining weight loss. Patients who achieve remission in binge eating after undergoing CBT or IPI often experience modest but stable weight loss.20-22 For example, in a comparison study of CBT and IPT:

  • After 20 weekly sessions, patients whose binge eating was in remission lost weight (mean body mass index [BMI] −0.5 ± 1.5 kg/m2), whereas those who continued to binge gained weight (mean BMI +0.4 ±2.0 kg/m2).
  • At 12 months’ follow-up, patients still in remission continued to lose weight (mean BMI −1.0 ± 3.0 kg/m2), whereas those no longer in remission gained weight (mean BMI +0.7 ±2.9 kg/m2[P = 0.01]).22

Self-help and dialectical behavioral therapy (DBT) may also help reduce binge eating in BED. As with CBT and IPT, they are less effective in weight loss. In the only controlled study of DBT,24 patients achieved an average 2.5-lb weight loss after 20 weeks of DBT, compared with an average 0.6-lb weight gain in the control group. This difference was not significant, and the report did not include data on weight loss maintenance.

In summary, CBT may be more effective than behavioral weight loss treatment for reducing binge eating, but behavioral weight loss is more effective for weight loss.

Medications for BED

Medications that have been tried for BED include antidepressants, appetite suppressants, and anticonvulsants.25,26 Antidepressants are used to treat BED because:

  • BED is often associated with depressive symptoms and disorders.
  • BED is related to bulimia nervosa, and placebo-controlled trials have shown that the binge eating of bulimia nervosa responds to several classes of antidepressants. The selective serotonin reuptake inhibitor (SSRI) fluoxetine is the only medication indicated for treating any eating disorder (bulimia nervosa).
  • Bupropion and venlafaxine—a serotonin-norepinephrine reuptake inhibitor (SNRI)—have weight-loss properties.

SSRIs are the most extensively studied antidepressants for treating BED. SSRIs have weightloss properties, but only short term.25-26 Citalopram, fluoxetine, fluvoxamine, and sertraline have reduced binge eating and body weight more effectively than placebo during 6 to 9 weeks of treatment (Table 2).25-26 However, one controlled study23 showed that fluoxetine was not significantly more effective than placebo in reducing binge frequency or body weight after 16 weeks.

TCAs. Studies of tricyclic antidepressants (TCAs) for BED are sparse, and results have been mixed. In one trial, imipramine was similar to placebo in reducing binge frequency and body weight. In a placebo-controlled study of patients with nonpurging bulimia nervosa, desipramine reduced binge eating but had no effect on body weight.25,26

Table 2

Drug therapies shown to be effective for BED*

MedicationBinge eatingWeightDepressionStudy sizeDuration (weeks)Dosage (mg/d)
Antidepressants
Citalopram++38620 to 60
Fluoxetine †+++60620 to 80
Fluvoxamine++85950 to 300
Sertraline++34650 to 200
Appetite suppressant
Sibutramine+++601215
Anticonvulsant
Topiramate++611450 to 600
+ Improvement
− No improvement
* Randomized, controlled trials. Antidepressants were studied in patients with BED; sibutramine and topiramate were studied in patients with BED and associated obesity.
† One 16-week trial of fluoxetine for BED (reference 23) did not show statistically significant differences in post-treatment binge frequency or body-mass index.

Venlafaxine. In a retrospective review of 35 consecutive obese women with BED, venlafaxine, mean 222 mg/d for 28 to 300 days (median 120 days), reduced binge eating, body weight, and depressive symptoms.27

 

 

Bupropion has been more effective than placebo for treating:

  • uncomplicated obesity (short- and long-term)
  • obesity associated with depressive symptoms
  • bulimia nervosa (although bupropion is contraindicated in these patients because of seizure risk).26,28,29

No controlled trials have studied bupropion for BED. When using dosages effective in depressive disorders, we find bupropion helpful in reducing binge eating, body weight, and depressive symptoms in BED patients.

Appetite suppressants decrease appetite and weight, may increase satiety, and may reduce depressive symptoms.

Sibutramine—a serotonin, norepinephrine, and dopamine reuptake inhibitor indicated for managing obesity—has been reported effective in BED in a 12-week, randomized, double-blind, placebo-controlled trial. A 15-mg/d dosage reduced binge frequency, body weight, and depressive symptoms more effectively than placebo in 60 obese patients with BED.30 Most-frequent adverse effects (dry mouth and constipation) were mild and benign, and no significant complications were observed.

Sibutramine’s mechanism of action in BED is unknown. However, it suppressed food intake during binge-eating episodes in patients with BED in a randomized, controlled, cross-over laboratory study.31

Orlistat. We know of no published controlled studies of the lipase inhibitor orlistat in treating BED. In our experience, some patients do well with this agent, though we have observed infrequent purging episodes with it in patients with BED.

With orlistat, 120 mg tid, our BED patients have experienced weight loss comparable to that seen in uncomplicated obesity at similar dosages. Orlistat seems most effective for:

  • patients whose binge eating is in remission
  • those who responded to behavioral weightloss treatment, a psychological treatment, or another medication.

Anticonvulsants such as topiramate and zonisamide have been shown effective in treating obesity32,33 and are sometimes used to treat BED. Obese BED patients with mood disorders often do best with psychotherapy plus medication

Topiramate at dosages of 50 to 600 mg/d (median 212 mg/d) reduced binge-eating frequency, obsessive-compulsive features of binge eating, and body weight more effectively than placebo in a 14-week study of 61 obese patients with BED. These effects were maintained across 48 weeks in an open-label extension trial.34

Zonisamide, mean 513 mg/d, produced similar results during a prospective, open-label, 12-week trial in 15 patients with BED.35 A controlled trial to replicate these findings is ongoing.

BED may respond to anticonvulsant therapy for several reasons:

  • Some anticonvulsants are effective in treating bipolar disorder, which may occur with BED.12
  • Some anticonvulsants have shown benefit in conditions associated with pathologic impulsivity, such as substance abuse, impulse-control, and cluster B disorders.10

Growing evidence shows that bulimia nervosa and BED may be associated with pathologic impulsivity.

Combination therapies are generally more effective than monotherapies in patients with mood disorders, uncomplicated obesity, and possibly bulimia nervosa. Even so, few trials have systematically studied combination therapy in managing patients with BED.

Two studies compared psychotherapy and antidepressants alone and in combination in treating BED.21,23 Both showed that CBT alone was more effective in decreasing binge frequency than desipramine alone,21 fluoxetine alone,23 and the combination of CBT and medication. On the other hand, patients who took desipramine either alone or in combination experienced a greater degree of weight loss than those who did not take desipramine.21

In another combination therapy, exercise has been shown to be an effective adjunct to CBT in maintenance treatment of obese women with BED.36

No studies have compared behavioral weight management or a specialized psychotherapy in combination with an antiobesity drug or a weight-loss anticonvulsant in treating BED.

Treatment recommendations

In our experience, BED patients—particularly those with obesity and psychopathology—often do best with some combination of psychological treatment and medication:

  • The psychological component may be behavioral weight-loss treatment, a specialized psychotherapy such as CBT or IPT, or some combination of behavioral weight-loss treatment and specialized psychotherapy.
  • The medication component may consist of an antidepressant, anticonvulsant, antiobesity drug, or multiple drugs (such as an SSRI or sibutramine with topiramate for BED with major depression, or topiramate with lithium for BED with bipolar disorder).

Although combination therapies may be optimal for some patients, this approach remains unproven in controlled trials.

Patient preference. In addition to comorbidities, patient preference is an important consideration when choosing BED treatments. We determine our patients’ preferences by educating them as much as possible about their options. We explain the benefits and weaknesses of all treatments and encourage them to participate in forming their individualized treatment plans.

Patients sometimes have strong treatment preferences. Some prefer psychological treatments, whereas others prefer medications. Working with patient preferences enhances treatment adherence. For example, patients who fail a preferred treatment are often more willing to adhere to another treatment modality about which they initially were skeptical.

 

 

Related resources

  • Bray GA, Bouchard C (eds). Handbook of obesity: clinical applications (2nd ed). New York, NY: Marcel Dekker, 2004.
  • Cooper Z, Zairburn CG, Hawker DM. Cognitive behavioral treatment of obesity. A clinician’s guide. New York: Guilford Press, 2003.
  • Carter WP, Hudson JI, Lalonde JK, et al. Pharmacologic treatment of binge eating disorder. Int J Eat Disord 2003;34(suppl):S74-88.

Drug brand names

  • Bupropion • Wellbutrin
  • Citalopram • Celexa
  • Desipramine • Norpramin
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Imipramine • Tofranil
  • Lamotrigine • Lamictal
  • Lithium • Eskalith, others
  • Orlistat • Xenical
  • Sertraline • Zoloft
  • Sibutramine • Meridia
  • Topiramate • Topamax
  • Venlafaxine • Effexor
  • Zonisamide • Zonegran

Disclosure

Dr. Kotwal receives grant support from Elan Corporation and is a speaker for Ortho-McNeil Pharmaceutical and Pfizer Inc.

Dr. Kaneria and Ms. Guerdjikova report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. McElroy is a consultant to Abbott Laboratories, Bristol-Myers Squibb Co., Elan Corporation, GlaxoSmithKline, Janssen Pharmaceutica, Eli Lilly and Co., and Ortho-McNeil Pharmaceutical. She receives grant/research support from Elan Pharmaceuticals, Forest Pharmaceuticals, Merck & Co., Ortho-McNeil Pharmaceutical, and Sanofi-Synthelabo and is a speaker for Eli Lilly and Co. and Ortho-McNeil Pharmaceutical.

References

1. Pope HG, Hudson JI. Bulimia nervosa: Persistent disorders requires equally persistent treatment. Current Psychiatry 2004;3(1):13-22.

2. Halmi KA. Anorexia nervosa: Dual therapy can bring patients back from the brink. Current Psychiatry 2004;3(3):39-56.

3. American Psychiatric Association. Diagnostic and statistical manual of mental disorders (4th ed). Washington, DC: American Psychiatric Association, 1994.

4. Agras WS. Treatment of binge eating disorder. In: Gabbard GO (ed). Treatments of psychiatric disorders (3rd ed). Washington, DC: American Psychiatric Press, 2001;2209-19.

5. de Zwaan M. Binge eating disorder and obesity. Int J Obes Relat Metab Disord 2001;25(suppl 1):S51-5.

6. Dingemans AE, Bruna MJ, van Furth EF. Binge eating disorder: a review. Int J Obes Relat Metab Disord 2002;26:299-307.

7. Fairburn CG, Harrison PJ. Eating disorders. Lancet 2003;361:407-16.

8. Walsh BT (ed). The current status of binge eating disorder. Int J Eat Disord 2003;34(suppl):S1-120.

9. Devlin MJ, Goldfein JA, Dobrow I. What is this thing called BED? Current status of binge eating disorder nosology. Int J Eat Disord 2003;34(suppl):S2-18.

10. McElroy SL, Kotwal R. Binge eating. In: Hollander E, Stein D (eds). Handbook of impulse control disorders Washington, DC: American Psychiatric Press (in press).

11. Smith DE, Marcus MD, Lewis CE, et al. Prevalence of binge eating disorder, obesity, and depression in a biracial cohort of young adults. Ann Behav Med 1998;20:227-32.

12. Kruger S, Shugar G, Cooke RG. Comorbidity of binge eating disorder and the partial binge eating syndrome with bipolar disorder. Int J Eat Disord 1996;19:45-52.

13. Fairburn CG, Cooper Z, Doll H, et al. The natural course of bulimia nervosa and binge eating disorder in young women. Arch Gen Psychiatry 2000;57:659-65.

14. Striegel-Moore RH, Franko DL. Epidemiology of binge eating disorder. Int J Eat Disord 2003;34(suppl):S19-29.

15. Tammela LI, Rissanen A, Kuikka JT, et al. Treatment improves serotonin transporter binding and reduces binge eating. Psychopharmacology (Berl) 2003;170:89-93.

16. Wang GJ, Volkow ND, Logan J, et al. Brain dopamine and obesity. Lancet 2001;357:354-7.

17. Branson R, Potoczna N, Kral JG, et al. Binge eating as a major phenotype of melanocortin 4 receptor gene mutations. N Engl J Med 2003;348:1096-103.

18. Fairburn CG, Doll HA, Welch SL, et al. Risk factors for binge eating disorder: a community-based, case-control study. Arch Gen Psychiatry 1998;55:425-32.

19. Striegel-Moore RH, Dohm FA, Pike KM, et al. Abuse, bullying, and discrimination as risk factors for binge eating disorder. Am J Psychiatry 2002;159:1902-7.

20. Wonderlich SA, de Zwaan M, Mitchell JE, et al. Psychological and dietary treatments of binge eating disorder: conceptual implications. Int J Eat Disord 2003;34(suppl):S58-78.

21. Agras WS, Telch DF, Arnow B, et al. Weight loss, cognitive-behavioral, and desipramine treatments in binge eating disorder. An additive design. Behav Ther 1994;25:225-38.

22. Wilfley DE, Welch RR, Stein RI, et al. A randomized comparison of group cognitive-behavioral therapy and group interpersonal psychotherapy for the treatment of overweight individuals with binge eating disorder. Arch Gen Psychiatry 2002;59:713-21.

23. Grilo CM. A controlled study of cognitive behavioral therapy and fluoxetine for binge eating disorder (presentation) Charleston, SC: Eating Disorders Research Society annual meeting, 2002.

24. Telch CF, Agras WS, Linehan MM. Dialectical behavior therapy for binge eating disorder. J Consult Clin Psychol 2001;69:1061-5.

25. Carter WP, Hudson JI, Lalonde JK, et al. Pharmacologic treatment of binge eating disorder. Int J Eat Disord 2003;34(suppl):S74-88.

26. Appolinario JC, McElroy SL. Pharmacologic approaches in the treatment of binge eating disorder. Curr Drug Targets (in press).

27. Malhotra S, King KH, Welge JA, et al. Venlafaxine treatment of binge-eating disorder associated with obesity: a series of 35 patients. J Clin Psychiatry 2002;63:802-6.

28. Anderson JW, Greenway FL, Fujioka K, et al. Bupropion SR enhances weight loss: a 48-week double-blind, placebo-controlled trial. Obes Res 2002;10:633-41.

29. McElroy SL, Kotwal R, Malhotra S, et al. Are mood disorders and obesity related? A review for the mental health professional. J Clin Psychiatry (in press).

30. Appolinario JC, Bacaltchuk J, Sichieri R, et al. A randomized, double-blind, placebo-controlled study of sibutramine in the treatment of binge-eating disorder. Arch Gen Psychiatry 2003;60:1109-16.

31. Mitchell JE, Gosnell BA, Roerig JL, et al. Effects of sibutramine on binge eating, hunger, and fullness in a laboratory human feeding paradigm. Obes Res 2003;11:599-602.

32. Bray GA, Hollander P, Klein S, et al. A 6-month randomized, placebo-controlled, dose-ranging trial of topiramate for weight loss in obesity. Obes Res 2003;11:722-33.

33. Gadde KM, Franciscy DM, Wagner HR, 2nd, Krishnan KR. Zonisamide for weight loss in obese adults: a randomized controlled trial. JAMA 2003;289:1820-5.

34. McElroy SL, Arnold LM, Shapira NA, et al. Topiramate in the treatment of binge eating disorder associated with obesity: a randomized, placebo-controlled trial. Am J Psychiatry 2003;160:255-61.

35. McElroy SL, Kotwal R, Hudson JI, et al. Zonisamide in the treatment of binge-eating disorder: an open-label, prospective trial. J Clin Psychiatry 2004;65:50-6.

36. Pendleton VR, Goodrick GK, Poston WSC, et al. Exercise augments the effects of cognitive-behavioral therapy in the treatment of binge eating. Int J Eat Disord 2002;31(2):172-84.

References

1. Pope HG, Hudson JI. Bulimia nervosa: Persistent disorders requires equally persistent treatment. Current Psychiatry 2004;3(1):13-22.

2. Halmi KA. Anorexia nervosa: Dual therapy can bring patients back from the brink. Current Psychiatry 2004;3(3):39-56.

3. American Psychiatric Association. Diagnostic and statistical manual of mental disorders (4th ed). Washington, DC: American Psychiatric Association, 1994.

4. Agras WS. Treatment of binge eating disorder. In: Gabbard GO (ed). Treatments of psychiatric disorders (3rd ed). Washington, DC: American Psychiatric Press, 2001;2209-19.

5. de Zwaan M. Binge eating disorder and obesity. Int J Obes Relat Metab Disord 2001;25(suppl 1):S51-5.

6. Dingemans AE, Bruna MJ, van Furth EF. Binge eating disorder: a review. Int J Obes Relat Metab Disord 2002;26:299-307.

7. Fairburn CG, Harrison PJ. Eating disorders. Lancet 2003;361:407-16.

8. Walsh BT (ed). The current status of binge eating disorder. Int J Eat Disord 2003;34(suppl):S1-120.

9. Devlin MJ, Goldfein JA, Dobrow I. What is this thing called BED? Current status of binge eating disorder nosology. Int J Eat Disord 2003;34(suppl):S2-18.

10. McElroy SL, Kotwal R. Binge eating. In: Hollander E, Stein D (eds). Handbook of impulse control disorders Washington, DC: American Psychiatric Press (in press).

11. Smith DE, Marcus MD, Lewis CE, et al. Prevalence of binge eating disorder, obesity, and depression in a biracial cohort of young adults. Ann Behav Med 1998;20:227-32.

12. Kruger S, Shugar G, Cooke RG. Comorbidity of binge eating disorder and the partial binge eating syndrome with bipolar disorder. Int J Eat Disord 1996;19:45-52.

13. Fairburn CG, Cooper Z, Doll H, et al. The natural course of bulimia nervosa and binge eating disorder in young women. Arch Gen Psychiatry 2000;57:659-65.

14. Striegel-Moore RH, Franko DL. Epidemiology of binge eating disorder. Int J Eat Disord 2003;34(suppl):S19-29.

15. Tammela LI, Rissanen A, Kuikka JT, et al. Treatment improves serotonin transporter binding and reduces binge eating. Psychopharmacology (Berl) 2003;170:89-93.

16. Wang GJ, Volkow ND, Logan J, et al. Brain dopamine and obesity. Lancet 2001;357:354-7.

17. Branson R, Potoczna N, Kral JG, et al. Binge eating as a major phenotype of melanocortin 4 receptor gene mutations. N Engl J Med 2003;348:1096-103.

18. Fairburn CG, Doll HA, Welch SL, et al. Risk factors for binge eating disorder: a community-based, case-control study. Arch Gen Psychiatry 1998;55:425-32.

19. Striegel-Moore RH, Dohm FA, Pike KM, et al. Abuse, bullying, and discrimination as risk factors for binge eating disorder. Am J Psychiatry 2002;159:1902-7.

20. Wonderlich SA, de Zwaan M, Mitchell JE, et al. Psychological and dietary treatments of binge eating disorder: conceptual implications. Int J Eat Disord 2003;34(suppl):S58-78.

21. Agras WS, Telch DF, Arnow B, et al. Weight loss, cognitive-behavioral, and desipramine treatments in binge eating disorder. An additive design. Behav Ther 1994;25:225-38.

22. Wilfley DE, Welch RR, Stein RI, et al. A randomized comparison of group cognitive-behavioral therapy and group interpersonal psychotherapy for the treatment of overweight individuals with binge eating disorder. Arch Gen Psychiatry 2002;59:713-21.

23. Grilo CM. A controlled study of cognitive behavioral therapy and fluoxetine for binge eating disorder (presentation) Charleston, SC: Eating Disorders Research Society annual meeting, 2002.

24. Telch CF, Agras WS, Linehan MM. Dialectical behavior therapy for binge eating disorder. J Consult Clin Psychol 2001;69:1061-5.

25. Carter WP, Hudson JI, Lalonde JK, et al. Pharmacologic treatment of binge eating disorder. Int J Eat Disord 2003;34(suppl):S74-88.

26. Appolinario JC, McElroy SL. Pharmacologic approaches in the treatment of binge eating disorder. Curr Drug Targets (in press).

27. Malhotra S, King KH, Welge JA, et al. Venlafaxine treatment of binge-eating disorder associated with obesity: a series of 35 patients. J Clin Psychiatry 2002;63:802-6.

28. Anderson JW, Greenway FL, Fujioka K, et al. Bupropion SR enhances weight loss: a 48-week double-blind, placebo-controlled trial. Obes Res 2002;10:633-41.

29. McElroy SL, Kotwal R, Malhotra S, et al. Are mood disorders and obesity related? A review for the mental health professional. J Clin Psychiatry (in press).

30. Appolinario JC, Bacaltchuk J, Sichieri R, et al. A randomized, double-blind, placebo-controlled study of sibutramine in the treatment of binge-eating disorder. Arch Gen Psychiatry 2003;60:1109-16.

31. Mitchell JE, Gosnell BA, Roerig JL, et al. Effects of sibutramine on binge eating, hunger, and fullness in a laboratory human feeding paradigm. Obes Res 2003;11:599-602.

32. Bray GA, Hollander P, Klein S, et al. A 6-month randomized, placebo-controlled, dose-ranging trial of topiramate for weight loss in obesity. Obes Res 2003;11:722-33.

33. Gadde KM, Franciscy DM, Wagner HR, 2nd, Krishnan KR. Zonisamide for weight loss in obese adults: a randomized controlled trial. JAMA 2003;289:1820-5.

34. McElroy SL, Arnold LM, Shapira NA, et al. Topiramate in the treatment of binge eating disorder associated with obesity: a randomized, placebo-controlled trial. Am J Psychiatry 2003;160:255-61.

35. McElroy SL, Kotwal R, Hudson JI, et al. Zonisamide in the treatment of binge-eating disorder: an open-label, prospective trial. J Clin Psychiatry 2004;65:50-6.

36. Pendleton VR, Goodrick GK, Poston WSC, et al. Exercise augments the effects of cognitive-behavioral therapy in the treatment of binge eating. Int J Eat Disord 2002;31(2):172-84.

Issue
Current Psychiatry - 03(04)
Issue
Current Psychiatry - 03(04)
Page Number
31-47
Page Number
31-47
Publications
Publications
Topics
Article Type
Display Headline
Update on eating disorders: Binge-eating disorder
Display Headline
Update on eating disorders: Binge-eating disorder
Sections
Article Source

PURLs Copyright

Inside the Article

Update on eating disorders Anorexia nervosa: Dual therapy can bring patients back from the brink

Article Type
Changed
Tue, 12/11/2018 - 15:10
Display Headline
Update on eating disorders Anorexia nervosa: Dual therapy can bring patients back from the brink

Ms. J started losing weight deliberately at age 14 while attending boarding school. She lost 25 lbs by jogging 6 miles per day, exercising another 2 hours, avoiding meat, abusing laxatives, and drinking large quantities of coffee.

She was referred to a school counselor because of her weight loss and returned home. She was happier at a local high school and recovered to normal weight. In college, however, she reverted to compulsive exercising and preoccupation with her weight after the break-up of her first intimate relationship.

Now at age 22, Ms. J has persistently failed to gain weight during outpatient therapy for anorexia nervosa. At 5′7″ she weighs 98 lbs. On the day she was to be hospitalized involuntarily, she took 25 diphenhydramine tablets, which her psychiatrist viewed as a suicide threat. The overdose was treated in the emergency room with ipecac syrup, and she was admitted for inpatient eating disorder treatment.

Like Ms. J, patients with anorexia nervosa resist treatment and deny having most diagnostic signs and symptoms. Based on the evidence and my 30 years of treating anorectic patients,1 this article offers suggestions to help you:

  • gather accurate histories from patients and their families
  • identify common psychiatric comorbidities
  • gain the patient’s trust during treatment
  • provide effective dual therapy, with cognitive-behavioral and pharmacologic components.

Table 1

Diagnostic criteria for anorexia nervosa

Underweight (<85% of normal for age and height)
Fear of gaining weight or becoming fat, even though underweight
Disturbed conceptualization of body shape and weight, denial of seriousness of low body weight, or overemphasis on body shape and weight in self-evaluation
Amenorrhea. Subtypes:
  • Restricting type (does not binge or purge)
  • Binge-eating/purging type
Source: Adapted with permission from Diagnostic and statistical manual of mental disorders (4th ed. text revision).
Copyright 2000 American Psychiatric Association.

Making the diagnosis

Anorexia nervosa is characterized by underweight, fear of gaining weight, disturbed body concept, and amenorrhea (Table 1). Its core psychological symptoms have been described as:

  • relentless pursuit of thinness
  • denial of cachexia
  • and feelings of general ineffectiveness.2

The patient may say she feels fat even though emaciated or that parts of her body are too large. This disturbed experience of body weight or shape may represent sublimation and displacement for feelings of inadequacy. Because anorectic patients stay thin so effectively, they may feel a sense of accomplishment by evaluating themselves in terms of their thinness. Cognitive therapy focuses on correcting patients’ pervasive sense of inadequacy, as manifest in maturity fears and lack of confidence in coping with life’s problems.3

Subtypes. Anorexia nervosa has two subtypes—restricting and binge eating/purging—that differ in behavioral and medical symptoms.4 Patients with binge eating/purging show:

  • higher rates of impulsivity (suicide attempts, self-mutilation, stealing, and alcohol and other substance abuse)
  • more-prevalent impulsive personality disorders (borderline personality disorder, hysterical personality disorder)
  • medical problems caused by purging.

Restricting-type patients are often dependent and submissive, with difficulty separating from parents. These patients may be preoccupied with orderliness, perfectionism, and control.

Recommendation. A structured interview to diagnose anorexia nervosa is summarized in Table 2. Because the patient will likely deny her symptoms, it is usually necessary to also interview family members or close friends.

Psychiatric comorbidity

Case report continued: A ‘perfectionist.’

School for Ms. J required great effort, and she spent many hours studying. Her upper-middle-class parents described her as “a perfectionist.” The family placed considerable emphasis on doing the “correct” thing.

During adolescence, Ms. J developed a major depressive episode that lasted 4 months. She also developed obsessions and compulsions unrelated to her eating disorder. She obsessively ruminated about the correct things to say in social circumstances and devoted 4 hours per day to cleaning and checking compulsions. She felt she had to wash her car every time before going out; if she could not, she would cancel her social plans.

Table 2

Diagnosis of anorexia nervosa: Questions to ask*

Weight history-What was her highest weight and lowest weight (after weight loss)
-At what ages did these weights occur?
-Ask about her present weight before you weigh her
Eating behavior-What does she eat and when from morning awakening to bedtime?
-Does she eat with the family less often than in the past?
-Is she binging?
Purging behavior-Is she inducing vomiting?
-Is she using laxatives, diuretics, ipecac, or enemas?
Preoccupations and rituals concerning food and weight-Does she constantly count calories and express concern about fat content in foods?
-Does she often gaze in the mirror and comment about being fat?
-How often does she weigh herself?
-Does she express fear of being unable to stop eating?
Activity-Is she jogging, bike riding, or doing aerobics?
-How often, and for how long?
-Is she overactive at home, such as pacing?
Menstrual history-At what age did menses begin?
-What was the date of her last period?
-How regular is her cycle?
Psychiatric comorbidity-Does she have symptoms of depression?
-Impulsive behavior (suicide attempts or self-mutilation)?
-Drug or alcohol abuse?
-Anxiety (obsessive-compulsive behaviors, social phobia, generalized anxiety, fearfulness)?
-Personality disorders?
* Because patients with anorexia nervosa often deny their symptoms and conceal their food intake, it is usually necessary to interview family members or close friends as well as the patient.
 

 

In college, she began abusing alcohol and was arrested once for driving while intoxicated.

Depression is the most common comorbidity in anorexia nervosa. Two-thirds of anorectic patients in a 10-year follow-up study reported a history of major depressive disorder.5 Suicide, starvation, and electrolyte imbalance are the three major causes of death. Among severely ill patients who require hospitalization, 10% to 20% die, though the suicide rate is undocumented.

Compulsions. Anorectics’ preoccupations about food and eating rituals have been compared with compulsions, though less than 20% of patients meet diagnostic criteria for obsessive-compulsive disorder.6

Substance abuse. Bulimic anorectics report more alcohol and substance use and abuse than restricting anorectics.7 The most common substances of abuse are cannabis, cocaine, stimulants, and over-the-counter pills such as diet aids.

Personality disorders. Up to 50% of patients with anorexia nervosa—particularly the binge/purge subtype—have personality disorders. Borderline personality disorder is especially common among binge/purge types,8 and avoidant personality disorder is more common among restricting types.

Table 3

Diagnostic signs of emaciation and purging in patients with anorexia nervosa

Emaciation
  • Dry, cracking skin
  • Lanugo hair
  • Bradycardia
  • Hypotension
  • Leukopenia with relative lymphocytosis
  • Anemia
  • Hypercholesterolemia
  • Reduced bone density
Purging
  • Calluses on dorsum of hand, produced by hand friction from self-induced vomiting
  • Perioral dermatitis
  • Enlarged parotid glands (“chipmunk” face)
  • Tooth enamel erosion, caries, periodontitis
  • Cardiac arrhythmias (hypokalemia from purging)
  • Hypochloremic metabolic alkalosis
  • Hyperamylasemia
  • QT interval and T-wave changes on ECG

Personality disorders usually reflect instability in interpersonal relationships, poor self-image, or fluctuating affect. Patients may show a pattern of social inhibition, feelings of inadequacy, and hypersensitivity to negative evaluation.

Sexuality. Psychosocial and sexual development is often delayed in adolescent anorectics. In adults, interest in sex often plummets with anorexia onset, although binge/purge-type patients occasionally become promiscuous.

Medical signs

Case report continued: Abnormal ECG.

Ms. J was hospitalized after her weight dropped below 75% of normal for her age, height, and body build. She showed signs of electrolyte disturbance, including severe bradycardia (pulse rate 40) and ST-segment abnormalities on ECG.

Clinical signs of emaciation and purging can assist with diagnosis and in making decisions about medical treatment, including hospitalization (Table 3). Patients who purge are often weak and have puffy cheeks or parotid gland enlargement. They may have fainting spells and scars on their hands from stimulating vomiting. Laxative abuse may decrease colon motility and worsen constipation.

Neuroendocrine changes secondary to dieting and weight loss include:

  • increased corticotropin-releasing hormone secretion
  • blunted diurnal cortisol fluctuation
  • decreased follicle-stimulating hormone (FSH) secretion
  • impaired growth hormone regulation
  • decreased luteinizing hormone (LH) secretion
  • mildly decreased triiodothyronine
  • erratic vasopressin secretion.

Measuring these changes is unnecessary, as general nutritional rehabilitation with weight gain will correct them.

Neurotransmitter function. Emaciated anorectics have a blunted response to pharmacologic probes for dopamine, reduced CSF norepinephrine turnover, and decreased CSF serotonin. Neuroimaging studies suggest that serotonin dysfunction may persist after weight is restored, although these findings require replication.

Treatment priorities

Effective therapies. Open studies indicate that multidimensional treatment—medical management, psychoeducation, and individual cognitive-behavioral therapy (CBT)—is most effective for anorexia nervosa. The fewer than 10 controlled trials that address anorexia nervosa treatment show:

  • the more severe the illness, the more intense the treatment required
  • outpatient therapy is most successful in patients who have had the illness <6 months, are not binging and vomiting, and have parents who participate in family therapy.

Hospitalization. An emaciated patient who is irritable, depressed, preoccupied with food, and sleep-deprived is unlikely to make progress toward behavioral change. The first goal, therefore, is to restore her nutritional state to normal.

Severely ill anorectic patients require hospitalization for daily monitoring of weight, calorie intake, urine output, and serum electrolytes and amylase (to assess purging behavior). Hospitalization is indicated for:

  • loss of >20% of normal weight for age, height, and bone structure
  • >6 months of repeated hospitalizations and underweight
  • psychotic depression or serious suicide attempt
  • incapacitating obsessions and compulsions, related or not to the eating disorder
  • serious comorbid medical conditions, such as edema, hypoproteinemia, severe anemia, cardiac arrhythmia, or hypokalemic alkalosis (serum K+ < 2.5 mEq/L).

Keeping a patient in the hospital long enough to provide effective medical and psychological therapy has become difficult, however, because of medical insurance restrictions (Box). The result: poorer outcomes and increased relapse rates compared with 10 years ago.9-12

Box

Shorter hospitalizations, worse outcomes for patients with eating disorders

Hospital treatment of eating disorders has shifted from long-term care of a chronic disorder to stabilization of acute episodes. For some patients, this change has been deleterious and not cost-effective.

A decade ago, eating disorder hospitalizations were covered primarily by private insurance. Today, health maintenance organizations, managed care oversight of private insurance, and public funding are the primary sources of payment. These insurers often limit payment for eating disorder hospitalization, the most costly aspect of psychiatric care.

Poor outcomes and a high relapse rate have been documented in anorexia nervosa patients who left the hospital while underweight.9-11 From 1984 to 1998:

  • average hospital stays for anorexia nervosa decreased from 150 days to 23.7 days
  • readmissions increased from 0% to 27% of total admissions
  • anorectic patients’ average body mass index at discharge dropped from 19.3 to 17.7, a statistically significant difference.12

For psychiatrists, this trend means many outpatients with anorexia nervosa will require repeated hospitalizations that will not substantially improve their anorectic behaviors.

 

 

Nutritional rehabilitation and behavior changes can often correct the medical complications of emaciation and purging. Lost bone density is seldom restored, but nutritional rehabilitation can prevent further bone loss.13 Women who remain amenorrheic for several years after weight restoration tend to be more psychologically disturbed than those who resume menses rapidly.14

Cognitive-behavioral therapy

Other authors have discussed CBT for anorexia nervosa.3,15 In general, the key tasks—operationalizing beliefs, evaluating autonomic thoughts, testing prospective hypotheses, and examining underlying assumptions—are accomplished by assessing anorexia’s distorted cognitions. No satisfactory controlled studies have examined any other type of individual psychotherapy for treating anorexia nervosa.

Alliance building. Patients with anorexia find it difficult to participate in therapeutic relationships. They are terrified of gaining weight and readily drop out of treatment. To build a therapeutic alliance:

  • begin by helping the patient develop a history of her significant life events
  • proceed slowly, praising her for every small attempt at changing her behavior
  • set realistic therapy goals, considering her degree of resistance.

Monitoring. Behavior therapy consists primarily of positive reinforcements for weight gain. For this, we weigh outpatients weekly and inpatients daily. Outpatients are taught to keep diaries of daily food intake, stressful events, and emotional responses to them. The therapist begins each session by examining the patient’s diary with her and discussing how life events affect her eating behavior.

Cognitive restructuring helps patients identify their disturbed cognitions and challenge core beliefs about self-image. In this process, they become aware of their negative thoughts and develop arguments and evidence to support and refute the thoughts’ validity. They then form a reasoned conclusion based on the evidence.

Even if patients do not accept this logical conclusion, we encourage them to behave as if they believe it to be true. By doing this repeatedly, they eventually obtain some symptom relief.

Response-prevention techniques can help stop binging and purging. For example, we may require inpatients to sit together for 1 hour after eating. Because most patients will not vomit in front of each other, they learn how to resist vomiting and eventually experience reduced anxiety without vomiting after a meal.

Problem solving helps patients to reason through difficult food-related or interpersonal situations. The patient states the problem, then generates as many solutions as possible with the therapist’s assistance. She chooses one solution and puts it into effect, usually for 1 week. She then discusses the results with her therapist and decides whether to try another solution.

Family therapy. A family analysis—including a brief psychiatric history and evaluation of interactions—is recommended for all patients who live at home. This analysis can help you decide what type of family therapy or counseling to recommend.

Some families respond well with the parents and patient together in therapy sessions, whereas others are more comfortable with separate counseling. In a recent controlled study, anorectic patients younger than age 18 did equally well whether they were counseled with the family or separately.16

Brief therapy sessions are sometimes the most effective method to address family issues. When this is not possible, you and the patient can discuss family relationships in individual therapy.

Medications

Many medications have been used to treat anorexia nervosa, though few randomized, placebo-controlled studies exist. Because evidence does not support using psychotropics as monotherapy for anorexia nervosa, medication is considered adjunctive to CBT.

Chlorpromazine can help the hospitalized, severely ill patient who is overwhelmed with uncontrollable behavioral rituals and thoughts of losing weight. This antipsychotic helps reduce anorectic preoccupations and anxiety and helps make patients more amenable to therapy.

Start chlorpromazine at 10 mg tid and increase gradually until the patient can eat without extreme anxiety. Usual maximum dosage is 50 mg tid. Monitor blood pressure, tardive dyskinesia, and decreased white blood cell count.

Olanzapine may help induce weight gain and reduce anxiety in anorectic patients.17 Controlled and open-label studies are under way.

We start olanzapine at 2.5 mg/d and increase gradually to 10 or 15 mg/d. At this dosage, patients’ anxiety about eating is usually substantially reduced. Sedation is the most common side effect.

Anorexia patients often refuse to take olanzapine for fear of weight gain. If a patient’s emaciation is life-threatening, we may seek court permission to medicate her involuntarily. We reassure her that we will discontinue olanzapine when she reaches her target weight.

Serotonin in anorexia. Central serotonin pathways modulate feeding behavior. Serotonin antagonists—such as cyproheptadine—increase food intake and weight gain, whereas serotonin agonists—such as selective serotonin reuptake inhibitors (SSRIs)—decrease food intake.

Serotonin pathways also may modulate obsessive-compulsive and impulsive behaviors. Both serotonin agonists and antagonists can be useful adjuncts in treating anorexia nervosa.

 

 

In a double-blind, placebo-controlled trial, cyproheptadine, 4 to 8 mg tid, was associated with weight gain and reduced depressive symptoms in anorexia nervosa patients.18 Unlike tricyclic antidepressants, cyproheptadine does not reduce blood pressure or increase heart rate, which makes it attractive for emaciated anorectic patients. Dosages up to 28 mg/d can be used safely.

The SSRI fluoxetine may help prevent weight loss relapse in anorexia nervosa and reduce obsessive-compulsive behaviors.19 In open studies of low-weight anorectics, however, fluoxetine had little impact on weight or other clinically meaningful variables.20 Thus, this agent is recommended for preventing weight-loss relapse only in patients who are within 10% to 15% of ideal body weight.

Outpatient care

Case report continued: Ongoing therapy

During hospitalization, Ms. J participated in all therapeutic modalities but had difficulty eating enough to gain weight. She reached her target weight of 127 lbs in about 7 weeks but gained no sense of purpose in life.

She is starting an intensive outpatient program using CBT to maintain her weight and further address the core psychopathology of her illness. Her maintenance therapy includes attending Alcoholics Anonymous meetings, ongoing fluoxetine (20 mg/d) to prevent weight-loss relapse, and CBT for obsessions and compulsions not related to her eating disorder.

Related resources

  • Halmi KA. Eating disorders: Anorexia, bulimia nervosa and obesity. In: Hales RE, Yudofsky SC (eds). Textbook of clinical psychiatry, 4th ed. Washington, DC: American Psychiatric Publishing, 2003:1001-21.
  • Academy of Eating Disorders. www.aedweb.org
  • Anorexia Nervosa & Associated Disorders (ANAD). www.anad.org

Drug brand names

  • Chlorpromazine • Thorazine
  • Cyproheptadine • Periactin
  • Fluoxetine • Prozac
  • Olanzapine • Zyprexa

Disclosure

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

References

1. Halmi KA. Anorexia nervosa: demographic and clinical features in 94 cases. Psychosom Med 1974;36:18-24.

2. Bruch AH. Eating disorders: obesity, anorexia nervosa, and the person within. New York, Basic books, 1973.

3. Kleifield E, Wagner S, Halmi KA. Cognitive behavioral treatment of anorexia nervosa. Psychiatr Clin North Am 1996;19:715-34.

4. Halmi KA. Eating disorders. In: Martin A, Scahill L, Charney DS Leckman JF (eds). Pediatric pharmacology. New York: Oxford University Press, 2002;592-602.

5. Halmi KA, Eckert E, Marci P, Cohen J. Comorbidity of psychiatric diagnoses in anorexia nervosa. Arch Gen Psychiatry 1991;48:712-18.

6. Braun DL, Sunday SR, Halmi KA. Psychiatric comorbidity in patients with eating disorders. Psychol Med 1994;24:859-67.

7. Holderness CC, Brooks-Gunn J, Warren MP. Comorbidity of eating disorders and substance abuse review of the literature. Int J Eat Disord 1994;16:1-34.

8. Herzog DB, Keller M, Lavori P. The prevalence of personality disorders in 210 women with eating disorders. J Clin Psychiatry 1992;53:147-52.

9. Baran S, Weltzin T, Kaye W. Low discharge weight and outcome in anorexia nervosa. Am J Psychiatry 1995;150:1070-2.

10. Commerford MC, Licinio J, Halmi KA. Guidelines for discharging eating disorder patients. Eat Disord 1997;5:69-74.

11. Howard W, Evans K, Quinter-Howard C, et al. Predictors of success or failure of transition to day hospital treatment for inpatients with anorexia nervosa. Am J Psychiatry 1999;156:1697-1702.

12. Wiseman C, Sunday SR, Klapper F, et al. Changing patterns of hospitalization in eating disorder patients. Int J Eat Disord 2001;30:69-74.

13. Newman M, Halmi KA. The relationship of bone density to estradiol and cortisol in anorexia nervosa and bulimia nervosa. Psychiatr Res 1989;29:105-12.

14. Falk JR, Halmi KA. Amenorrhea in anorexia nervosa: examination of the critical body hypothesis. Biol Psychiatr 1982;17:799-806.

15. Garner DM, Bemis KM. A cognitive-behavioral approach to anorexia nervosa. Cognit Ther Res 1982;6:1223-50.

16. Dare C. Eisler. Family therapy and eating disorders. In: Fairburn CR, Brownell KD (eds). Eating disorders and obesity. New York: Guilford Press, 2002;314-19.

17. Powers P, Santana CA, Bannon YS. Olanzapine in the treatment of anorexia nervosa: an open label trial. Int J Eat Disord 2002;32:146-54.

18. Halmi KA, Eckert ED, Ladu T, Cohen J. Anorexia nervosa: treatment efficacy of cyproheptadine and amitriptyline. Arch Gen Psychiatry 1986;43:177-81.

19. Kaye W. The use of fluoxetine to prevent relapse in anorexia nervosa (presentation). Pittsburgh, PA: Eating Disorder Research Society annual meeting, 1996.

20. Ferguson C, Lavia M, Crossan P. Are serotonin selective reuptake inhibitors effective in underweight anorexia nervosa? Int J Eat Disord 1999;25:11-17.

Article PDF
Author and Disclosure Information

Paul E. Keck, Jr, MD
Katherine A. Halmi, MD
Professor, Department of psychiatry Weill Medical College of Cornell University Director, Eating Disorder Program New York Presbyterian Hospital, Westchester division

Issue
Current Psychiatry - 03(03)
Publications
Topics
Page Number
39-59
Sections
Author and Disclosure Information

Paul E. Keck, Jr, MD
Katherine A. Halmi, MD
Professor, Department of psychiatry Weill Medical College of Cornell University Director, Eating Disorder Program New York Presbyterian Hospital, Westchester division

Author and Disclosure Information

Paul E. Keck, Jr, MD
Katherine A. Halmi, MD
Professor, Department of psychiatry Weill Medical College of Cornell University Director, Eating Disorder Program New York Presbyterian Hospital, Westchester division

Article PDF
Article PDF

Ms. J started losing weight deliberately at age 14 while attending boarding school. She lost 25 lbs by jogging 6 miles per day, exercising another 2 hours, avoiding meat, abusing laxatives, and drinking large quantities of coffee.

She was referred to a school counselor because of her weight loss and returned home. She was happier at a local high school and recovered to normal weight. In college, however, she reverted to compulsive exercising and preoccupation with her weight after the break-up of her first intimate relationship.

Now at age 22, Ms. J has persistently failed to gain weight during outpatient therapy for anorexia nervosa. At 5′7″ she weighs 98 lbs. On the day she was to be hospitalized involuntarily, she took 25 diphenhydramine tablets, which her psychiatrist viewed as a suicide threat. The overdose was treated in the emergency room with ipecac syrup, and she was admitted for inpatient eating disorder treatment.

Like Ms. J, patients with anorexia nervosa resist treatment and deny having most diagnostic signs and symptoms. Based on the evidence and my 30 years of treating anorectic patients,1 this article offers suggestions to help you:

  • gather accurate histories from patients and their families
  • identify common psychiatric comorbidities
  • gain the patient’s trust during treatment
  • provide effective dual therapy, with cognitive-behavioral and pharmacologic components.

Table 1

Diagnostic criteria for anorexia nervosa

Underweight (<85% of normal for age and height)
Fear of gaining weight or becoming fat, even though underweight
Disturbed conceptualization of body shape and weight, denial of seriousness of low body weight, or overemphasis on body shape and weight in self-evaluation
Amenorrhea. Subtypes:
  • Restricting type (does not binge or purge)
  • Binge-eating/purging type
Source: Adapted with permission from Diagnostic and statistical manual of mental disorders (4th ed. text revision).
Copyright 2000 American Psychiatric Association.

Making the diagnosis

Anorexia nervosa is characterized by underweight, fear of gaining weight, disturbed body concept, and amenorrhea (Table 1). Its core psychological symptoms have been described as:

  • relentless pursuit of thinness
  • denial of cachexia
  • and feelings of general ineffectiveness.2

The patient may say she feels fat even though emaciated or that parts of her body are too large. This disturbed experience of body weight or shape may represent sublimation and displacement for feelings of inadequacy. Because anorectic patients stay thin so effectively, they may feel a sense of accomplishment by evaluating themselves in terms of their thinness. Cognitive therapy focuses on correcting patients’ pervasive sense of inadequacy, as manifest in maturity fears and lack of confidence in coping with life’s problems.3

Subtypes. Anorexia nervosa has two subtypes—restricting and binge eating/purging—that differ in behavioral and medical symptoms.4 Patients with binge eating/purging show:

  • higher rates of impulsivity (suicide attempts, self-mutilation, stealing, and alcohol and other substance abuse)
  • more-prevalent impulsive personality disorders (borderline personality disorder, hysterical personality disorder)
  • medical problems caused by purging.

Restricting-type patients are often dependent and submissive, with difficulty separating from parents. These patients may be preoccupied with orderliness, perfectionism, and control.

Recommendation. A structured interview to diagnose anorexia nervosa is summarized in Table 2. Because the patient will likely deny her symptoms, it is usually necessary to also interview family members or close friends.

Psychiatric comorbidity

Case report continued: A ‘perfectionist.’

School for Ms. J required great effort, and she spent many hours studying. Her upper-middle-class parents described her as “a perfectionist.” The family placed considerable emphasis on doing the “correct” thing.

During adolescence, Ms. J developed a major depressive episode that lasted 4 months. She also developed obsessions and compulsions unrelated to her eating disorder. She obsessively ruminated about the correct things to say in social circumstances and devoted 4 hours per day to cleaning and checking compulsions. She felt she had to wash her car every time before going out; if she could not, she would cancel her social plans.

Table 2

Diagnosis of anorexia nervosa: Questions to ask*

Weight history-What was her highest weight and lowest weight (after weight loss)
-At what ages did these weights occur?
-Ask about her present weight before you weigh her
Eating behavior-What does she eat and when from morning awakening to bedtime?
-Does she eat with the family less often than in the past?
-Is she binging?
Purging behavior-Is she inducing vomiting?
-Is she using laxatives, diuretics, ipecac, or enemas?
Preoccupations and rituals concerning food and weight-Does she constantly count calories and express concern about fat content in foods?
-Does she often gaze in the mirror and comment about being fat?
-How often does she weigh herself?
-Does she express fear of being unable to stop eating?
Activity-Is she jogging, bike riding, or doing aerobics?
-How often, and for how long?
-Is she overactive at home, such as pacing?
Menstrual history-At what age did menses begin?
-What was the date of her last period?
-How regular is her cycle?
Psychiatric comorbidity-Does she have symptoms of depression?
-Impulsive behavior (suicide attempts or self-mutilation)?
-Drug or alcohol abuse?
-Anxiety (obsessive-compulsive behaviors, social phobia, generalized anxiety, fearfulness)?
-Personality disorders?
* Because patients with anorexia nervosa often deny their symptoms and conceal their food intake, it is usually necessary to interview family members or close friends as well as the patient.
 

 

In college, she began abusing alcohol and was arrested once for driving while intoxicated.

Depression is the most common comorbidity in anorexia nervosa. Two-thirds of anorectic patients in a 10-year follow-up study reported a history of major depressive disorder.5 Suicide, starvation, and electrolyte imbalance are the three major causes of death. Among severely ill patients who require hospitalization, 10% to 20% die, though the suicide rate is undocumented.

Compulsions. Anorectics’ preoccupations about food and eating rituals have been compared with compulsions, though less than 20% of patients meet diagnostic criteria for obsessive-compulsive disorder.6

Substance abuse. Bulimic anorectics report more alcohol and substance use and abuse than restricting anorectics.7 The most common substances of abuse are cannabis, cocaine, stimulants, and over-the-counter pills such as diet aids.

Personality disorders. Up to 50% of patients with anorexia nervosa—particularly the binge/purge subtype—have personality disorders. Borderline personality disorder is especially common among binge/purge types,8 and avoidant personality disorder is more common among restricting types.

Table 3

Diagnostic signs of emaciation and purging in patients with anorexia nervosa

Emaciation
  • Dry, cracking skin
  • Lanugo hair
  • Bradycardia
  • Hypotension
  • Leukopenia with relative lymphocytosis
  • Anemia
  • Hypercholesterolemia
  • Reduced bone density
Purging
  • Calluses on dorsum of hand, produced by hand friction from self-induced vomiting
  • Perioral dermatitis
  • Enlarged parotid glands (“chipmunk” face)
  • Tooth enamel erosion, caries, periodontitis
  • Cardiac arrhythmias (hypokalemia from purging)
  • Hypochloremic metabolic alkalosis
  • Hyperamylasemia
  • QT interval and T-wave changes on ECG

Personality disorders usually reflect instability in interpersonal relationships, poor self-image, or fluctuating affect. Patients may show a pattern of social inhibition, feelings of inadequacy, and hypersensitivity to negative evaluation.

Sexuality. Psychosocial and sexual development is often delayed in adolescent anorectics. In adults, interest in sex often plummets with anorexia onset, although binge/purge-type patients occasionally become promiscuous.

Medical signs

Case report continued: Abnormal ECG.

Ms. J was hospitalized after her weight dropped below 75% of normal for her age, height, and body build. She showed signs of electrolyte disturbance, including severe bradycardia (pulse rate 40) and ST-segment abnormalities on ECG.

Clinical signs of emaciation and purging can assist with diagnosis and in making decisions about medical treatment, including hospitalization (Table 3). Patients who purge are often weak and have puffy cheeks or parotid gland enlargement. They may have fainting spells and scars on their hands from stimulating vomiting. Laxative abuse may decrease colon motility and worsen constipation.

Neuroendocrine changes secondary to dieting and weight loss include:

  • increased corticotropin-releasing hormone secretion
  • blunted diurnal cortisol fluctuation
  • decreased follicle-stimulating hormone (FSH) secretion
  • impaired growth hormone regulation
  • decreased luteinizing hormone (LH) secretion
  • mildly decreased triiodothyronine
  • erratic vasopressin secretion.

Measuring these changes is unnecessary, as general nutritional rehabilitation with weight gain will correct them.

Neurotransmitter function. Emaciated anorectics have a blunted response to pharmacologic probes for dopamine, reduced CSF norepinephrine turnover, and decreased CSF serotonin. Neuroimaging studies suggest that serotonin dysfunction may persist after weight is restored, although these findings require replication.

Treatment priorities

Effective therapies. Open studies indicate that multidimensional treatment—medical management, psychoeducation, and individual cognitive-behavioral therapy (CBT)—is most effective for anorexia nervosa. The fewer than 10 controlled trials that address anorexia nervosa treatment show:

  • the more severe the illness, the more intense the treatment required
  • outpatient therapy is most successful in patients who have had the illness <6 months, are not binging and vomiting, and have parents who participate in family therapy.

Hospitalization. An emaciated patient who is irritable, depressed, preoccupied with food, and sleep-deprived is unlikely to make progress toward behavioral change. The first goal, therefore, is to restore her nutritional state to normal.

Severely ill anorectic patients require hospitalization for daily monitoring of weight, calorie intake, urine output, and serum electrolytes and amylase (to assess purging behavior). Hospitalization is indicated for:

  • loss of >20% of normal weight for age, height, and bone structure
  • >6 months of repeated hospitalizations and underweight
  • psychotic depression or serious suicide attempt
  • incapacitating obsessions and compulsions, related or not to the eating disorder
  • serious comorbid medical conditions, such as edema, hypoproteinemia, severe anemia, cardiac arrhythmia, or hypokalemic alkalosis (serum K+ < 2.5 mEq/L).

Keeping a patient in the hospital long enough to provide effective medical and psychological therapy has become difficult, however, because of medical insurance restrictions (Box). The result: poorer outcomes and increased relapse rates compared with 10 years ago.9-12

Box

Shorter hospitalizations, worse outcomes for patients with eating disorders

Hospital treatment of eating disorders has shifted from long-term care of a chronic disorder to stabilization of acute episodes. For some patients, this change has been deleterious and not cost-effective.

A decade ago, eating disorder hospitalizations were covered primarily by private insurance. Today, health maintenance organizations, managed care oversight of private insurance, and public funding are the primary sources of payment. These insurers often limit payment for eating disorder hospitalization, the most costly aspect of psychiatric care.

Poor outcomes and a high relapse rate have been documented in anorexia nervosa patients who left the hospital while underweight.9-11 From 1984 to 1998:

  • average hospital stays for anorexia nervosa decreased from 150 days to 23.7 days
  • readmissions increased from 0% to 27% of total admissions
  • anorectic patients’ average body mass index at discharge dropped from 19.3 to 17.7, a statistically significant difference.12

For psychiatrists, this trend means many outpatients with anorexia nervosa will require repeated hospitalizations that will not substantially improve their anorectic behaviors.

 

 

Nutritional rehabilitation and behavior changes can often correct the medical complications of emaciation and purging. Lost bone density is seldom restored, but nutritional rehabilitation can prevent further bone loss.13 Women who remain amenorrheic for several years after weight restoration tend to be more psychologically disturbed than those who resume menses rapidly.14

Cognitive-behavioral therapy

Other authors have discussed CBT for anorexia nervosa.3,15 In general, the key tasks—operationalizing beliefs, evaluating autonomic thoughts, testing prospective hypotheses, and examining underlying assumptions—are accomplished by assessing anorexia’s distorted cognitions. No satisfactory controlled studies have examined any other type of individual psychotherapy for treating anorexia nervosa.

Alliance building. Patients with anorexia find it difficult to participate in therapeutic relationships. They are terrified of gaining weight and readily drop out of treatment. To build a therapeutic alliance:

  • begin by helping the patient develop a history of her significant life events
  • proceed slowly, praising her for every small attempt at changing her behavior
  • set realistic therapy goals, considering her degree of resistance.

Monitoring. Behavior therapy consists primarily of positive reinforcements for weight gain. For this, we weigh outpatients weekly and inpatients daily. Outpatients are taught to keep diaries of daily food intake, stressful events, and emotional responses to them. The therapist begins each session by examining the patient’s diary with her and discussing how life events affect her eating behavior.

Cognitive restructuring helps patients identify their disturbed cognitions and challenge core beliefs about self-image. In this process, they become aware of their negative thoughts and develop arguments and evidence to support and refute the thoughts’ validity. They then form a reasoned conclusion based on the evidence.

Even if patients do not accept this logical conclusion, we encourage them to behave as if they believe it to be true. By doing this repeatedly, they eventually obtain some symptom relief.

Response-prevention techniques can help stop binging and purging. For example, we may require inpatients to sit together for 1 hour after eating. Because most patients will not vomit in front of each other, they learn how to resist vomiting and eventually experience reduced anxiety without vomiting after a meal.

Problem solving helps patients to reason through difficult food-related or interpersonal situations. The patient states the problem, then generates as many solutions as possible with the therapist’s assistance. She chooses one solution and puts it into effect, usually for 1 week. She then discusses the results with her therapist and decides whether to try another solution.

Family therapy. A family analysis—including a brief psychiatric history and evaluation of interactions—is recommended for all patients who live at home. This analysis can help you decide what type of family therapy or counseling to recommend.

Some families respond well with the parents and patient together in therapy sessions, whereas others are more comfortable with separate counseling. In a recent controlled study, anorectic patients younger than age 18 did equally well whether they were counseled with the family or separately.16

Brief therapy sessions are sometimes the most effective method to address family issues. When this is not possible, you and the patient can discuss family relationships in individual therapy.

Medications

Many medications have been used to treat anorexia nervosa, though few randomized, placebo-controlled studies exist. Because evidence does not support using psychotropics as monotherapy for anorexia nervosa, medication is considered adjunctive to CBT.

Chlorpromazine can help the hospitalized, severely ill patient who is overwhelmed with uncontrollable behavioral rituals and thoughts of losing weight. This antipsychotic helps reduce anorectic preoccupations and anxiety and helps make patients more amenable to therapy.

Start chlorpromazine at 10 mg tid and increase gradually until the patient can eat without extreme anxiety. Usual maximum dosage is 50 mg tid. Monitor blood pressure, tardive dyskinesia, and decreased white blood cell count.

Olanzapine may help induce weight gain and reduce anxiety in anorectic patients.17 Controlled and open-label studies are under way.

We start olanzapine at 2.5 mg/d and increase gradually to 10 or 15 mg/d. At this dosage, patients’ anxiety about eating is usually substantially reduced. Sedation is the most common side effect.

Anorexia patients often refuse to take olanzapine for fear of weight gain. If a patient’s emaciation is life-threatening, we may seek court permission to medicate her involuntarily. We reassure her that we will discontinue olanzapine when she reaches her target weight.

Serotonin in anorexia. Central serotonin pathways modulate feeding behavior. Serotonin antagonists—such as cyproheptadine—increase food intake and weight gain, whereas serotonin agonists—such as selective serotonin reuptake inhibitors (SSRIs)—decrease food intake.

Serotonin pathways also may modulate obsessive-compulsive and impulsive behaviors. Both serotonin agonists and antagonists can be useful adjuncts in treating anorexia nervosa.

 

 

In a double-blind, placebo-controlled trial, cyproheptadine, 4 to 8 mg tid, was associated with weight gain and reduced depressive symptoms in anorexia nervosa patients.18 Unlike tricyclic antidepressants, cyproheptadine does not reduce blood pressure or increase heart rate, which makes it attractive for emaciated anorectic patients. Dosages up to 28 mg/d can be used safely.

The SSRI fluoxetine may help prevent weight loss relapse in anorexia nervosa and reduce obsessive-compulsive behaviors.19 In open studies of low-weight anorectics, however, fluoxetine had little impact on weight or other clinically meaningful variables.20 Thus, this agent is recommended for preventing weight-loss relapse only in patients who are within 10% to 15% of ideal body weight.

Outpatient care

Case report continued: Ongoing therapy

During hospitalization, Ms. J participated in all therapeutic modalities but had difficulty eating enough to gain weight. She reached her target weight of 127 lbs in about 7 weeks but gained no sense of purpose in life.

She is starting an intensive outpatient program using CBT to maintain her weight and further address the core psychopathology of her illness. Her maintenance therapy includes attending Alcoholics Anonymous meetings, ongoing fluoxetine (20 mg/d) to prevent weight-loss relapse, and CBT for obsessions and compulsions not related to her eating disorder.

Related resources

  • Halmi KA. Eating disorders: Anorexia, bulimia nervosa and obesity. In: Hales RE, Yudofsky SC (eds). Textbook of clinical psychiatry, 4th ed. Washington, DC: American Psychiatric Publishing, 2003:1001-21.
  • Academy of Eating Disorders. www.aedweb.org
  • Anorexia Nervosa & Associated Disorders (ANAD). www.anad.org

Drug brand names

  • Chlorpromazine • Thorazine
  • Cyproheptadine • Periactin
  • Fluoxetine • Prozac
  • Olanzapine • Zyprexa

Disclosure

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

Ms. J started losing weight deliberately at age 14 while attending boarding school. She lost 25 lbs by jogging 6 miles per day, exercising another 2 hours, avoiding meat, abusing laxatives, and drinking large quantities of coffee.

She was referred to a school counselor because of her weight loss and returned home. She was happier at a local high school and recovered to normal weight. In college, however, she reverted to compulsive exercising and preoccupation with her weight after the break-up of her first intimate relationship.

Now at age 22, Ms. J has persistently failed to gain weight during outpatient therapy for anorexia nervosa. At 5′7″ she weighs 98 lbs. On the day she was to be hospitalized involuntarily, she took 25 diphenhydramine tablets, which her psychiatrist viewed as a suicide threat. The overdose was treated in the emergency room with ipecac syrup, and she was admitted for inpatient eating disorder treatment.

Like Ms. J, patients with anorexia nervosa resist treatment and deny having most diagnostic signs and symptoms. Based on the evidence and my 30 years of treating anorectic patients,1 this article offers suggestions to help you:

  • gather accurate histories from patients and their families
  • identify common psychiatric comorbidities
  • gain the patient’s trust during treatment
  • provide effective dual therapy, with cognitive-behavioral and pharmacologic components.

Table 1

Diagnostic criteria for anorexia nervosa

Underweight (<85% of normal for age and height)
Fear of gaining weight or becoming fat, even though underweight
Disturbed conceptualization of body shape and weight, denial of seriousness of low body weight, or overemphasis on body shape and weight in self-evaluation
Amenorrhea. Subtypes:
  • Restricting type (does not binge or purge)
  • Binge-eating/purging type
Source: Adapted with permission from Diagnostic and statistical manual of mental disorders (4th ed. text revision).
Copyright 2000 American Psychiatric Association.

Making the diagnosis

Anorexia nervosa is characterized by underweight, fear of gaining weight, disturbed body concept, and amenorrhea (Table 1). Its core psychological symptoms have been described as:

  • relentless pursuit of thinness
  • denial of cachexia
  • and feelings of general ineffectiveness.2

The patient may say she feels fat even though emaciated or that parts of her body are too large. This disturbed experience of body weight or shape may represent sublimation and displacement for feelings of inadequacy. Because anorectic patients stay thin so effectively, they may feel a sense of accomplishment by evaluating themselves in terms of their thinness. Cognitive therapy focuses on correcting patients’ pervasive sense of inadequacy, as manifest in maturity fears and lack of confidence in coping with life’s problems.3

Subtypes. Anorexia nervosa has two subtypes—restricting and binge eating/purging—that differ in behavioral and medical symptoms.4 Patients with binge eating/purging show:

  • higher rates of impulsivity (suicide attempts, self-mutilation, stealing, and alcohol and other substance abuse)
  • more-prevalent impulsive personality disorders (borderline personality disorder, hysterical personality disorder)
  • medical problems caused by purging.

Restricting-type patients are often dependent and submissive, with difficulty separating from parents. These patients may be preoccupied with orderliness, perfectionism, and control.

Recommendation. A structured interview to diagnose anorexia nervosa is summarized in Table 2. Because the patient will likely deny her symptoms, it is usually necessary to also interview family members or close friends.

Psychiatric comorbidity

Case report continued: A ‘perfectionist.’

School for Ms. J required great effort, and she spent many hours studying. Her upper-middle-class parents described her as “a perfectionist.” The family placed considerable emphasis on doing the “correct” thing.

During adolescence, Ms. J developed a major depressive episode that lasted 4 months. She also developed obsessions and compulsions unrelated to her eating disorder. She obsessively ruminated about the correct things to say in social circumstances and devoted 4 hours per day to cleaning and checking compulsions. She felt she had to wash her car every time before going out; if she could not, she would cancel her social plans.

Table 2

Diagnosis of anorexia nervosa: Questions to ask*

Weight history-What was her highest weight and lowest weight (after weight loss)
-At what ages did these weights occur?
-Ask about her present weight before you weigh her
Eating behavior-What does she eat and when from morning awakening to bedtime?
-Does she eat with the family less often than in the past?
-Is she binging?
Purging behavior-Is she inducing vomiting?
-Is she using laxatives, diuretics, ipecac, or enemas?
Preoccupations and rituals concerning food and weight-Does she constantly count calories and express concern about fat content in foods?
-Does she often gaze in the mirror and comment about being fat?
-How often does she weigh herself?
-Does she express fear of being unable to stop eating?
Activity-Is she jogging, bike riding, or doing aerobics?
-How often, and for how long?
-Is she overactive at home, such as pacing?
Menstrual history-At what age did menses begin?
-What was the date of her last period?
-How regular is her cycle?
Psychiatric comorbidity-Does she have symptoms of depression?
-Impulsive behavior (suicide attempts or self-mutilation)?
-Drug or alcohol abuse?
-Anxiety (obsessive-compulsive behaviors, social phobia, generalized anxiety, fearfulness)?
-Personality disorders?
* Because patients with anorexia nervosa often deny their symptoms and conceal their food intake, it is usually necessary to interview family members or close friends as well as the patient.
 

 

In college, she began abusing alcohol and was arrested once for driving while intoxicated.

Depression is the most common comorbidity in anorexia nervosa. Two-thirds of anorectic patients in a 10-year follow-up study reported a history of major depressive disorder.5 Suicide, starvation, and electrolyte imbalance are the three major causes of death. Among severely ill patients who require hospitalization, 10% to 20% die, though the suicide rate is undocumented.

Compulsions. Anorectics’ preoccupations about food and eating rituals have been compared with compulsions, though less than 20% of patients meet diagnostic criteria for obsessive-compulsive disorder.6

Substance abuse. Bulimic anorectics report more alcohol and substance use and abuse than restricting anorectics.7 The most common substances of abuse are cannabis, cocaine, stimulants, and over-the-counter pills such as diet aids.

Personality disorders. Up to 50% of patients with anorexia nervosa—particularly the binge/purge subtype—have personality disorders. Borderline personality disorder is especially common among binge/purge types,8 and avoidant personality disorder is more common among restricting types.

Table 3

Diagnostic signs of emaciation and purging in patients with anorexia nervosa

Emaciation
  • Dry, cracking skin
  • Lanugo hair
  • Bradycardia
  • Hypotension
  • Leukopenia with relative lymphocytosis
  • Anemia
  • Hypercholesterolemia
  • Reduced bone density
Purging
  • Calluses on dorsum of hand, produced by hand friction from self-induced vomiting
  • Perioral dermatitis
  • Enlarged parotid glands (“chipmunk” face)
  • Tooth enamel erosion, caries, periodontitis
  • Cardiac arrhythmias (hypokalemia from purging)
  • Hypochloremic metabolic alkalosis
  • Hyperamylasemia
  • QT interval and T-wave changes on ECG

Personality disorders usually reflect instability in interpersonal relationships, poor self-image, or fluctuating affect. Patients may show a pattern of social inhibition, feelings of inadequacy, and hypersensitivity to negative evaluation.

Sexuality. Psychosocial and sexual development is often delayed in adolescent anorectics. In adults, interest in sex often plummets with anorexia onset, although binge/purge-type patients occasionally become promiscuous.

Medical signs

Case report continued: Abnormal ECG.

Ms. J was hospitalized after her weight dropped below 75% of normal for her age, height, and body build. She showed signs of electrolyte disturbance, including severe bradycardia (pulse rate 40) and ST-segment abnormalities on ECG.

Clinical signs of emaciation and purging can assist with diagnosis and in making decisions about medical treatment, including hospitalization (Table 3). Patients who purge are often weak and have puffy cheeks or parotid gland enlargement. They may have fainting spells and scars on their hands from stimulating vomiting. Laxative abuse may decrease colon motility and worsen constipation.

Neuroendocrine changes secondary to dieting and weight loss include:

  • increased corticotropin-releasing hormone secretion
  • blunted diurnal cortisol fluctuation
  • decreased follicle-stimulating hormone (FSH) secretion
  • impaired growth hormone regulation
  • decreased luteinizing hormone (LH) secretion
  • mildly decreased triiodothyronine
  • erratic vasopressin secretion.

Measuring these changes is unnecessary, as general nutritional rehabilitation with weight gain will correct them.

Neurotransmitter function. Emaciated anorectics have a blunted response to pharmacologic probes for dopamine, reduced CSF norepinephrine turnover, and decreased CSF serotonin. Neuroimaging studies suggest that serotonin dysfunction may persist after weight is restored, although these findings require replication.

Treatment priorities

Effective therapies. Open studies indicate that multidimensional treatment—medical management, psychoeducation, and individual cognitive-behavioral therapy (CBT)—is most effective for anorexia nervosa. The fewer than 10 controlled trials that address anorexia nervosa treatment show:

  • the more severe the illness, the more intense the treatment required
  • outpatient therapy is most successful in patients who have had the illness <6 months, are not binging and vomiting, and have parents who participate in family therapy.

Hospitalization. An emaciated patient who is irritable, depressed, preoccupied with food, and sleep-deprived is unlikely to make progress toward behavioral change. The first goal, therefore, is to restore her nutritional state to normal.

Severely ill anorectic patients require hospitalization for daily monitoring of weight, calorie intake, urine output, and serum electrolytes and amylase (to assess purging behavior). Hospitalization is indicated for:

  • loss of >20% of normal weight for age, height, and bone structure
  • >6 months of repeated hospitalizations and underweight
  • psychotic depression or serious suicide attempt
  • incapacitating obsessions and compulsions, related or not to the eating disorder
  • serious comorbid medical conditions, such as edema, hypoproteinemia, severe anemia, cardiac arrhythmia, or hypokalemic alkalosis (serum K+ < 2.5 mEq/L).

Keeping a patient in the hospital long enough to provide effective medical and psychological therapy has become difficult, however, because of medical insurance restrictions (Box). The result: poorer outcomes and increased relapse rates compared with 10 years ago.9-12

Box

Shorter hospitalizations, worse outcomes for patients with eating disorders

Hospital treatment of eating disorders has shifted from long-term care of a chronic disorder to stabilization of acute episodes. For some patients, this change has been deleterious and not cost-effective.

A decade ago, eating disorder hospitalizations were covered primarily by private insurance. Today, health maintenance organizations, managed care oversight of private insurance, and public funding are the primary sources of payment. These insurers often limit payment for eating disorder hospitalization, the most costly aspect of psychiatric care.

Poor outcomes and a high relapse rate have been documented in anorexia nervosa patients who left the hospital while underweight.9-11 From 1984 to 1998:

  • average hospital stays for anorexia nervosa decreased from 150 days to 23.7 days
  • readmissions increased from 0% to 27% of total admissions
  • anorectic patients’ average body mass index at discharge dropped from 19.3 to 17.7, a statistically significant difference.12

For psychiatrists, this trend means many outpatients with anorexia nervosa will require repeated hospitalizations that will not substantially improve their anorectic behaviors.

 

 

Nutritional rehabilitation and behavior changes can often correct the medical complications of emaciation and purging. Lost bone density is seldom restored, but nutritional rehabilitation can prevent further bone loss.13 Women who remain amenorrheic for several years after weight restoration tend to be more psychologically disturbed than those who resume menses rapidly.14

Cognitive-behavioral therapy

Other authors have discussed CBT for anorexia nervosa.3,15 In general, the key tasks—operationalizing beliefs, evaluating autonomic thoughts, testing prospective hypotheses, and examining underlying assumptions—are accomplished by assessing anorexia’s distorted cognitions. No satisfactory controlled studies have examined any other type of individual psychotherapy for treating anorexia nervosa.

Alliance building. Patients with anorexia find it difficult to participate in therapeutic relationships. They are terrified of gaining weight and readily drop out of treatment. To build a therapeutic alliance:

  • begin by helping the patient develop a history of her significant life events
  • proceed slowly, praising her for every small attempt at changing her behavior
  • set realistic therapy goals, considering her degree of resistance.

Monitoring. Behavior therapy consists primarily of positive reinforcements for weight gain. For this, we weigh outpatients weekly and inpatients daily. Outpatients are taught to keep diaries of daily food intake, stressful events, and emotional responses to them. The therapist begins each session by examining the patient’s diary with her and discussing how life events affect her eating behavior.

Cognitive restructuring helps patients identify their disturbed cognitions and challenge core beliefs about self-image. In this process, they become aware of their negative thoughts and develop arguments and evidence to support and refute the thoughts’ validity. They then form a reasoned conclusion based on the evidence.

Even if patients do not accept this logical conclusion, we encourage them to behave as if they believe it to be true. By doing this repeatedly, they eventually obtain some symptom relief.

Response-prevention techniques can help stop binging and purging. For example, we may require inpatients to sit together for 1 hour after eating. Because most patients will not vomit in front of each other, they learn how to resist vomiting and eventually experience reduced anxiety without vomiting after a meal.

Problem solving helps patients to reason through difficult food-related or interpersonal situations. The patient states the problem, then generates as many solutions as possible with the therapist’s assistance. She chooses one solution and puts it into effect, usually for 1 week. She then discusses the results with her therapist and decides whether to try another solution.

Family therapy. A family analysis—including a brief psychiatric history and evaluation of interactions—is recommended for all patients who live at home. This analysis can help you decide what type of family therapy or counseling to recommend.

Some families respond well with the parents and patient together in therapy sessions, whereas others are more comfortable with separate counseling. In a recent controlled study, anorectic patients younger than age 18 did equally well whether they were counseled with the family or separately.16

Brief therapy sessions are sometimes the most effective method to address family issues. When this is not possible, you and the patient can discuss family relationships in individual therapy.

Medications

Many medications have been used to treat anorexia nervosa, though few randomized, placebo-controlled studies exist. Because evidence does not support using psychotropics as monotherapy for anorexia nervosa, medication is considered adjunctive to CBT.

Chlorpromazine can help the hospitalized, severely ill patient who is overwhelmed with uncontrollable behavioral rituals and thoughts of losing weight. This antipsychotic helps reduce anorectic preoccupations and anxiety and helps make patients more amenable to therapy.

Start chlorpromazine at 10 mg tid and increase gradually until the patient can eat without extreme anxiety. Usual maximum dosage is 50 mg tid. Monitor blood pressure, tardive dyskinesia, and decreased white blood cell count.

Olanzapine may help induce weight gain and reduce anxiety in anorectic patients.17 Controlled and open-label studies are under way.

We start olanzapine at 2.5 mg/d and increase gradually to 10 or 15 mg/d. At this dosage, patients’ anxiety about eating is usually substantially reduced. Sedation is the most common side effect.

Anorexia patients often refuse to take olanzapine for fear of weight gain. If a patient’s emaciation is life-threatening, we may seek court permission to medicate her involuntarily. We reassure her that we will discontinue olanzapine when she reaches her target weight.

Serotonin in anorexia. Central serotonin pathways modulate feeding behavior. Serotonin antagonists—such as cyproheptadine—increase food intake and weight gain, whereas serotonin agonists—such as selective serotonin reuptake inhibitors (SSRIs)—decrease food intake.

Serotonin pathways also may modulate obsessive-compulsive and impulsive behaviors. Both serotonin agonists and antagonists can be useful adjuncts in treating anorexia nervosa.

 

 

In a double-blind, placebo-controlled trial, cyproheptadine, 4 to 8 mg tid, was associated with weight gain and reduced depressive symptoms in anorexia nervosa patients.18 Unlike tricyclic antidepressants, cyproheptadine does not reduce blood pressure or increase heart rate, which makes it attractive for emaciated anorectic patients. Dosages up to 28 mg/d can be used safely.

The SSRI fluoxetine may help prevent weight loss relapse in anorexia nervosa and reduce obsessive-compulsive behaviors.19 In open studies of low-weight anorectics, however, fluoxetine had little impact on weight or other clinically meaningful variables.20 Thus, this agent is recommended for preventing weight-loss relapse only in patients who are within 10% to 15% of ideal body weight.

Outpatient care

Case report continued: Ongoing therapy

During hospitalization, Ms. J participated in all therapeutic modalities but had difficulty eating enough to gain weight. She reached her target weight of 127 lbs in about 7 weeks but gained no sense of purpose in life.

She is starting an intensive outpatient program using CBT to maintain her weight and further address the core psychopathology of her illness. Her maintenance therapy includes attending Alcoholics Anonymous meetings, ongoing fluoxetine (20 mg/d) to prevent weight-loss relapse, and CBT for obsessions and compulsions not related to her eating disorder.

Related resources

  • Halmi KA. Eating disorders: Anorexia, bulimia nervosa and obesity. In: Hales RE, Yudofsky SC (eds). Textbook of clinical psychiatry, 4th ed. Washington, DC: American Psychiatric Publishing, 2003:1001-21.
  • Academy of Eating Disorders. www.aedweb.org
  • Anorexia Nervosa & Associated Disorders (ANAD). www.anad.org

Drug brand names

  • Chlorpromazine • Thorazine
  • Cyproheptadine • Periactin
  • Fluoxetine • Prozac
  • Olanzapine • Zyprexa

Disclosure

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

References

1. Halmi KA. Anorexia nervosa: demographic and clinical features in 94 cases. Psychosom Med 1974;36:18-24.

2. Bruch AH. Eating disorders: obesity, anorexia nervosa, and the person within. New York, Basic books, 1973.

3. Kleifield E, Wagner S, Halmi KA. Cognitive behavioral treatment of anorexia nervosa. Psychiatr Clin North Am 1996;19:715-34.

4. Halmi KA. Eating disorders. In: Martin A, Scahill L, Charney DS Leckman JF (eds). Pediatric pharmacology. New York: Oxford University Press, 2002;592-602.

5. Halmi KA, Eckert E, Marci P, Cohen J. Comorbidity of psychiatric diagnoses in anorexia nervosa. Arch Gen Psychiatry 1991;48:712-18.

6. Braun DL, Sunday SR, Halmi KA. Psychiatric comorbidity in patients with eating disorders. Psychol Med 1994;24:859-67.

7. Holderness CC, Brooks-Gunn J, Warren MP. Comorbidity of eating disorders and substance abuse review of the literature. Int J Eat Disord 1994;16:1-34.

8. Herzog DB, Keller M, Lavori P. The prevalence of personality disorders in 210 women with eating disorders. J Clin Psychiatry 1992;53:147-52.

9. Baran S, Weltzin T, Kaye W. Low discharge weight and outcome in anorexia nervosa. Am J Psychiatry 1995;150:1070-2.

10. Commerford MC, Licinio J, Halmi KA. Guidelines for discharging eating disorder patients. Eat Disord 1997;5:69-74.

11. Howard W, Evans K, Quinter-Howard C, et al. Predictors of success or failure of transition to day hospital treatment for inpatients with anorexia nervosa. Am J Psychiatry 1999;156:1697-1702.

12. Wiseman C, Sunday SR, Klapper F, et al. Changing patterns of hospitalization in eating disorder patients. Int J Eat Disord 2001;30:69-74.

13. Newman M, Halmi KA. The relationship of bone density to estradiol and cortisol in anorexia nervosa and bulimia nervosa. Psychiatr Res 1989;29:105-12.

14. Falk JR, Halmi KA. Amenorrhea in anorexia nervosa: examination of the critical body hypothesis. Biol Psychiatr 1982;17:799-806.

15. Garner DM, Bemis KM. A cognitive-behavioral approach to anorexia nervosa. Cognit Ther Res 1982;6:1223-50.

16. Dare C. Eisler. Family therapy and eating disorders. In: Fairburn CR, Brownell KD (eds). Eating disorders and obesity. New York: Guilford Press, 2002;314-19.

17. Powers P, Santana CA, Bannon YS. Olanzapine in the treatment of anorexia nervosa: an open label trial. Int J Eat Disord 2002;32:146-54.

18. Halmi KA, Eckert ED, Ladu T, Cohen J. Anorexia nervosa: treatment efficacy of cyproheptadine and amitriptyline. Arch Gen Psychiatry 1986;43:177-81.

19. Kaye W. The use of fluoxetine to prevent relapse in anorexia nervosa (presentation). Pittsburgh, PA: Eating Disorder Research Society annual meeting, 1996.

20. Ferguson C, Lavia M, Crossan P. Are serotonin selective reuptake inhibitors effective in underweight anorexia nervosa? Int J Eat Disord 1999;25:11-17.

References

1. Halmi KA. Anorexia nervosa: demographic and clinical features in 94 cases. Psychosom Med 1974;36:18-24.

2. Bruch AH. Eating disorders: obesity, anorexia nervosa, and the person within. New York, Basic books, 1973.

3. Kleifield E, Wagner S, Halmi KA. Cognitive behavioral treatment of anorexia nervosa. Psychiatr Clin North Am 1996;19:715-34.

4. Halmi KA. Eating disorders. In: Martin A, Scahill L, Charney DS Leckman JF (eds). Pediatric pharmacology. New York: Oxford University Press, 2002;592-602.

5. Halmi KA, Eckert E, Marci P, Cohen J. Comorbidity of psychiatric diagnoses in anorexia nervosa. Arch Gen Psychiatry 1991;48:712-18.

6. Braun DL, Sunday SR, Halmi KA. Psychiatric comorbidity in patients with eating disorders. Psychol Med 1994;24:859-67.

7. Holderness CC, Brooks-Gunn J, Warren MP. Comorbidity of eating disorders and substance abuse review of the literature. Int J Eat Disord 1994;16:1-34.

8. Herzog DB, Keller M, Lavori P. The prevalence of personality disorders in 210 women with eating disorders. J Clin Psychiatry 1992;53:147-52.

9. Baran S, Weltzin T, Kaye W. Low discharge weight and outcome in anorexia nervosa. Am J Psychiatry 1995;150:1070-2.

10. Commerford MC, Licinio J, Halmi KA. Guidelines for discharging eating disorder patients. Eat Disord 1997;5:69-74.

11. Howard W, Evans K, Quinter-Howard C, et al. Predictors of success or failure of transition to day hospital treatment for inpatients with anorexia nervosa. Am J Psychiatry 1999;156:1697-1702.

12. Wiseman C, Sunday SR, Klapper F, et al. Changing patterns of hospitalization in eating disorder patients. Int J Eat Disord 2001;30:69-74.

13. Newman M, Halmi KA. The relationship of bone density to estradiol and cortisol in anorexia nervosa and bulimia nervosa. Psychiatr Res 1989;29:105-12.

14. Falk JR, Halmi KA. Amenorrhea in anorexia nervosa: examination of the critical body hypothesis. Biol Psychiatr 1982;17:799-806.

15. Garner DM, Bemis KM. A cognitive-behavioral approach to anorexia nervosa. Cognit Ther Res 1982;6:1223-50.

16. Dare C. Eisler. Family therapy and eating disorders. In: Fairburn CR, Brownell KD (eds). Eating disorders and obesity. New York: Guilford Press, 2002;314-19.

17. Powers P, Santana CA, Bannon YS. Olanzapine in the treatment of anorexia nervosa: an open label trial. Int J Eat Disord 2002;32:146-54.

18. Halmi KA, Eckert ED, Ladu T, Cohen J. Anorexia nervosa: treatment efficacy of cyproheptadine and amitriptyline. Arch Gen Psychiatry 1986;43:177-81.

19. Kaye W. The use of fluoxetine to prevent relapse in anorexia nervosa (presentation). Pittsburgh, PA: Eating Disorder Research Society annual meeting, 1996.

20. Ferguson C, Lavia M, Crossan P. Are serotonin selective reuptake inhibitors effective in underweight anorexia nervosa? Int J Eat Disord 1999;25:11-17.

Issue
Current Psychiatry - 03(03)
Issue
Current Psychiatry - 03(03)
Page Number
39-59
Page Number
39-59
Publications
Publications
Topics
Article Type
Display Headline
Update on eating disorders Anorexia nervosa: Dual therapy can bring patients back from the brink
Display Headline
Update on eating disorders Anorexia nervosa: Dual therapy can bring patients back from the brink
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Update on eating disorders Eating disorders: Which treatments are most effective

Article Type
Changed
Mon, 04/16/2018 - 14:15
Display Headline
Update on eating disorders Eating disorders: Which treatments are most effective

Compared with other psychiatric diagnoses, eating disorders are relatively new. Bulimia nervosa was first described as a distinct syndrome in 1979, Update on Eating Disorders.” In part 1 Harrison G. Pope, Jr., MD, and James I. Hudson, MD, ScD, of Harvard Medical School describe how to avoid undertreating bulimia nervosa. Future issues will feature insights on:

 

  • anorexia nervosa by Katherine A. Halmi, MD, Weill Medical College of Cornell University
  • binge eating by Susan L. McElroy, MD, Renu Kotwal, MD, and Rakesh M. Kaneria, MD, University of Cincinnati College of Medicine.

Rigorous clinical research by these experts and others has helped weed out unsubstantiated theories while providing empiric evidence of eating disorders’ biological and psychological roots. This series updates the evidence for choosing medications and psychotherapies that have shown the greatest therapeutic promise.

References

 

1. Russell GFM. Bulimia nervosa: an ominous variant of anorexia nervosa. Psychol Med 1979;;9::492.-

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

Article PDF
Author and Disclosure Information

 


Paul E. Keck, Jr, MD
Deputy Editor, Current Psychiatry

Issue
Current Psychiatry - 03(01)
Publications
Topics
Page Number
11-11
Sections
Author and Disclosure Information

 


Paul E. Keck, Jr, MD
Deputy Editor, Current Psychiatry

Author and Disclosure Information

 


Paul E. Keck, Jr, MD
Deputy Editor, Current Psychiatry

Article PDF
Article PDF

Compared with other psychiatric diagnoses, eating disorders are relatively new. Bulimia nervosa was first described as a distinct syndrome in 1979, Update on Eating Disorders.” In part 1 Harrison G. Pope, Jr., MD, and James I. Hudson, MD, ScD, of Harvard Medical School describe how to avoid undertreating bulimia nervosa. Future issues will feature insights on:

 

  • anorexia nervosa by Katherine A. Halmi, MD, Weill Medical College of Cornell University
  • binge eating by Susan L. McElroy, MD, Renu Kotwal, MD, and Rakesh M. Kaneria, MD, University of Cincinnati College of Medicine.

Rigorous clinical research by these experts and others has helped weed out unsubstantiated theories while providing empiric evidence of eating disorders’ biological and psychological roots. This series updates the evidence for choosing medications and psychotherapies that have shown the greatest therapeutic promise.

Compared with other psychiatric diagnoses, eating disorders are relatively new. Bulimia nervosa was first described as a distinct syndrome in 1979, Update on Eating Disorders.” In part 1 Harrison G. Pope, Jr., MD, and James I. Hudson, MD, ScD, of Harvard Medical School describe how to avoid undertreating bulimia nervosa. Future issues will feature insights on:

 

  • anorexia nervosa by Katherine A. Halmi, MD, Weill Medical College of Cornell University
  • binge eating by Susan L. McElroy, MD, Renu Kotwal, MD, and Rakesh M. Kaneria, MD, University of Cincinnati College of Medicine.

Rigorous clinical research by these experts and others has helped weed out unsubstantiated theories while providing empiric evidence of eating disorders’ biological and psychological roots. This series updates the evidence for choosing medications and psychotherapies that have shown the greatest therapeutic promise.

References

 

1. Russell GFM. Bulimia nervosa: an ominous variant of anorexia nervosa. Psychol Med 1979;;9::492.-

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

References

 

1. Russell GFM. Bulimia nervosa: an ominous variant of anorexia nervosa. Psychol Med 1979;;9::492.-

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

Issue
Current Psychiatry - 03(01)
Issue
Current Psychiatry - 03(01)
Page Number
11-11
Page Number
11-11
Publications
Publications
Topics
Article Type
Display Headline
Update on eating disorders Eating disorders: Which treatments are most effective
Display Headline
Update on eating disorders Eating disorders: Which treatments are most effective
Sections
Article PDF Media

Update on eating disorders Bulimia nervosa: Persistent disorder requires equally persistent treatment

Article Type
Changed
Tue, 12/11/2018 - 15:11
Display Headline
Update on eating disorders Bulimia nervosa: Persistent disorder requires equally persistent treatment

Thousands of scientific papers have been written about bulimia, but not all patients receive effective treatments that produce remission.

To set the record straight and help psychiatrists avoid undertreating bulimia, this article discusses:

  • evidence for using antidepressants, even when patients are not “depressed”
  • merits of psychotherapies, including those shown to work and those that can harm
  • augmentation therapies that can help increase response from partial to full remission.

Initial evaluation

Diagnosis. Bulimia nervosa is characterized by eating binges, followed by purging behaviors such as self-induced vomiting or laxative abuse1,2 (Table 1). It affects 1% to 3% of adolescent girls and young women and occurs in women 5 to 10 times more often than in men.

Bulimia is often persistent. About one-half of bulimic patients—including those who have been treated—continue to show eating disorder features on long-term follow-up.3,4

Psychiatric comorbidity. Most bulimic patients report a history of other psychiatric disorders, especially major depressive and bipolar disorders and anxiety disorders such as panic disorder, social phobia, and obsessive-compulsive disorder (OCD).5 Because these psychiatric comorbidities may occur before, during, or after bulimia nervosa, one cannot assume that mood or anxiety disorders are a cause or consequence of bulimia. Instead, bulimia nervosa, mood disorders, and anxiety disorders may be different expressions of a shared etiologic abnormality.

Table 1

DSM-IV-TR diagnostic criteria for bulimia nervosa

  1. Recurrent episodes of binge eating, characterized by both of the following:
  2. Recurrent inappropriate compensatory behavior in order to prevent weight gain, such as self-induced vomiting; misuse of laxatives, diuretics, enemas, or other medications; fasting; or excessive exercise
  3. The binge eating and inappropriate compensatory behaviors both occur, on average, at least twice a week for 3 months
  4. Self-evaluation is unduly influenced by body shape and weight
  5. The disturbance does not occur exclusively during episodes of anorexia nervosa
Specify type:
Purging type: during the current episode of bulimia nervosa, the person has regularly engaged in self-induced vomiting or the misuse of laxatives, diuretics, or enemas
Non-purging type: during the current episode of bulimia nervosa, the person has used other inappropriate compensatory behaviors, such as fasting or excessive exercise but has not regularly engaged in self-induced vomiting or the misuse of laxatives, diuretics, or enemas
Source: Reprinted with permission from the Diagnostic and Statistical Manual of Mental Disorders, 4th ed, text rev. Copyright 2000 American Psychiatric Association.

Evidence supporting this hypothesis comes from studies showing that these disorders:

  • respond to several chemically unrelated families of antidepressants6,7
  • frequently co-occur in individual patients5,7
  • frequently co-aggregate in families.7-9

We have published this evidence6,7 and proposed that bulimia nervosa may be one form of a larger underlying disorder, which we termed “affective spectrum disorder.”

Antidepressants are often rapidly effective in treating bulimic symptoms,10 regardless of whether patients exhibit depressive symptoms. Thus, there is no reason to withhold antidepressant therapy simply because a bulimic patient is not depressed. The term “antidepressant” may be a misnomer; these drugs are effective for numerous conditions, of which depression is only one.

Anorexic symptoms. Co-occurring depressive or anxiety disorders in a bulimic patient will not greatly alter treatment. The antidepressants and psychotherapies typically used to treat bulimia are often equally effective for affective disorders. Co-occurring anorexia nervosa, however, is a more serious concern.

Bulimic patients often display a history of anorexia nervosa; in many cases, the patient develops anorexia nervosa as a teenager and then progresses to bulimia nervosa across several years. Her prognosis is much better if her weight normalizes with the shift to bulimia nervosa, than if her weight remains well below normal for her height. It is unclear why medications and psychotherapy are much less effective in bulimic patients with anorexic symptoms than in those with bulimia alone. Watch for further details on anorexia nervosa as this series continues in future issues of.

Medical considerations. Potential medical complications—mostly consequences of vomiting or laxative use—are important to consider when you assess a bulimic patient:1

  • The acid in vomitus may gradually erode tooth enamel, requiring dental consultation.
  • Vomitus may inflame salivary gland ducts, though the swelling is usually benign.
  • Frequent vomiting may result in hypokalemia and alkalosis, although aggressive medical treatment usually is not needed.

Ask about ipecac use. To induce vomiting, some patients may abuse ipecac syrup, which can cause cardiomyopathy.11

Inpatient or outpatient? Unless the bulimic patient displays severe and medically dangerous anorexic symptoms, she can usually be treated as an outpatient. However, evaluate her carefully for suicidal ideation—which is not uncommon in bulimia nervosa—and consider inpatient treatment if necessary.

Medication vs. psychotherapy

The relative merits of medication versus psychotherapy in treating bulimia nervosa continue to be debated. The Cochrane Database of Systematic Reviews includes meta-analyses of both drug therapy12 and psychotherapy13 for bulimia nervosa. The 2001 drug therapy review found that “the use of a single antidepressant agent was clinically effective,” with no one drug clearly superior to another. Notably, this review was published before recent findings on topiramate.

 

 

The corresponding 2002 review of psychotherapy concludes—somewhat more cautiously—that “there is a small body of evidence for the efficacy of cognitive-behavior therapy in bulimia nervosa and similar syndromes, but the quality of trials is very variable and sample sizes are often small.”

In bulimia nervosa and other psychiatric disorders, comparing psychotherapy with drug therapy is hazardous because several factors bias the comparison in favor of psychotherapy. These factors include an expectational effect, a responsibility effect, and differential generalizability of study results.

Expectational effect. Patients in clinical trials are aware that they are receiving psychotherapy and, presumably, that study investigators hope to demonstrate its efficacy. This might account for much of psychotherapy’s apparent effect, as even placebos can produce 30% to 50% improvement in bulimia.14

Responsibility effect. If a patient fails to improve in a drug study, she will conclude that the drug has failed. But if she fails to improve in a psychotherapy study, she may conclude that she has failed. Because psychological treatments generally require patients to work in therapy, the patient may feel partially responsible for the outcome. Thus, to avoid cognitive dissonance, she may consciously or unconsciously exaggerate her improvement, both in her own mind and when reporting to treaters.

Differential generalizability. Psychological study protocols, such as administering several months of a behavioral treatment, usually mimic clinical practice fairly well. This is not the case with drug study protocols.

No responsible clinician would inflexibly administer a single dosage of a single drug for a fixed period to every bulimic patient and then declare failure for all nonresponders, as is done in study protocols. In practice, the clinician can offer nonresponders augmentation strategies and additional drug trials. Thus, calculations of bulimia response rates in drug studies substantially understate response to drug therapy in clinical practice.

Table 2

How effective are medications in treating bulimia nervosa?

MedicationEvidence for efficacyRemarks
Antidepressants
Selective serotonin reuptake inhibitors+++Fluoxetine is only SSRI studied in controlled trials
Tricyclics+++Generally more side effects than SSRIs
Monoamine oxidase inhibitors++High rates of remission, but dietary restrictions
Trazodone++Only one controlled trial
Venlafaxine, mirtazapine, nefazodone?No controlled trials, but probably effective
Bupropion(++)Not recommended; caused seizures in bulimic patients
Anticonvulsants
Topiramate++Only one controlled trial, but substantial effect size
Phenytoin+Little efficacy in only controlled study
Carbamazepine+May be useful in bulimia with comorbid bipolar disorder
Valproate+May be useful in bulimia with comorbid bipolar disorder
Other agents
Liothyronine+Augmentation agent in patients with incomplete antidepressant response
Lithium+Ineffective in only controlled trial; possible augmentation strategy
Naltrexone0Ineffective in two controlled trials
Ondansetron+One controlled trial
0 No apparent efficacy
+ Occasional effect; limited evidence
++ Clear effect; good evidence from controlled trial(s)
+++ Strongly documented effect; evidence from multiple controlled trials.
( ) Negative effect

One also might note that psychological study findings have not “sold” well in the clinical practice marketplace. For example, in a recent survey of more than 220 bulimic women treated with psychotherapy, only 6.9% said they received a full course of cognitive behavioral therapy (CBT)14 —despite two decades of evidence of its efficacy. By contrast, untested, inefficacious, and possibly harmful psychotherapies for bulimia—including recovered-memory therapy—appear to be thriving.

Recommendation. Interpret with caution any head-to-head comparisons of psychological versus drug therapies—especially when clinical practice recommendations are made. Certain psychological therapies provided by specifically-trained individuals likely do help patients with bulimia nervosa. However, biases inherent to the studies may inflate psychological therapies’ efficacy when compared with that of drug therapy.

Therefore, for a psychiatrist who does not specialize in eating disorders to offer exclusively psychological therapy to a bulimic patient—while withholding or postponing drug therapy—may now be a questionable practice.

Choosing drug therapies

Although consensus is lacking on an optimal treatment trial sequence for bulimia nervosa, we suggest a rational approach based on the evidence and our experience (Algorithm).

First-line antidepressants. A selective serotonin reuptake inhibitor (SSRI) trial is usually the first choice (Table 2), and some data suggest that higher-than-usual dosages may be required. For example, in a large multicenter trial of fluoxetine in bulimia nervosa, 60 mg/d was considerably more effective than 20 mg/d for reducing binge eating behavior and vomiting frequency.15

Based on our observations, however, we believe that noncompliance or irregular compliance may account for this difference in response. Bulimic patients’ impulsive and obsessional behavior may keep them from taking their medications as prescribed. The higher fluoxetine dosage may therefore have been more effective simply because it ensured adequate plasma levels, even when patients missed or forgot multiple doses.

Augmenting agents. A first antidepressant trial rarely leads to complete remission of bulimic symptoms. This is not a serious concern, however, because many other options are available.

Liothyronine. Partial responders to SSRIs often become complete responders when we add a 10-day trial of liothyronine (T3), 25 μg/d. If this fails, we may try augmenting with lithium carbonate, although bulimic patients are often afraid of weight gain or lithium’s other side effects.

 

 

Topiramate. A newer augmentation strategy is to add the anticonvulsant topiramate. Used alone, topiramate demonstrated effectiveness for bulimia nervosa in one placebo-controlled, double-blind trial.16

Adding topiramate to an antidepressant regimen will likely reduce any remaining bulimic symptoms. In addition, topiramate often produces weight loss—a side effect that bulimic patients usually welcome. It remains unclear whether topiramate’s weight-loss effects might pose a hazard in patients with simultaneous bulimic and anorexic symptoms.

Other antidepressants. If the above strategies fail, other antidepressant options include venlafaxine, tricyclics, and monoamine oxidase inhibitors. Bupropion is not recommended in bulimia nervosa; one trial17 of this agent resulted in a much higher rate of grand mal seizures in bulimic patients than in patients taking bupropion for depression.

In bulimic patients with concomitant bipolar disorder, the anticonvulsants carbamazepine and valproate often reduce affective and bulimic symptoms. By contrast, the anticonvulsant phenytoin—once thought to be useful in bulimia nervosa10 —offers little benefit for either bulimic or affective symptoms.

Persistence is important when initial medication trials fail. One unblinded study followed 36 bulimic patients 9 to 19 months after they completed a controlled study with trazodone.18 Of the 26 patients who tried a second or third antidepressant, 17 (65%) achieved remission of bulimia on follow-up. Of the 10 patients who declined a second or third trial, only 1 (10%) attained remission.

Notably, these study results were obtained before the SSRIs and other newer antidepressants or topiramate became available. Cooperative patients using present-day medications might be able to achieve remission rates that exceed 65%.

Algorithm Proposed treatment approach to bulimia nervosa



Table 3

How effective are psychotherapies in treating bulimia nervosa?

PsychotherapyEvidence for efficacyRemarks
Cognitive behavioral therapy (CBT)+++Controlled evidence for efficacy in individual and group treatment
Interpersonal psychotherapy (IPT)++Effective, but slower than CBT
Exposure with response prevention+May be added to other behavioral techniques, though additive benefit questionable
Dialectical behavior therapy+Highly structured behavioral technique originally developed for borderline personality disorder
Self-help groups+Frequently considered very helpful by patients
Psychodynamic psychotherapy0“Recovered memory” approaches are frankly harmful
Eye movement desensitization and reprocessing (EMDR)0Dubious theoretical basis; no methodologically acceptable evidence for efficacy
0 No apparent efficacy
+ Occasional effect; limited evidence
++ Clear effect; good evidence from controlled trial(s)
+++ Strongly documented effect; evidence from multiple controlled trials.

Psychotherapy

Cognitive-behavioral therapy. CBT—given either individually or in groups—is the most effective psychotherapy for bulimia (Table 3).19 CBT typically involves 3 to 6 months of helping the patient focus on her bulimic behaviors and on specific attitudes—such as unrealistic preoccupations with being “too fat”—that perpetuate the behaviors.

In practice, unfortunately, few bulimic patients are offered CBT, perhaps because few clinicians are trained in the specific approach used for bulimia nervosa.19 If you are not trained in using CBT for bulimia and do not have access to colleagues who offer this treatment, you may begin with medication plus simple behavioral treatments, such as:

  • offering supportive therapy in the office
  • referring patients to self-help groups for persons with eating disorders.

If this strategy fails, encourage patients to consider CBT—even if they must travel some distance to obtain it.

Other specialized psychotherapies. Dialectical behavior therapy and interpersonal psychotherapy appear to be effective in bulimia. Again, however, clinicians who lack training in these techniques or access to local experts may be unable to offer them. Psychodynamic therapy does not appear to offer greater benefit in bulimia nervosa than ordinary supportive counseling.

Dubious therapies. One psychodynamic approach—regrettably still practiced—is “recovered memory therapy.” Therapists who use it claim that childhood sexual abuse or other trauma can cause bulimic symptoms but patients have repressed the memory of these events.20

No methodologically sound evidence has shown that childhood sexual abuse can cause bulimia nervosa years or decades later.21 Nor is there acceptable evidence that people can repress the memory of a traumatic experience.22 Therapists administering recovered memory therapy have been subjected to malpractice judgments totaling tens of millions of dollars from suits filed by patients who eventually realized that so-called “recovered” memories were false.23

Another dubious therapy—eye movement desensitization and reprocessing (EMDR)—also may involve attempts to “recover” memories of putative traumatic events.24 No methodologically sound evidence has shown that EMDR is effective in bulimic patients, and the technique’s theoretical basis is questionable.24,25

Related resources

Drug brand names

  • Bupropion • Wellbutrin
  • Citalopram • Celexa
  • Desipramine • Norpramin
  • Fluoxetine • Prozac
  • Lithium • Lithobid, Eskalith
  • Nortriptyline • Pamelor, Aventyl
  • Sertraline • Zoloft
  • Topiramate • Topamax
  • Trazodone • Desyrel
  • Liothyronine • Cytomel
  • Venlafaxine • Effexor

Disclosure

Dr. Pope receives research support from Ortho-McNeil Pharmaceuticals and is a consultant to Solvay Pharmaceuticals and Auxilium Pharmaceuticals.

 

 

Dr. Hudson receives research support from and is a consultant to Eli Lilly & Co. and Ortho-McNeil Pharmaceuticals.

References

1. Mehler PS. Clinical practice. Bulimia nervosa. N Engl J Med 2003;349(9):875-81.

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

3. Fisher M. The course and outcome of eating disorders in adults and in adolescents: a review. Adolesc Med 2003;14(1):149-58.

4. Fairburn CG, Norman PA, Welch SL, et al. A prospective study of outcome in bulimia nervosa and the long-term effects of three psychological treatments. Arch Gen Psychiatry 1995;52(4):304-12.

5. Hudson JI, Pope HG, Jr, Yurgelun-Todd D, et al. A controlled study of lifetime prevalence of affective and other psychiatric disorders in bulimic outpatients. Am J Psychiatry 1987;144(10):1283-7.

6. Hudson JI, Pope HG, Jr. Affective spectrum disorder: does antidepressant response identify a family of disorders with a common pathophysiology? Am J Psychiatry 1990;147(5):552-64.

7. Hudson JI, Mangweth B, Pope HG, Jr, et al. Family study of affective spectrum disorder. Arch Gen Psychiatry 2003;60:170-7.

8. Hudson JI, Laird NM, Betensky RA, et al. Multivariate logistic regression for familial aggregation of two disorders: II. Analysis of studies of eating and mood disorders. Am J Epidemiology 2001;153(5):506-14.

9. Mangweth B, Hudson JI, Pope HG, Jr, et al. Family study of the aggregation of eating disorders and mood disorders. Psychol Med (in press).

10. Hudson JI, Pope HG, Jr, Carter WP. Pharmacologic therapy of bulimia nervosa. In: Goldstein D (ed). The management of eating disorders and obesity (2nd ed) Totowa, NJ: Humana Press, Inc.(in press).

11. Pope HG, Jr, Hudson JI, Nixon RA, Herridge PL. The epidemiology of ipecac abuse. N Engl J Med 1986;14(4):245-6.

12. Hay PJ, Bacaltchuk J. Psychotherapy for bulimia nervosa and binging. Cochrane Database Syst Rev 2003;(1):CD000562.-

13. Bacaltchuk J, Hay P. Antidepressants versus placebo for people with bulimia nervosa. Cochrane Database Syst Rev 2001;(4):CD003391.-

14. Crow S, Mussell MP, Peterson C, et al. Prior treatment received by patients with bulimia nervosa. Int J Eating Disord 1999;25(1):39-44.

15. Fluoxetine Bulimia Collaborative Study Group Fluoxetine in the treatment of bulimia nervosa: a multicenter placebo-controlled, double-blind trial. Arch Gen Psychiatry 1992;49:139-47.

16. Hoopes S, Reimherr F, Hedges D, et al. Part 1:Topiramate in the treatment of bulimia nervosa: a randomized, double-blind, placebocontrolled trial. J Clin Psychiatry (in press).

17. Horne RL, Ferguson JM, Pope HG, Jr, et al. Treatment of bulimia with bupropion: a controlled multi-center trial. J Clin Psychiatry 1988;49(7):262-6.

18. Pope HG, Jr, McElroy SL, Keck PE, Jr, Hudson JI. Long-term pharmacotherapy of bulimia nervosa. J Clin Psychopharmacol 1989;9(5):385-6.

19. Fairburn CG, Harrison PJ. Eating disorders. Lancet 2003;361(9355):407-16.

20. Pope HG, Jr, Hudson JI. “Recovered memory” therapy for eating disorders: implications of the Ramona verdict. Int J Eat Disord 1996;19(2):139-45.

21. Pope HG, Jr, Hudson JI. Does childhood sexual abuse cause adult psychiatric disorders? Essentials of methodology. J Psychiatry Law 1995;Fall:363-81.

22. Pope HG, Jr, Oliva PS, Hudson JI. Repressed memories. The scientific status of research on repressed memories. In: Faigman DL, Kaye DH, Saks MJ, Sanders J (eds). Science in the law: social and behavioral science issues St. Paul, MN: West Group, 2002;487-526.

23. Cannell J, Hudson JI, Pope HG, Jr. Standards for informed consent in recovered memory therapy. J Am Acad Psychiatry Law 2001;29(2):138-47.

24. Hudson JI, Chase EA, Pope HG, Jr. Eye movement desensitization and reprocessing in eating disorders: caution against premature acceptance. Int J Eat Disord 1998;23:1-5.

25. McNally RJ. EMDR and mesmerism: a comparative historical analysis. J Anxiety Disord 1999;13(1-2):225-36.

Article PDF
Author and Disclosure Information

Paul E. Keck, Jr, MD

Harrison G. Pope, Jr, MD
Professor of psychiatry Harvard Medical School, Boston Director, Biological Psychiatry Laboratory McLean Hospital, Belmont, MA

James I. Hudson, MD, ScD
Associate professor of psychiatry Harvard Medical School, Boston Director, Biological Psychiatry Laboratory McLean Hospital, Belmont, MA

Issue
Current Psychiatry - 03(01)
Publications
Topics
Page Number
12-22
Sections
Author and Disclosure Information

Paul E. Keck, Jr, MD

Harrison G. Pope, Jr, MD
Professor of psychiatry Harvard Medical School, Boston Director, Biological Psychiatry Laboratory McLean Hospital, Belmont, MA

James I. Hudson, MD, ScD
Associate professor of psychiatry Harvard Medical School, Boston Director, Biological Psychiatry Laboratory McLean Hospital, Belmont, MA

Author and Disclosure Information

Paul E. Keck, Jr, MD

Harrison G. Pope, Jr, MD
Professor of psychiatry Harvard Medical School, Boston Director, Biological Psychiatry Laboratory McLean Hospital, Belmont, MA

James I. Hudson, MD, ScD
Associate professor of psychiatry Harvard Medical School, Boston Director, Biological Psychiatry Laboratory McLean Hospital, Belmont, MA

Article PDF
Article PDF

Thousands of scientific papers have been written about bulimia, but not all patients receive effective treatments that produce remission.

To set the record straight and help psychiatrists avoid undertreating bulimia, this article discusses:

  • evidence for using antidepressants, even when patients are not “depressed”
  • merits of psychotherapies, including those shown to work and those that can harm
  • augmentation therapies that can help increase response from partial to full remission.

Initial evaluation

Diagnosis. Bulimia nervosa is characterized by eating binges, followed by purging behaviors such as self-induced vomiting or laxative abuse1,2 (Table 1). It affects 1% to 3% of adolescent girls and young women and occurs in women 5 to 10 times more often than in men.

Bulimia is often persistent. About one-half of bulimic patients—including those who have been treated—continue to show eating disorder features on long-term follow-up.3,4

Psychiatric comorbidity. Most bulimic patients report a history of other psychiatric disorders, especially major depressive and bipolar disorders and anxiety disorders such as panic disorder, social phobia, and obsessive-compulsive disorder (OCD).5 Because these psychiatric comorbidities may occur before, during, or after bulimia nervosa, one cannot assume that mood or anxiety disorders are a cause or consequence of bulimia. Instead, bulimia nervosa, mood disorders, and anxiety disorders may be different expressions of a shared etiologic abnormality.

Table 1

DSM-IV-TR diagnostic criteria for bulimia nervosa

  1. Recurrent episodes of binge eating, characterized by both of the following:
  2. Recurrent inappropriate compensatory behavior in order to prevent weight gain, such as self-induced vomiting; misuse of laxatives, diuretics, enemas, or other medications; fasting; or excessive exercise
  3. The binge eating and inappropriate compensatory behaviors both occur, on average, at least twice a week for 3 months
  4. Self-evaluation is unduly influenced by body shape and weight
  5. The disturbance does not occur exclusively during episodes of anorexia nervosa
Specify type:
Purging type: during the current episode of bulimia nervosa, the person has regularly engaged in self-induced vomiting or the misuse of laxatives, diuretics, or enemas
Non-purging type: during the current episode of bulimia nervosa, the person has used other inappropriate compensatory behaviors, such as fasting or excessive exercise but has not regularly engaged in self-induced vomiting or the misuse of laxatives, diuretics, or enemas
Source: Reprinted with permission from the Diagnostic and Statistical Manual of Mental Disorders, 4th ed, text rev. Copyright 2000 American Psychiatric Association.

Evidence supporting this hypothesis comes from studies showing that these disorders:

  • respond to several chemically unrelated families of antidepressants6,7
  • frequently co-occur in individual patients5,7
  • frequently co-aggregate in families.7-9

We have published this evidence6,7 and proposed that bulimia nervosa may be one form of a larger underlying disorder, which we termed “affective spectrum disorder.”

Antidepressants are often rapidly effective in treating bulimic symptoms,10 regardless of whether patients exhibit depressive symptoms. Thus, there is no reason to withhold antidepressant therapy simply because a bulimic patient is not depressed. The term “antidepressant” may be a misnomer; these drugs are effective for numerous conditions, of which depression is only one.

Anorexic symptoms. Co-occurring depressive or anxiety disorders in a bulimic patient will not greatly alter treatment. The antidepressants and psychotherapies typically used to treat bulimia are often equally effective for affective disorders. Co-occurring anorexia nervosa, however, is a more serious concern.

Bulimic patients often display a history of anorexia nervosa; in many cases, the patient develops anorexia nervosa as a teenager and then progresses to bulimia nervosa across several years. Her prognosis is much better if her weight normalizes with the shift to bulimia nervosa, than if her weight remains well below normal for her height. It is unclear why medications and psychotherapy are much less effective in bulimic patients with anorexic symptoms than in those with bulimia alone. Watch for further details on anorexia nervosa as this series continues in future issues of.

Medical considerations. Potential medical complications—mostly consequences of vomiting or laxative use—are important to consider when you assess a bulimic patient:1

  • The acid in vomitus may gradually erode tooth enamel, requiring dental consultation.
  • Vomitus may inflame salivary gland ducts, though the swelling is usually benign.
  • Frequent vomiting may result in hypokalemia and alkalosis, although aggressive medical treatment usually is not needed.

Ask about ipecac use. To induce vomiting, some patients may abuse ipecac syrup, which can cause cardiomyopathy.11

Inpatient or outpatient? Unless the bulimic patient displays severe and medically dangerous anorexic symptoms, she can usually be treated as an outpatient. However, evaluate her carefully for suicidal ideation—which is not uncommon in bulimia nervosa—and consider inpatient treatment if necessary.

Medication vs. psychotherapy

The relative merits of medication versus psychotherapy in treating bulimia nervosa continue to be debated. The Cochrane Database of Systematic Reviews includes meta-analyses of both drug therapy12 and psychotherapy13 for bulimia nervosa. The 2001 drug therapy review found that “the use of a single antidepressant agent was clinically effective,” with no one drug clearly superior to another. Notably, this review was published before recent findings on topiramate.

 

 

The corresponding 2002 review of psychotherapy concludes—somewhat more cautiously—that “there is a small body of evidence for the efficacy of cognitive-behavior therapy in bulimia nervosa and similar syndromes, but the quality of trials is very variable and sample sizes are often small.”

In bulimia nervosa and other psychiatric disorders, comparing psychotherapy with drug therapy is hazardous because several factors bias the comparison in favor of psychotherapy. These factors include an expectational effect, a responsibility effect, and differential generalizability of study results.

Expectational effect. Patients in clinical trials are aware that they are receiving psychotherapy and, presumably, that study investigators hope to demonstrate its efficacy. This might account for much of psychotherapy’s apparent effect, as even placebos can produce 30% to 50% improvement in bulimia.14

Responsibility effect. If a patient fails to improve in a drug study, she will conclude that the drug has failed. But if she fails to improve in a psychotherapy study, she may conclude that she has failed. Because psychological treatments generally require patients to work in therapy, the patient may feel partially responsible for the outcome. Thus, to avoid cognitive dissonance, she may consciously or unconsciously exaggerate her improvement, both in her own mind and when reporting to treaters.

Differential generalizability. Psychological study protocols, such as administering several months of a behavioral treatment, usually mimic clinical practice fairly well. This is not the case with drug study protocols.

No responsible clinician would inflexibly administer a single dosage of a single drug for a fixed period to every bulimic patient and then declare failure for all nonresponders, as is done in study protocols. In practice, the clinician can offer nonresponders augmentation strategies and additional drug trials. Thus, calculations of bulimia response rates in drug studies substantially understate response to drug therapy in clinical practice.

Table 2

How effective are medications in treating bulimia nervosa?

MedicationEvidence for efficacyRemarks
Antidepressants
Selective serotonin reuptake inhibitors+++Fluoxetine is only SSRI studied in controlled trials
Tricyclics+++Generally more side effects than SSRIs
Monoamine oxidase inhibitors++High rates of remission, but dietary restrictions
Trazodone++Only one controlled trial
Venlafaxine, mirtazapine, nefazodone?No controlled trials, but probably effective
Bupropion(++)Not recommended; caused seizures in bulimic patients
Anticonvulsants
Topiramate++Only one controlled trial, but substantial effect size
Phenytoin+Little efficacy in only controlled study
Carbamazepine+May be useful in bulimia with comorbid bipolar disorder
Valproate+May be useful in bulimia with comorbid bipolar disorder
Other agents
Liothyronine+Augmentation agent in patients with incomplete antidepressant response
Lithium+Ineffective in only controlled trial; possible augmentation strategy
Naltrexone0Ineffective in two controlled trials
Ondansetron+One controlled trial
0 No apparent efficacy
+ Occasional effect; limited evidence
++ Clear effect; good evidence from controlled trial(s)
+++ Strongly documented effect; evidence from multiple controlled trials.
( ) Negative effect

One also might note that psychological study findings have not “sold” well in the clinical practice marketplace. For example, in a recent survey of more than 220 bulimic women treated with psychotherapy, only 6.9% said they received a full course of cognitive behavioral therapy (CBT)14 —despite two decades of evidence of its efficacy. By contrast, untested, inefficacious, and possibly harmful psychotherapies for bulimia—including recovered-memory therapy—appear to be thriving.

Recommendation. Interpret with caution any head-to-head comparisons of psychological versus drug therapies—especially when clinical practice recommendations are made. Certain psychological therapies provided by specifically-trained individuals likely do help patients with bulimia nervosa. However, biases inherent to the studies may inflate psychological therapies’ efficacy when compared with that of drug therapy.

Therefore, for a psychiatrist who does not specialize in eating disorders to offer exclusively psychological therapy to a bulimic patient—while withholding or postponing drug therapy—may now be a questionable practice.

Choosing drug therapies

Although consensus is lacking on an optimal treatment trial sequence for bulimia nervosa, we suggest a rational approach based on the evidence and our experience (Algorithm).

First-line antidepressants. A selective serotonin reuptake inhibitor (SSRI) trial is usually the first choice (Table 2), and some data suggest that higher-than-usual dosages may be required. For example, in a large multicenter trial of fluoxetine in bulimia nervosa, 60 mg/d was considerably more effective than 20 mg/d for reducing binge eating behavior and vomiting frequency.15

Based on our observations, however, we believe that noncompliance or irregular compliance may account for this difference in response. Bulimic patients’ impulsive and obsessional behavior may keep them from taking their medications as prescribed. The higher fluoxetine dosage may therefore have been more effective simply because it ensured adequate plasma levels, even when patients missed or forgot multiple doses.

Augmenting agents. A first antidepressant trial rarely leads to complete remission of bulimic symptoms. This is not a serious concern, however, because many other options are available.

Liothyronine. Partial responders to SSRIs often become complete responders when we add a 10-day trial of liothyronine (T3), 25 μg/d. If this fails, we may try augmenting with lithium carbonate, although bulimic patients are often afraid of weight gain or lithium’s other side effects.

 

 

Topiramate. A newer augmentation strategy is to add the anticonvulsant topiramate. Used alone, topiramate demonstrated effectiveness for bulimia nervosa in one placebo-controlled, double-blind trial.16

Adding topiramate to an antidepressant regimen will likely reduce any remaining bulimic symptoms. In addition, topiramate often produces weight loss—a side effect that bulimic patients usually welcome. It remains unclear whether topiramate’s weight-loss effects might pose a hazard in patients with simultaneous bulimic and anorexic symptoms.

Other antidepressants. If the above strategies fail, other antidepressant options include venlafaxine, tricyclics, and monoamine oxidase inhibitors. Bupropion is not recommended in bulimia nervosa; one trial17 of this agent resulted in a much higher rate of grand mal seizures in bulimic patients than in patients taking bupropion for depression.

In bulimic patients with concomitant bipolar disorder, the anticonvulsants carbamazepine and valproate often reduce affective and bulimic symptoms. By contrast, the anticonvulsant phenytoin—once thought to be useful in bulimia nervosa10 —offers little benefit for either bulimic or affective symptoms.

Persistence is important when initial medication trials fail. One unblinded study followed 36 bulimic patients 9 to 19 months after they completed a controlled study with trazodone.18 Of the 26 patients who tried a second or third antidepressant, 17 (65%) achieved remission of bulimia on follow-up. Of the 10 patients who declined a second or third trial, only 1 (10%) attained remission.

Notably, these study results were obtained before the SSRIs and other newer antidepressants or topiramate became available. Cooperative patients using present-day medications might be able to achieve remission rates that exceed 65%.

Algorithm Proposed treatment approach to bulimia nervosa



Table 3

How effective are psychotherapies in treating bulimia nervosa?

PsychotherapyEvidence for efficacyRemarks
Cognitive behavioral therapy (CBT)+++Controlled evidence for efficacy in individual and group treatment
Interpersonal psychotherapy (IPT)++Effective, but slower than CBT
Exposure with response prevention+May be added to other behavioral techniques, though additive benefit questionable
Dialectical behavior therapy+Highly structured behavioral technique originally developed for borderline personality disorder
Self-help groups+Frequently considered very helpful by patients
Psychodynamic psychotherapy0“Recovered memory” approaches are frankly harmful
Eye movement desensitization and reprocessing (EMDR)0Dubious theoretical basis; no methodologically acceptable evidence for efficacy
0 No apparent efficacy
+ Occasional effect; limited evidence
++ Clear effect; good evidence from controlled trial(s)
+++ Strongly documented effect; evidence from multiple controlled trials.

Psychotherapy

Cognitive-behavioral therapy. CBT—given either individually or in groups—is the most effective psychotherapy for bulimia (Table 3).19 CBT typically involves 3 to 6 months of helping the patient focus on her bulimic behaviors and on specific attitudes—such as unrealistic preoccupations with being “too fat”—that perpetuate the behaviors.

In practice, unfortunately, few bulimic patients are offered CBT, perhaps because few clinicians are trained in the specific approach used for bulimia nervosa.19 If you are not trained in using CBT for bulimia and do not have access to colleagues who offer this treatment, you may begin with medication plus simple behavioral treatments, such as:

  • offering supportive therapy in the office
  • referring patients to self-help groups for persons with eating disorders.

If this strategy fails, encourage patients to consider CBT—even if they must travel some distance to obtain it.

Other specialized psychotherapies. Dialectical behavior therapy and interpersonal psychotherapy appear to be effective in bulimia. Again, however, clinicians who lack training in these techniques or access to local experts may be unable to offer them. Psychodynamic therapy does not appear to offer greater benefit in bulimia nervosa than ordinary supportive counseling.

Dubious therapies. One psychodynamic approach—regrettably still practiced—is “recovered memory therapy.” Therapists who use it claim that childhood sexual abuse or other trauma can cause bulimic symptoms but patients have repressed the memory of these events.20

No methodologically sound evidence has shown that childhood sexual abuse can cause bulimia nervosa years or decades later.21 Nor is there acceptable evidence that people can repress the memory of a traumatic experience.22 Therapists administering recovered memory therapy have been subjected to malpractice judgments totaling tens of millions of dollars from suits filed by patients who eventually realized that so-called “recovered” memories were false.23

Another dubious therapy—eye movement desensitization and reprocessing (EMDR)—also may involve attempts to “recover” memories of putative traumatic events.24 No methodologically sound evidence has shown that EMDR is effective in bulimic patients, and the technique’s theoretical basis is questionable.24,25

Related resources

Drug brand names

  • Bupropion • Wellbutrin
  • Citalopram • Celexa
  • Desipramine • Norpramin
  • Fluoxetine • Prozac
  • Lithium • Lithobid, Eskalith
  • Nortriptyline • Pamelor, Aventyl
  • Sertraline • Zoloft
  • Topiramate • Topamax
  • Trazodone • Desyrel
  • Liothyronine • Cytomel
  • Venlafaxine • Effexor

Disclosure

Dr. Pope receives research support from Ortho-McNeil Pharmaceuticals and is a consultant to Solvay Pharmaceuticals and Auxilium Pharmaceuticals.

 

 

Dr. Hudson receives research support from and is a consultant to Eli Lilly & Co. and Ortho-McNeil Pharmaceuticals.

Thousands of scientific papers have been written about bulimia, but not all patients receive effective treatments that produce remission.

To set the record straight and help psychiatrists avoid undertreating bulimia, this article discusses:

  • evidence for using antidepressants, even when patients are not “depressed”
  • merits of psychotherapies, including those shown to work and those that can harm
  • augmentation therapies that can help increase response from partial to full remission.

Initial evaluation

Diagnosis. Bulimia nervosa is characterized by eating binges, followed by purging behaviors such as self-induced vomiting or laxative abuse1,2 (Table 1). It affects 1% to 3% of adolescent girls and young women and occurs in women 5 to 10 times more often than in men.

Bulimia is often persistent. About one-half of bulimic patients—including those who have been treated—continue to show eating disorder features on long-term follow-up.3,4

Psychiatric comorbidity. Most bulimic patients report a history of other psychiatric disorders, especially major depressive and bipolar disorders and anxiety disorders such as panic disorder, social phobia, and obsessive-compulsive disorder (OCD).5 Because these psychiatric comorbidities may occur before, during, or after bulimia nervosa, one cannot assume that mood or anxiety disorders are a cause or consequence of bulimia. Instead, bulimia nervosa, mood disorders, and anxiety disorders may be different expressions of a shared etiologic abnormality.

Table 1

DSM-IV-TR diagnostic criteria for bulimia nervosa

  1. Recurrent episodes of binge eating, characterized by both of the following:
  2. Recurrent inappropriate compensatory behavior in order to prevent weight gain, such as self-induced vomiting; misuse of laxatives, diuretics, enemas, or other medications; fasting; or excessive exercise
  3. The binge eating and inappropriate compensatory behaviors both occur, on average, at least twice a week for 3 months
  4. Self-evaluation is unduly influenced by body shape and weight
  5. The disturbance does not occur exclusively during episodes of anorexia nervosa
Specify type:
Purging type: during the current episode of bulimia nervosa, the person has regularly engaged in self-induced vomiting or the misuse of laxatives, diuretics, or enemas
Non-purging type: during the current episode of bulimia nervosa, the person has used other inappropriate compensatory behaviors, such as fasting or excessive exercise but has not regularly engaged in self-induced vomiting or the misuse of laxatives, diuretics, or enemas
Source: Reprinted with permission from the Diagnostic and Statistical Manual of Mental Disorders, 4th ed, text rev. Copyright 2000 American Psychiatric Association.

Evidence supporting this hypothesis comes from studies showing that these disorders:

  • respond to several chemically unrelated families of antidepressants6,7
  • frequently co-occur in individual patients5,7
  • frequently co-aggregate in families.7-9

We have published this evidence6,7 and proposed that bulimia nervosa may be one form of a larger underlying disorder, which we termed “affective spectrum disorder.”

Antidepressants are often rapidly effective in treating bulimic symptoms,10 regardless of whether patients exhibit depressive symptoms. Thus, there is no reason to withhold antidepressant therapy simply because a bulimic patient is not depressed. The term “antidepressant” may be a misnomer; these drugs are effective for numerous conditions, of which depression is only one.

Anorexic symptoms. Co-occurring depressive or anxiety disorders in a bulimic patient will not greatly alter treatment. The antidepressants and psychotherapies typically used to treat bulimia are often equally effective for affective disorders. Co-occurring anorexia nervosa, however, is a more serious concern.

Bulimic patients often display a history of anorexia nervosa; in many cases, the patient develops anorexia nervosa as a teenager and then progresses to bulimia nervosa across several years. Her prognosis is much better if her weight normalizes with the shift to bulimia nervosa, than if her weight remains well below normal for her height. It is unclear why medications and psychotherapy are much less effective in bulimic patients with anorexic symptoms than in those with bulimia alone. Watch for further details on anorexia nervosa as this series continues in future issues of.

Medical considerations. Potential medical complications—mostly consequences of vomiting or laxative use—are important to consider when you assess a bulimic patient:1

  • The acid in vomitus may gradually erode tooth enamel, requiring dental consultation.
  • Vomitus may inflame salivary gland ducts, though the swelling is usually benign.
  • Frequent vomiting may result in hypokalemia and alkalosis, although aggressive medical treatment usually is not needed.

Ask about ipecac use. To induce vomiting, some patients may abuse ipecac syrup, which can cause cardiomyopathy.11

Inpatient or outpatient? Unless the bulimic patient displays severe and medically dangerous anorexic symptoms, she can usually be treated as an outpatient. However, evaluate her carefully for suicidal ideation—which is not uncommon in bulimia nervosa—and consider inpatient treatment if necessary.

Medication vs. psychotherapy

The relative merits of medication versus psychotherapy in treating bulimia nervosa continue to be debated. The Cochrane Database of Systematic Reviews includes meta-analyses of both drug therapy12 and psychotherapy13 for bulimia nervosa. The 2001 drug therapy review found that “the use of a single antidepressant agent was clinically effective,” with no one drug clearly superior to another. Notably, this review was published before recent findings on topiramate.

 

 

The corresponding 2002 review of psychotherapy concludes—somewhat more cautiously—that “there is a small body of evidence for the efficacy of cognitive-behavior therapy in bulimia nervosa and similar syndromes, but the quality of trials is very variable and sample sizes are often small.”

In bulimia nervosa and other psychiatric disorders, comparing psychotherapy with drug therapy is hazardous because several factors bias the comparison in favor of psychotherapy. These factors include an expectational effect, a responsibility effect, and differential generalizability of study results.

Expectational effect. Patients in clinical trials are aware that they are receiving psychotherapy and, presumably, that study investigators hope to demonstrate its efficacy. This might account for much of psychotherapy’s apparent effect, as even placebos can produce 30% to 50% improvement in bulimia.14

Responsibility effect. If a patient fails to improve in a drug study, she will conclude that the drug has failed. But if she fails to improve in a psychotherapy study, she may conclude that she has failed. Because psychological treatments generally require patients to work in therapy, the patient may feel partially responsible for the outcome. Thus, to avoid cognitive dissonance, she may consciously or unconsciously exaggerate her improvement, both in her own mind and when reporting to treaters.

Differential generalizability. Psychological study protocols, such as administering several months of a behavioral treatment, usually mimic clinical practice fairly well. This is not the case with drug study protocols.

No responsible clinician would inflexibly administer a single dosage of a single drug for a fixed period to every bulimic patient and then declare failure for all nonresponders, as is done in study protocols. In practice, the clinician can offer nonresponders augmentation strategies and additional drug trials. Thus, calculations of bulimia response rates in drug studies substantially understate response to drug therapy in clinical practice.

Table 2

How effective are medications in treating bulimia nervosa?

MedicationEvidence for efficacyRemarks
Antidepressants
Selective serotonin reuptake inhibitors+++Fluoxetine is only SSRI studied in controlled trials
Tricyclics+++Generally more side effects than SSRIs
Monoamine oxidase inhibitors++High rates of remission, but dietary restrictions
Trazodone++Only one controlled trial
Venlafaxine, mirtazapine, nefazodone?No controlled trials, but probably effective
Bupropion(++)Not recommended; caused seizures in bulimic patients
Anticonvulsants
Topiramate++Only one controlled trial, but substantial effect size
Phenytoin+Little efficacy in only controlled study
Carbamazepine+May be useful in bulimia with comorbid bipolar disorder
Valproate+May be useful in bulimia with comorbid bipolar disorder
Other agents
Liothyronine+Augmentation agent in patients with incomplete antidepressant response
Lithium+Ineffective in only controlled trial; possible augmentation strategy
Naltrexone0Ineffective in two controlled trials
Ondansetron+One controlled trial
0 No apparent efficacy
+ Occasional effect; limited evidence
++ Clear effect; good evidence from controlled trial(s)
+++ Strongly documented effect; evidence from multiple controlled trials.
( ) Negative effect

One also might note that psychological study findings have not “sold” well in the clinical practice marketplace. For example, in a recent survey of more than 220 bulimic women treated with psychotherapy, only 6.9% said they received a full course of cognitive behavioral therapy (CBT)14 —despite two decades of evidence of its efficacy. By contrast, untested, inefficacious, and possibly harmful psychotherapies for bulimia—including recovered-memory therapy—appear to be thriving.

Recommendation. Interpret with caution any head-to-head comparisons of psychological versus drug therapies—especially when clinical practice recommendations are made. Certain psychological therapies provided by specifically-trained individuals likely do help patients with bulimia nervosa. However, biases inherent to the studies may inflate psychological therapies’ efficacy when compared with that of drug therapy.

Therefore, for a psychiatrist who does not specialize in eating disorders to offer exclusively psychological therapy to a bulimic patient—while withholding or postponing drug therapy—may now be a questionable practice.

Choosing drug therapies

Although consensus is lacking on an optimal treatment trial sequence for bulimia nervosa, we suggest a rational approach based on the evidence and our experience (Algorithm).

First-line antidepressants. A selective serotonin reuptake inhibitor (SSRI) trial is usually the first choice (Table 2), and some data suggest that higher-than-usual dosages may be required. For example, in a large multicenter trial of fluoxetine in bulimia nervosa, 60 mg/d was considerably more effective than 20 mg/d for reducing binge eating behavior and vomiting frequency.15

Based on our observations, however, we believe that noncompliance or irregular compliance may account for this difference in response. Bulimic patients’ impulsive and obsessional behavior may keep them from taking their medications as prescribed. The higher fluoxetine dosage may therefore have been more effective simply because it ensured adequate plasma levels, even when patients missed or forgot multiple doses.

Augmenting agents. A first antidepressant trial rarely leads to complete remission of bulimic symptoms. This is not a serious concern, however, because many other options are available.

Liothyronine. Partial responders to SSRIs often become complete responders when we add a 10-day trial of liothyronine (T3), 25 μg/d. If this fails, we may try augmenting with lithium carbonate, although bulimic patients are often afraid of weight gain or lithium’s other side effects.

 

 

Topiramate. A newer augmentation strategy is to add the anticonvulsant topiramate. Used alone, topiramate demonstrated effectiveness for bulimia nervosa in one placebo-controlled, double-blind trial.16

Adding topiramate to an antidepressant regimen will likely reduce any remaining bulimic symptoms. In addition, topiramate often produces weight loss—a side effect that bulimic patients usually welcome. It remains unclear whether topiramate’s weight-loss effects might pose a hazard in patients with simultaneous bulimic and anorexic symptoms.

Other antidepressants. If the above strategies fail, other antidepressant options include venlafaxine, tricyclics, and monoamine oxidase inhibitors. Bupropion is not recommended in bulimia nervosa; one trial17 of this agent resulted in a much higher rate of grand mal seizures in bulimic patients than in patients taking bupropion for depression.

In bulimic patients with concomitant bipolar disorder, the anticonvulsants carbamazepine and valproate often reduce affective and bulimic symptoms. By contrast, the anticonvulsant phenytoin—once thought to be useful in bulimia nervosa10 —offers little benefit for either bulimic or affective symptoms.

Persistence is important when initial medication trials fail. One unblinded study followed 36 bulimic patients 9 to 19 months after they completed a controlled study with trazodone.18 Of the 26 patients who tried a second or third antidepressant, 17 (65%) achieved remission of bulimia on follow-up. Of the 10 patients who declined a second or third trial, only 1 (10%) attained remission.

Notably, these study results were obtained before the SSRIs and other newer antidepressants or topiramate became available. Cooperative patients using present-day medications might be able to achieve remission rates that exceed 65%.

Algorithm Proposed treatment approach to bulimia nervosa



Table 3

How effective are psychotherapies in treating bulimia nervosa?

PsychotherapyEvidence for efficacyRemarks
Cognitive behavioral therapy (CBT)+++Controlled evidence for efficacy in individual and group treatment
Interpersonal psychotherapy (IPT)++Effective, but slower than CBT
Exposure with response prevention+May be added to other behavioral techniques, though additive benefit questionable
Dialectical behavior therapy+Highly structured behavioral technique originally developed for borderline personality disorder
Self-help groups+Frequently considered very helpful by patients
Psychodynamic psychotherapy0“Recovered memory” approaches are frankly harmful
Eye movement desensitization and reprocessing (EMDR)0Dubious theoretical basis; no methodologically acceptable evidence for efficacy
0 No apparent efficacy
+ Occasional effect; limited evidence
++ Clear effect; good evidence from controlled trial(s)
+++ Strongly documented effect; evidence from multiple controlled trials.

Psychotherapy

Cognitive-behavioral therapy. CBT—given either individually or in groups—is the most effective psychotherapy for bulimia (Table 3).19 CBT typically involves 3 to 6 months of helping the patient focus on her bulimic behaviors and on specific attitudes—such as unrealistic preoccupations with being “too fat”—that perpetuate the behaviors.

In practice, unfortunately, few bulimic patients are offered CBT, perhaps because few clinicians are trained in the specific approach used for bulimia nervosa.19 If you are not trained in using CBT for bulimia and do not have access to colleagues who offer this treatment, you may begin with medication plus simple behavioral treatments, such as:

  • offering supportive therapy in the office
  • referring patients to self-help groups for persons with eating disorders.

If this strategy fails, encourage patients to consider CBT—even if they must travel some distance to obtain it.

Other specialized psychotherapies. Dialectical behavior therapy and interpersonal psychotherapy appear to be effective in bulimia. Again, however, clinicians who lack training in these techniques or access to local experts may be unable to offer them. Psychodynamic therapy does not appear to offer greater benefit in bulimia nervosa than ordinary supportive counseling.

Dubious therapies. One psychodynamic approach—regrettably still practiced—is “recovered memory therapy.” Therapists who use it claim that childhood sexual abuse or other trauma can cause bulimic symptoms but patients have repressed the memory of these events.20

No methodologically sound evidence has shown that childhood sexual abuse can cause bulimia nervosa years or decades later.21 Nor is there acceptable evidence that people can repress the memory of a traumatic experience.22 Therapists administering recovered memory therapy have been subjected to malpractice judgments totaling tens of millions of dollars from suits filed by patients who eventually realized that so-called “recovered” memories were false.23

Another dubious therapy—eye movement desensitization and reprocessing (EMDR)—also may involve attempts to “recover” memories of putative traumatic events.24 No methodologically sound evidence has shown that EMDR is effective in bulimic patients, and the technique’s theoretical basis is questionable.24,25

Related resources

Drug brand names

  • Bupropion • Wellbutrin
  • Citalopram • Celexa
  • Desipramine • Norpramin
  • Fluoxetine • Prozac
  • Lithium • Lithobid, Eskalith
  • Nortriptyline • Pamelor, Aventyl
  • Sertraline • Zoloft
  • Topiramate • Topamax
  • Trazodone • Desyrel
  • Liothyronine • Cytomel
  • Venlafaxine • Effexor

Disclosure

Dr. Pope receives research support from Ortho-McNeil Pharmaceuticals and is a consultant to Solvay Pharmaceuticals and Auxilium Pharmaceuticals.

 

 

Dr. Hudson receives research support from and is a consultant to Eli Lilly & Co. and Ortho-McNeil Pharmaceuticals.

References

1. Mehler PS. Clinical practice. Bulimia nervosa. N Engl J Med 2003;349(9):875-81.

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

3. Fisher M. The course and outcome of eating disorders in adults and in adolescents: a review. Adolesc Med 2003;14(1):149-58.

4. Fairburn CG, Norman PA, Welch SL, et al. A prospective study of outcome in bulimia nervosa and the long-term effects of three psychological treatments. Arch Gen Psychiatry 1995;52(4):304-12.

5. Hudson JI, Pope HG, Jr, Yurgelun-Todd D, et al. A controlled study of lifetime prevalence of affective and other psychiatric disorders in bulimic outpatients. Am J Psychiatry 1987;144(10):1283-7.

6. Hudson JI, Pope HG, Jr. Affective spectrum disorder: does antidepressant response identify a family of disorders with a common pathophysiology? Am J Psychiatry 1990;147(5):552-64.

7. Hudson JI, Mangweth B, Pope HG, Jr, et al. Family study of affective spectrum disorder. Arch Gen Psychiatry 2003;60:170-7.

8. Hudson JI, Laird NM, Betensky RA, et al. Multivariate logistic regression for familial aggregation of two disorders: II. Analysis of studies of eating and mood disorders. Am J Epidemiology 2001;153(5):506-14.

9. Mangweth B, Hudson JI, Pope HG, Jr, et al. Family study of the aggregation of eating disorders and mood disorders. Psychol Med (in press).

10. Hudson JI, Pope HG, Jr, Carter WP. Pharmacologic therapy of bulimia nervosa. In: Goldstein D (ed). The management of eating disorders and obesity (2nd ed) Totowa, NJ: Humana Press, Inc.(in press).

11. Pope HG, Jr, Hudson JI, Nixon RA, Herridge PL. The epidemiology of ipecac abuse. N Engl J Med 1986;14(4):245-6.

12. Hay PJ, Bacaltchuk J. Psychotherapy for bulimia nervosa and binging. Cochrane Database Syst Rev 2003;(1):CD000562.-

13. Bacaltchuk J, Hay P. Antidepressants versus placebo for people with bulimia nervosa. Cochrane Database Syst Rev 2001;(4):CD003391.-

14. Crow S, Mussell MP, Peterson C, et al. Prior treatment received by patients with bulimia nervosa. Int J Eating Disord 1999;25(1):39-44.

15. Fluoxetine Bulimia Collaborative Study Group Fluoxetine in the treatment of bulimia nervosa: a multicenter placebo-controlled, double-blind trial. Arch Gen Psychiatry 1992;49:139-47.

16. Hoopes S, Reimherr F, Hedges D, et al. Part 1:Topiramate in the treatment of bulimia nervosa: a randomized, double-blind, placebocontrolled trial. J Clin Psychiatry (in press).

17. Horne RL, Ferguson JM, Pope HG, Jr, et al. Treatment of bulimia with bupropion: a controlled multi-center trial. J Clin Psychiatry 1988;49(7):262-6.

18. Pope HG, Jr, McElroy SL, Keck PE, Jr, Hudson JI. Long-term pharmacotherapy of bulimia nervosa. J Clin Psychopharmacol 1989;9(5):385-6.

19. Fairburn CG, Harrison PJ. Eating disorders. Lancet 2003;361(9355):407-16.

20. Pope HG, Jr, Hudson JI. “Recovered memory” therapy for eating disorders: implications of the Ramona verdict. Int J Eat Disord 1996;19(2):139-45.

21. Pope HG, Jr, Hudson JI. Does childhood sexual abuse cause adult psychiatric disorders? Essentials of methodology. J Psychiatry Law 1995;Fall:363-81.

22. Pope HG, Jr, Oliva PS, Hudson JI. Repressed memories. The scientific status of research on repressed memories. In: Faigman DL, Kaye DH, Saks MJ, Sanders J (eds). Science in the law: social and behavioral science issues St. Paul, MN: West Group, 2002;487-526.

23. Cannell J, Hudson JI, Pope HG, Jr. Standards for informed consent in recovered memory therapy. J Am Acad Psychiatry Law 2001;29(2):138-47.

24. Hudson JI, Chase EA, Pope HG, Jr. Eye movement desensitization and reprocessing in eating disorders: caution against premature acceptance. Int J Eat Disord 1998;23:1-5.

25. McNally RJ. EMDR and mesmerism: a comparative historical analysis. J Anxiety Disord 1999;13(1-2):225-36.

References

1. Mehler PS. Clinical practice. Bulimia nervosa. N Engl J Med 2003;349(9):875-81.

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

3. Fisher M. The course and outcome of eating disorders in adults and in adolescents: a review. Adolesc Med 2003;14(1):149-58.

4. Fairburn CG, Norman PA, Welch SL, et al. A prospective study of outcome in bulimia nervosa and the long-term effects of three psychological treatments. Arch Gen Psychiatry 1995;52(4):304-12.

5. Hudson JI, Pope HG, Jr, Yurgelun-Todd D, et al. A controlled study of lifetime prevalence of affective and other psychiatric disorders in bulimic outpatients. Am J Psychiatry 1987;144(10):1283-7.

6. Hudson JI, Pope HG, Jr. Affective spectrum disorder: does antidepressant response identify a family of disorders with a common pathophysiology? Am J Psychiatry 1990;147(5):552-64.

7. Hudson JI, Mangweth B, Pope HG, Jr, et al. Family study of affective spectrum disorder. Arch Gen Psychiatry 2003;60:170-7.

8. Hudson JI, Laird NM, Betensky RA, et al. Multivariate logistic regression for familial aggregation of two disorders: II. Analysis of studies of eating and mood disorders. Am J Epidemiology 2001;153(5):506-14.

9. Mangweth B, Hudson JI, Pope HG, Jr, et al. Family study of the aggregation of eating disorders and mood disorders. Psychol Med (in press).

10. Hudson JI, Pope HG, Jr, Carter WP. Pharmacologic therapy of bulimia nervosa. In: Goldstein D (ed). The management of eating disorders and obesity (2nd ed) Totowa, NJ: Humana Press, Inc.(in press).

11. Pope HG, Jr, Hudson JI, Nixon RA, Herridge PL. The epidemiology of ipecac abuse. N Engl J Med 1986;14(4):245-6.

12. Hay PJ, Bacaltchuk J. Psychotherapy for bulimia nervosa and binging. Cochrane Database Syst Rev 2003;(1):CD000562.-

13. Bacaltchuk J, Hay P. Antidepressants versus placebo for people with bulimia nervosa. Cochrane Database Syst Rev 2001;(4):CD003391.-

14. Crow S, Mussell MP, Peterson C, et al. Prior treatment received by patients with bulimia nervosa. Int J Eating Disord 1999;25(1):39-44.

15. Fluoxetine Bulimia Collaborative Study Group Fluoxetine in the treatment of bulimia nervosa: a multicenter placebo-controlled, double-blind trial. Arch Gen Psychiatry 1992;49:139-47.

16. Hoopes S, Reimherr F, Hedges D, et al. Part 1:Topiramate in the treatment of bulimia nervosa: a randomized, double-blind, placebocontrolled trial. J Clin Psychiatry (in press).

17. Horne RL, Ferguson JM, Pope HG, Jr, et al. Treatment of bulimia with bupropion: a controlled multi-center trial. J Clin Psychiatry 1988;49(7):262-6.

18. Pope HG, Jr, McElroy SL, Keck PE, Jr, Hudson JI. Long-term pharmacotherapy of bulimia nervosa. J Clin Psychopharmacol 1989;9(5):385-6.

19. Fairburn CG, Harrison PJ. Eating disorders. Lancet 2003;361(9355):407-16.

20. Pope HG, Jr, Hudson JI. “Recovered memory” therapy for eating disorders: implications of the Ramona verdict. Int J Eat Disord 1996;19(2):139-45.

21. Pope HG, Jr, Hudson JI. Does childhood sexual abuse cause adult psychiatric disorders? Essentials of methodology. J Psychiatry Law 1995;Fall:363-81.

22. Pope HG, Jr, Oliva PS, Hudson JI. Repressed memories. The scientific status of research on repressed memories. In: Faigman DL, Kaye DH, Saks MJ, Sanders J (eds). Science in the law: social and behavioral science issues St. Paul, MN: West Group, 2002;487-526.

23. Cannell J, Hudson JI, Pope HG, Jr. Standards for informed consent in recovered memory therapy. J Am Acad Psychiatry Law 2001;29(2):138-47.

24. Hudson JI, Chase EA, Pope HG, Jr. Eye movement desensitization and reprocessing in eating disorders: caution against premature acceptance. Int J Eat Disord 1998;23:1-5.

25. McNally RJ. EMDR and mesmerism: a comparative historical analysis. J Anxiety Disord 1999;13(1-2):225-36.

Issue
Current Psychiatry - 03(01)
Issue
Current Psychiatry - 03(01)
Page Number
12-22
Page Number
12-22
Publications
Publications
Topics
Article Type
Display Headline
Update on eating disorders Bulimia nervosa: Persistent disorder requires equally persistent treatment
Display Headline
Update on eating disorders Bulimia nervosa: Persistent disorder requires equally persistent treatment
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Update on bipolar disorder: How to better predict response to maintenance therapy

Article Type
Changed
Mon, 04/16/2018 - 14:28
Display Headline
Update on bipolar disorder: How to better predict response to maintenance therapy

What are we trying to accomplish in the maintenance treatment of patients with bipolar disorder? Given that the disorder recurs in more than 90% of patients who experience a manic episode,1 there are 5 important goals:

  1. Prevention of recurrent episodes;
  2. Amelioration of subsyndromal symptoms;
  3. Reduction of suicide risk;
  4. Compliance enhancement;
  5. Optimization of interpersonal, social, and vocational functioning.

There is a great premium on preventing mood episode recurrence in patients with bipolar disorder. Mood episodes themselves produce substantial morbidity, but morbidity is not confined to these episodes alone. Full recovery of functioning often lags many months behind remission of symptoms.2 Recurrent mood episodes also may lead to progressive loss of function between episodes. Mood episodes also carry risks of mortality from suicide, violence, and impulsive risk taking.

Clearly, mood-stabilizing medications form the cornerstone of maintenance treatment,3,4 along with a strong therapeutic alliance between patient and clinician and targeted psychosocial therapies. In the Expert Consensus Guidelines for medication treatment of bipolar I disorder, maintenance treatment was recommended for 1 year following an initial manic or mixed episode; longer (indefinite) treatment was recommended for patients with a family history of bipolar disorder or if 2 episodes occurred.3

Compared with clinical trials of agents for acute bipolar mania (and mixed episodes), there are relatively few randomized controlled trials of medications for the maintenance phase of bipolar disorder. Some naturalistic studies have provided data on relapse rates associated with treatment with a variety of different agents. Even fewer studies have examined psychosocial interventions designed specifically to reduce relapse rates.

But new data are beginning to emerge regarding the efficacy of divalproex, lamotrigine, and olanzapine as maintenance therapies. A number of clinical predictors of response to these agents have begun to be identified, as well as to lithium and carbamazepine. Eliciting these characteristics is important when making recommendations to patients about available drug therapies.

In addition, effective maintenance treatment often requires combinations of mood-stabilizing, antidepressant, and antipsychotic agents to control or eliminate subsyndromal and breakthrough symptoms.

In this review, we will cover the available new data on pharmacologic maintenance treatments of bipolar disorder and their clinical implications.

What the studies show

Lithium has been the mainstay of therapy for bipolar disorder for more than 35 years. Most randomized, controlled trials of lithium maintenance therapy were conducted in the 1960s and 1970s5 (Table 1). Unfortunately, these studies had several design limitations that inflated the expectations of lithium’s efficacy as a maintenance treatment.6 These included discontinuation designs in which patients stabilized on lithium were abruptly switched to placebo; exaggerated early placebo relapse rates; enrollment of both unipolar and bipolar patients; lack of specific diagnostic criteria; and reported results only for patients completing studies. Pooled data from these trials indicated that lithium reduced the risk of relapse fourfold compared with placebo at 6 months and 1 year.5

Two contemporary randomized, placebo-controlled maintenance studies utilizing more rigorous designs provided further evidence of lithium’s superiority over placebo in extending time to manic relapse.7,8 Both studies enrolled patients who were currently or had recently been manic and had been stabilized in open-label treatment that included study medications.

In the first study, comparing 1-year relapse rates among patients randomized to lithium, divalproex, or placebo, lithium extended time to recurrence of mania by 55% compared with placebo.7 In the second, an 18-month trial comparing lithium, lamotrigine, and placebo, lithium significantly increased the time to intervention for recurrence of mania compared with placebo.8 The overall manic relapse rates were 17% for lithium-treated patients and 41% for those on placebo. However, lithium did not significantly extend time to depressive relapse or intervention for depressive relapse, respectively, in either study. Moreover, in the first study, patients who received lithium tended to have greater subthreshold depressive symptoms.8

Table 1

What works in maintenance treatment of bipolar disorder (ranked by number of randomized, controlled trials)

Medication RCTs (n)Trial resultsStrong evidenceSome evidence
Lithium (15)Equal to divalproex and lamotrigine; equal to or better than carbamazepine; better than placebo4-fold reduction of relapse risk vs. placebo at 6, 12 months; more efficacious in mania than in depression; higher lithium level correlated with lower relapse rates, but more side effects 
Carbamazepine (7)No significant benefit vs. lithium; better than placebo May be more efficacious in mania than in depression; may be less tolerable than lithium; no data on serum levels
Divalproex (4)Equal to lithium and olanzapine; better than placebo May be more efficacious in mania than in depression; may be more tolerable than lithium; data on serum levels pending
Lamotrigine (2)Better than placeboMore efficacious in depression than in maniaDosage 200-400 mg/d
Olanzapine (1)Equal to divalproex Data on dosage and differential efficacy related to dosage pending
Clozapine (1)Better than treatment as usualEfficacy in treatment-resistant mania (bipolar schizoaffective disorder, bipolar type) 
 

 

These findings were consistent with earlier placebo-controlled studies of lithium maintenance treatment and a recent crossover comparison trial with carbamazepine.9 Lithium was also recently compared with carbamazepine in a 2.5-year maintenance multisite study in Europe.10 There was no significant difference in efficacy between the two in time to hospitalization, the primary outcome measure. On other outcome measures, including time to relapse or need for additional medication, lithium was superior to carbamazepine.

Pooled results from a number of naturalistic studies, which mirror clinical practice, indicated that approximately one-third of patients maintained on lithium had good functional outcomes without relapses and only minimal symptoms.11 In general, these studies found higher rates of relapse with longer durations of follow-up.

Valproate maintenance treatment has been studied in two randomized, controlled trials (one vs. placebo,7 and one vs. olanzapine12) and two open-label comparison studies against lithium.13,14 In the placebo-controlled trial, which also included a lithium comparison group described earlier, there was no significant difference in the time to development of any mood episode among the three treatment groups.7 However, divalproex was superior to placebo on a number of other outcome measures, including rate of study termination for any mood episode, termination for depression, and termination for noncompliance.

Divalproex was superior to placebo in patients who received divalproex in the open-label treatment phase before randomization. This is clinically relevant, as this group reflects expected relapse rates in patients treated initially with divalproex for acute mania who then remain on the drug for maintenance therapy. Patients receiving divalproex had significantly lower rates of intolerance and noncompliance compared to those treated with lithium.

In the second comparison study, 167 patients initially randomized and responding to divalproex or olanzapine in a 3-week acute bipolar mania trial continued in a double-blind 44-week extension study.12 There were no significant differences in relapse into mania between the two groups (olanzapine 41%, divalproex 50%, p = 0.4) or time to manic relapse (olanzapine 270 days, divalproex 74 days, p = 0.4). There was no significant difference in tolerability between the two.

Two open-label studies compared valproate with lithium. In an 18-month study conducted in France, patients randomized to the valpromide formulation of valproate displayed a 20% lower relapse rate than those receiving lithium.13 The second open-label comparison trial found comparable efficacy between lithium and divalproex in a 1-year naturalistic pharmacoeconomic study that allowed additional medications as needed for recurrent symptoms.14

Carbamazepine has been evaluated in a limited number of studies for maintenance treatment of bipolar disorder. The results of many early randomized, controlled maintenance trials were criticized on methodologic grounds.15 But two recent comparison trials with lithium, as noted earlier, provided clinically important data regarding carbamazepine’s prophylactic efficacy.9,10 Several additional clinically relevant observations emerged from analyses of secondary outcome measures in these two trials.

In the first study, 52 patients with bipolar I or II disorder received lithium or carbamazepine for 1 year, crossed over to the alternate drug the second year, and received both drugs the third year.9 There was little difference in relapse rates during the first year between the lithium (31%) and carbamazepine (37%) groups. Similarly, in the overall trial, there was little difference in the percentage of patients who were rated as having a moderate or better response—33% on lithium, 31% on carbamazepine, and 55% on the combination. A higher proportion of patients receiving carbamazepine withdrew due to side effects. In the second trial, significantly more patients receiving carbamazepine required additional medications for breakthrough symptoms and experienced side effects requiring treatment discontinuation.10

Lamotrigine has been studied in two placebo-controlled, randomized maintenance studies in patients with bipolar disorder.8,16 The first evaluated bipolar I patients who had experienced a manic or hypomanic episode within 60 days of entry into an open-label treatment phase with lamotrigine.8 Patients who, in turn, improved or remained stable during the open-label treatment phase were then randomized to treatment with lamotrigine 200-400 mg/d, lithium, or placebo for up to 18 months. Both lamotrigine and lithium were superior to placebo on the primary outcome measure, which was time to need for additional medication for a mood episode. The median time until 25% of patients relapsed was 72 weeks for lamotrigine, 58 weeks for lithium, and 35 weeks for placebo.

Table 2

PUTATIVE PREDICTORS OF RESPONSE TO MAINTENANCE MEDICATIONS

MedicationPredictor of responseStrength of evidence
LithiumNonrap id cycling
Few episodes
Few depressive symptoms
Family history bipolar disorder
Episode sequence M-D-I
No substance/alcohol use disorder
◊◊◊




DivalproexEqual efficacy in rapid & non-rapid cycling, manic & mixed,
No personality disorder

CarbamazepineEqual efficacy in rapid & non-rapid cycling, manic & mixed
Mood-incongruent symptoms
◊◊
LamotrigineBipolar II > bipolar I
Depression > mania

◊◊
OlanzapineEqual efficacy in rapid & non-rapid cycling, manic & mixed, psychotic & nonpsychotic mania in acute studies; data pending in maintenance
ClozapineEfficacy in treatment-resistant mania rapid & nonrapid cycling, manic & mixed, psychotic & nonpsychotic in naturalistic studies◊◊◊
Key ◊ = reported in 1 study; ◊◊ = reported in 2 studies; ◊◊◊ = reported in > 3 studies.
 

 

On secondary outcome measures, lamotrigine, but not lithium, was superior to placebo in delaying time to depressive relapse. In contrast, lithium, but not lamotrigine, was superior to placebo in delaying time to manic relapse. Finally, lamotrigine, but not lithium, was superior to placebo in time to discontinuation for any reason.

From this study, it appears that lamotrigine is most effective in preventing depressive relapse, whereas lithium is most effective in preventing manic relapse. Thus, lithium and lamotrigine may complement each other in maintenance treatment by preventing manic and depressive episodes in combination.

The findings of the first trial were consistent with those of the second placebo-controlled lamotrigine (100-500 mg/d) maintenance study conducted specifically in patients with rapid cycling bipolar I and II disorders.16 In this 6-month trial, there was no significant difference in time to need for additional medications, the primary outcome measure, between the lamotrigine and placebo groups.

Median survival time was significantly greater for the lamotrigine group (18 weeks) than for the placebo group (12 weeks). The lamotrigine group had significantly longer time to need for additional medications in bipolar II, but not bipolar I, patients. Patients with bipolar II disorder also displayed significantly greater improvement with lamotrigine on global scales compared with bipolar I patients. Because bipolar II disorder is characterized predominantly by depressive episodes, these findings suggest that lamotrigine was again more beneficial in preventing recurrent depression, in this case, specifically, in rapid cycling bipolar II patients.

Taken together, the results of these studies are also consistent with the results of two placebo-controlled trials of lamotrigine in the treatment of acute bipolar depression.17,18 Its efficacy in acute bipolar I depression was first demonstrated in a 6-week, double-blind, randomized, parallel-group trial in which patients received lamotrigine 50 mg/d, 200 mg/d (after gradual titration over the first 4 weeks), and placebo.17

Both dosage groups of lamotrigine displayed significantly greater improvement in depressive symptoms as measured by the Montgomery-Asberg Depression Rating Scale (but not the Hamilton Depression Rating Scale) and by the Clinical Global Improvement Scale compared with the placebo group. There was a trend for the 200 mg/d group to have greater improvement than the 50 mg/d group, a trend that might have become significant had the study been longer, as the 200 mg/d group did not receive this dose for the full trial. There was no significant difference in the incidence of hypomanic or manic switches among the three study groups.

Frye and colleagues also found lamotrigine to be superior to placebo in global improvement in depressive (but not manic) symptoms in a crossover monotherapy trial in treatment-refractory bipolar I and II patients.18

Olanzapine, as described earlier, was comparable to divalproex in a head-to-head comparison 44-week extension trial in patients who responded to double-blind treatment in an acute mania study.12 These results were consistent with the preliminary findings of an open-label extension study of olanzapine.19 There are no placebo-controlled trials of olanzapine or any other atypical antipsychotic in the maintenance phase of bipolar disorder published to date.

Clozapine was more effective than “treatment as usual” (combinations of mood-stabilizers and typical antipsychotics) in patients with treatment-refractory bipolar and schizoaffective (bipolar subtype) disorders.20 Many patients with bipolar disorder now receive adjunctive maintenance treatment with typical antipsychotics, but the limited data from the few controlled trials of typical agents administered in combination with mood-stabilizers do not support the efficacy of this strategy.21 Moreover, some reports suggest that maintenance treatment incorporating typical antipsychotics may increase the frequency and severity of depressive episodes in patients with bipolar disorder.

Predicting response to pharmacologic treatment

Clinical experience and data from the randomized, controlled trials reviewed earlier have identified several tentative predictors of response—or nonresponse—to specific medications. Lithium, divalproex, and carbamazepine appear to have greater efficacy in the prevention of manic rather than depressive episodes.11 In contrast, lamotrigine appears to have greater efficacy in preventing depressive rather than manic episodes.

It is unclear whether olanzapine and perhaps other atypical antipsychotics may have a more favorable effect in preventing one pole of the illness over another (Table 2). The currently available atypicals (clozapine, risperidone, olanzapine, quetiapine, and ziprasidone) in general appear to differ from typical antipsychotics in having bidirectional (antimanic and antidepressant) effects on mood symptoms.

Atypicals may exert antidepressant effects via a number of different mechanisms, including 5HT2 receptor antagonism (a property shared by all atypicals and the antidepressants trazodone, nefazodone, and mirtazapine); alpha2-adrenergic antagonism (clozapine and risperidone); and 5HT1D antagonism, 5HT1A agonism, and 5HT and NE reuptake inhibition (ziprasidone).22

Patients with rapid-cycling bipolar disorder have a relatively poor response to lithium (response rates of approximately 20%).11 Patients with rapid-cycling bipolar I disorder may have a greater likelihood of response to combined treatment with lithium and carbamazepine,9 or divalproex.11 Alternately, patients with rapid-cycling bipolar II disorder have lower relapse rates on lamotrigine compared with placebo.16 Anecdotal reports and the results of acute treatment studies suggest that clozapine and olanzapine may be beneficial maintenance agents for rapid-cycling bipolar I patients.

 

 

Other predictors of favorable response to lithium prophylaxis include a family history of bipolar disorder, illness course characterized by maniadepression-euthymia episode sequence, and few prior mood episodes.21 Conversely, co-occurring alcohol or substance use disorder, multiple prior mood episodes, and familial negative affective style have been associated with poor response to lithium maintenance treatment.

Tentative predictors of response to divalproex maintenance treatment include mixed episodes, rapid cycling, and absence of co-occurring personality disorder. In one lithium comparison trial, patients with mixed episodes, bipolar II, and NOS disorders and mood-incongruent symptoms appeared to have a better response to carbamazepine.10

The dosage and relevant plasma concentrations of maintenance agents may also affect long-term efficacy. Gelenberg et al observed that the risk of relapse in bipolar patients maintained on low (0.4-0.6 mmol/L) serum concentrations was 2.6 times higher than for patients maintained on high (0.8-1.0 mmol/L) serum concentrations.23 However, there was a tradeoff in tolerability. Patients treated at the higher concentrations experienced significantly more and often treatment-limiting side effects.

No data are available regarding a possible plasma concentration-response relationship between maintenance treatment with divalproex or carbamazepine, but based on data from acute treatment trials, concentrations well within the therapeutic range of each drug appear essential.

The problem of subsyndromal symptoms

Evidence from a number of long-term outcome studies in bipolar disorder indicates that many patients spend protracted periods of time neither well nor in syndromal manic or depressive episodes, but rather experiencing chronic subsyndromal symptoms.24,25 In these studies, depressive symptoms were twice as prevalent as hypomanic symptoms between acute episodes of illness. Furthermore, persistent subsyndromal depressive symptoms were strongly associated with an increased risk for relapse and poor occupational functioning.

Keller et al26 found that patients maintained on low lithium serum concentrations (0.4-0.6 mmol/L) were more likely to experience subsyndromal symptoms and that their symptoms were more likely to worsen at any time than were symptoms of patients maintained at higher serum concentrations (0.8-10 mmol/L). The first occurrence of subsyndromal symptoms increased the risk of fullepisode relapse fourfold. These findings indicate that the optimal pharmacological maintenance treatment of bipolar disorder requires titration of mood-stabilizer medications to eradicate subsyndromal symptoms. Eliminating these residual or recurrent symptoms, in turn, substantially decreases the risk of relapse and of enduring functional impairment.

Conventional wisdom, in part supported by limited data from a small number of studies, suggests that antidepressants be used sparingly and for limited periods of time in conjunction with mood-stabilizers for bipolar depression. However, new data indicate that this strategy may significantly increase the risk of recurrence of depressive symptoms and episodes.27 Thus, combined treatment with mood-stabilizing agents and antidepressants for patients without rapid cycling and with recurrent depressive episodes may be indicated more often than previously thought.

In practice, perhaps the majority of patients with bipolar disorder require treatment with more than one mood-stabilizing medication to suppress subsyndromal symptoms as well as to prevent full episode recurrence. The use of combinations has been very poorly studied in randomized, controlled trials. In a pilot study of 12 patients with bipolar I disorder, Solomon et al compared the efficacy of lithium alone versus the combination of lithium and divalproex for 1 year.28 Not surprisingly, the combination significantly reduced the risk of recurrence of mania and depression but was also associated with more bothersome side effects.

Clinical practice has greatly outstripped the limited data available from formal studies. Recently reported as useful maintenance treatment strategies are combinations of:

  • lithium and divalproex
  • lithium and carbamazepine
  • divalproex and carbamazepine
  • lithium, divalproex, and carbamazepine
  • lithium and/or divalproex with atypical antipsychotics, antidepressants, and lamotrigine.

Related resources

  • Nathan PE, Gorman JM, eds. A Guide to Treatments that Work. 2nd ed. New York: Oxford University Press, 2001.
  • Buckley PF, Waddington JL, eds. Schizophrenia and Mood Disorders: the New Drug Therapies in Clinical Practice. Boston: Butterworth-Heiman, 2000.
  • Goldberg JF, Harrow M. Bipolar Disorders. Clinical Course and Outcome. Washington, DC: American Psychiatric Press, 1999.
  • Sachs GS, Thase ME. Bipolar disorder therapeutics: maintenance treatment. Biol Psychiatry 2000;48:573-81.
  • Hirschfeld RMA, Bowden CL, Gitlin MJ, et al. Practice guideline for the treatment of bipolar disorder (revision). Am J Psychiatry, in press.

Drug brand names

  • Clozapine • Clozaril
  • Divalproex • Depakote, Depakote Sprinkle
  • Lamotrigine • Lamictal
  • Mirtazapine • Remeron, Remeron Soltab
  • Nefazodone • Serzone
  • Olanzapine • Zyprexa
  • Quetiapine • Seroquel
  • Risperidone • Risperdal
  • Trazodone • Desyrel
  • Valproate sodium • Depacon
  • Ziprasidone • Geodon

Disclosure

Dr. Keck reports that he receives grant/research support from and serves as a consultant to Abbott Laboratories, AstraZeneca, Pfizer Inc., and Eli Lilly and Co. He also receives grant/research support from Merck and Co. and Otsuka America Pharmaceutical, and serves as a consultant to Bristol-Myers Squibb Co., GlaxoSmithKline, and Janssen Pharmaceutica.

 

 

Dr. McElroy reports that she receives grant/research support from and serves as a consultant to Abbott Laboratories, Elan Pharmaceuticals, Cephalon Inc. GlaxoSmithKline, and Eli Lilly and Co. She also receives grant/research support from Forest Pharmaceuticals and Solvay Pharmaceuticals, and serves as a consultant to Bristol-Myers Squibb Co., Ortho-McNeil Pharmaceutical, and Janssen Pharmaceutica.

Dr. Nelson reports no financial relationship with any company whose products are mentioned in this article.

References

1. Solomon DA, Keitner GI, Miller IW, et al. Course of illness and maintenance treatments for patients with bipolar disorder. J Clin Psychiatry 1995;56:5-13.

2. Keck PE, Jr, McElroy SL, et al. Twelve-month outcome of bipolar patients following hospitalization for a manic or mixed episode. Am J Psychiatry 1998;155:646-52.

3. Sachs GS, Printz DJ, et al. The Expert Consensus Guideline Series: medication treatment of bipolar disorders 2000. Postgrad Med Special Report 2000;4:1-104.

4. Sachs GS, Thase ME. Bipolar disorder therapeutics: maintenance treatment. Biol Psychiatry 2000;48:573-81.

5. Keck PE, Jr, Welge JA, et al. Placebo effect in randomized, controlled maintenance studies of patients with bipolar disorder. Biol Psychiatry 2000;47:756-65.

6. Calabrese JR, Rapport DJ, Shelton MD, et al. Evolving methodologies in bipolar disorder maintenance research. Br J Psychiatry 2001;178 [Suppl 41]:157-63.

7. Bowden CL, Calabrese JR, McElroy SL, et al. A randomized, placebo-controlled 12-month trial of divalproex and lithium in treatment of outpatients with bipolar I disorder. Arch Gen Psychiatry 2000;57:481-9.

8. Calabrese JR, Bowden CL, DeVeaugh-Geiss J, et al. Lamotrigine demonstrates long term mood stabilization in recently manic patients. Lamictal 606 Study Group [Abstract]. Presented at the Annual Meeting of the American Psychiatric Association Meeting, New Orleans, LA, May 5-10, 2001.

9. Denicoff KD, et al. Comparative prophylactic efficacy of lithium, carbamazepine, and the combination in bipolar disorder. J Clin Psychiatry 1997;58:470-8.

10. Greil W, et al. Lithium versus carbamazepine in the maintenance treatment of bipolar disorders—a randomized study. J Affect Disord 1997;43:151-61.

11. Hirschfeld RMA, Bowden CL, Gitlin MJ, et al. Practice guideline for the treatment of patients with bipolar disorder (revision). Am J Psychiatry, in press.

12. Tohen M, Baker R. Olanzapine versus divalproex sodium for the treatment of acute mania and maintenance of remission: a 47-week study. Presented at the Annual Meeting of the American Psychiatric Association. New Orleans, La, May 5-10, 2001.

13. Lambert PA, Venaud G. Comparative study of valpromide versus lithium as prophylactic treatment in affective disorders. Nervure J Psychiatrie 1982;4:1-9.

14. Revicki DA, Hirschfeld RMA, Keck PE, Jr, et al. Cost-effectiveness of divalproex sodium vs. lithium in long-term therapy for bipolar disorder. Presented at the Annual Meeting of the American College of Neuropsychopharmacology. San Juan, Puerto Rico, Dec. 13-17, 1999.

15. Dardennnes R, Even C, et al. Comparison of carbamazepine and lithium in the prophylaxis of bipolar disorders. A meta-analysis. Br J Psychiatry 1995;166:375-81.

16. Calabrese JR, Suppes T, et al. A double-blind, placebo-controlled, prophylaxis study of lamotrigine in rapid-cycling bipolar disorder. J Clin Psychiatry 2000;61:841-50.

17. Calabrese JR, Bowden CL, Sachs GS, et al. A double-blind placebo-controlled study of lamotrigine monotherapy in outpatients with bipolar I depression. J Clin Psychiatry 1999;60:79-88.

18. Frye MA, Ketter TA, Kimbrell TA, et al. A double-blind, placebo-controlled study of lamotrigine and gabapentin monotherapy in refractory mood disorders. J Clin Psychopharmacol 2000;24:133-40.

19. Tohen M. Olanazpine versus placebo in the treatment of acute bipolar mania. Presented at the XI World Congress of Psychiatry, World Psychiatric Association. Hamburg, Germany, Aug. 7, 1999.

20. Suppes T, Webb A, Paul B, et al. Clinical outcome in a randomized 1-year trial of clozapine versus treatment as usual for patients with treatment-resistant illness and a history of mania. Am J Psychiatry 1999;156:1164-9.

21. Keck PE, Jr, McElroy SL. Pharmacological treatment of bipolar disorder. In: Nathan PE, Gorman JM, eds. A Guide to Treatments that Work. New York: Oxford University Press, 2001, in press.

22. Keck PE, Jr, Licht R. Antipsychotic medications in the treatment of mood disorders. In: Buckley PF, Waddington JL, eds. Schizophrenia and Mood Disorders: The New Drug Therapies in Clinical Practice. Boston: Butterworth-Heiman, 2000;199-211.

23. Gelenberg AJ, Kane JM, et al. Comparison of standard and low serum levels of lithium for maintenance treatment of bipolar disorder. N Engl J Med 1989;321:1489-93.

24. Judd LL, Akiskal HS. Delineating the longitudinal structure of depressive illness: beyond clinical subtypes and duration thresholds. Pharmacopsychiatry 2000;33:3-7.

25. Goodnick PJ, Fieve RR, Schlegel A, et al. Inter-episode major and subclinical symptoms in affective disorder. Acta Psychiatr Scand 1987;75:592-600.

26. Keller MB, Lavori PW, et al. Subsyndromal symptoms in bipolar disorder: a comparison of standard and low serum lithium levels. Arch Gen Psychiatry 1992;49:371-6.

27. Post RM, Altshuler LL, Frye MA, et al. Rate of switch in bipolar patients prospectively treated with second-generation antidepressants as augmentation to mood stabilizers. Bipolar Disorders 2001;3:259-65.

28. Solomon DA, Ryan CE, Keitner GI, et al. A pilot study of lithium carbonate plus divalproex sodium for the continuation and maintenance treatment of patients with bipolar I disorder. J Clin Psychiatry 1997;58:95-9.

Article PDF
Author and Disclosure Information

Paul E. Keck, Jr, MD
Erik B. Nelson, MD
Susan L. McElroy, MD
Biological Psychiatry Program, Department of Psychiatry University of Cincinnati College of Medicine, Cincinnati, Ohio

Issue
Current Psychiatry - 01(04)
Publications
Topics
Page Number
40-47
Sections
Author and Disclosure Information

Paul E. Keck, Jr, MD
Erik B. Nelson, MD
Susan L. McElroy, MD
Biological Psychiatry Program, Department of Psychiatry University of Cincinnati College of Medicine, Cincinnati, Ohio

Author and Disclosure Information

Paul E. Keck, Jr, MD
Erik B. Nelson, MD
Susan L. McElroy, MD
Biological Psychiatry Program, Department of Psychiatry University of Cincinnati College of Medicine, Cincinnati, Ohio

Article PDF
Article PDF

What are we trying to accomplish in the maintenance treatment of patients with bipolar disorder? Given that the disorder recurs in more than 90% of patients who experience a manic episode,1 there are 5 important goals:

  1. Prevention of recurrent episodes;
  2. Amelioration of subsyndromal symptoms;
  3. Reduction of suicide risk;
  4. Compliance enhancement;
  5. Optimization of interpersonal, social, and vocational functioning.

There is a great premium on preventing mood episode recurrence in patients with bipolar disorder. Mood episodes themselves produce substantial morbidity, but morbidity is not confined to these episodes alone. Full recovery of functioning often lags many months behind remission of symptoms.2 Recurrent mood episodes also may lead to progressive loss of function between episodes. Mood episodes also carry risks of mortality from suicide, violence, and impulsive risk taking.

Clearly, mood-stabilizing medications form the cornerstone of maintenance treatment,3,4 along with a strong therapeutic alliance between patient and clinician and targeted psychosocial therapies. In the Expert Consensus Guidelines for medication treatment of bipolar I disorder, maintenance treatment was recommended for 1 year following an initial manic or mixed episode; longer (indefinite) treatment was recommended for patients with a family history of bipolar disorder or if 2 episodes occurred.3

Compared with clinical trials of agents for acute bipolar mania (and mixed episodes), there are relatively few randomized controlled trials of medications for the maintenance phase of bipolar disorder. Some naturalistic studies have provided data on relapse rates associated with treatment with a variety of different agents. Even fewer studies have examined psychosocial interventions designed specifically to reduce relapse rates.

But new data are beginning to emerge regarding the efficacy of divalproex, lamotrigine, and olanzapine as maintenance therapies. A number of clinical predictors of response to these agents have begun to be identified, as well as to lithium and carbamazepine. Eliciting these characteristics is important when making recommendations to patients about available drug therapies.

In addition, effective maintenance treatment often requires combinations of mood-stabilizing, antidepressant, and antipsychotic agents to control or eliminate subsyndromal and breakthrough symptoms.

In this review, we will cover the available new data on pharmacologic maintenance treatments of bipolar disorder and their clinical implications.

What the studies show

Lithium has been the mainstay of therapy for bipolar disorder for more than 35 years. Most randomized, controlled trials of lithium maintenance therapy were conducted in the 1960s and 1970s5 (Table 1). Unfortunately, these studies had several design limitations that inflated the expectations of lithium’s efficacy as a maintenance treatment.6 These included discontinuation designs in which patients stabilized on lithium were abruptly switched to placebo; exaggerated early placebo relapse rates; enrollment of both unipolar and bipolar patients; lack of specific diagnostic criteria; and reported results only for patients completing studies. Pooled data from these trials indicated that lithium reduced the risk of relapse fourfold compared with placebo at 6 months and 1 year.5

Two contemporary randomized, placebo-controlled maintenance studies utilizing more rigorous designs provided further evidence of lithium’s superiority over placebo in extending time to manic relapse.7,8 Both studies enrolled patients who were currently or had recently been manic and had been stabilized in open-label treatment that included study medications.

In the first study, comparing 1-year relapse rates among patients randomized to lithium, divalproex, or placebo, lithium extended time to recurrence of mania by 55% compared with placebo.7 In the second, an 18-month trial comparing lithium, lamotrigine, and placebo, lithium significantly increased the time to intervention for recurrence of mania compared with placebo.8 The overall manic relapse rates were 17% for lithium-treated patients and 41% for those on placebo. However, lithium did not significantly extend time to depressive relapse or intervention for depressive relapse, respectively, in either study. Moreover, in the first study, patients who received lithium tended to have greater subthreshold depressive symptoms.8

Table 1

What works in maintenance treatment of bipolar disorder (ranked by number of randomized, controlled trials)

Medication RCTs (n)Trial resultsStrong evidenceSome evidence
Lithium (15)Equal to divalproex and lamotrigine; equal to or better than carbamazepine; better than placebo4-fold reduction of relapse risk vs. placebo at 6, 12 months; more efficacious in mania than in depression; higher lithium level correlated with lower relapse rates, but more side effects 
Carbamazepine (7)No significant benefit vs. lithium; better than placebo May be more efficacious in mania than in depression; may be less tolerable than lithium; no data on serum levels
Divalproex (4)Equal to lithium and olanzapine; better than placebo May be more efficacious in mania than in depression; may be more tolerable than lithium; data on serum levels pending
Lamotrigine (2)Better than placeboMore efficacious in depression than in maniaDosage 200-400 mg/d
Olanzapine (1)Equal to divalproex Data on dosage and differential efficacy related to dosage pending
Clozapine (1)Better than treatment as usualEfficacy in treatment-resistant mania (bipolar schizoaffective disorder, bipolar type) 
 

 

These findings were consistent with earlier placebo-controlled studies of lithium maintenance treatment and a recent crossover comparison trial with carbamazepine.9 Lithium was also recently compared with carbamazepine in a 2.5-year maintenance multisite study in Europe.10 There was no significant difference in efficacy between the two in time to hospitalization, the primary outcome measure. On other outcome measures, including time to relapse or need for additional medication, lithium was superior to carbamazepine.

Pooled results from a number of naturalistic studies, which mirror clinical practice, indicated that approximately one-third of patients maintained on lithium had good functional outcomes without relapses and only minimal symptoms.11 In general, these studies found higher rates of relapse with longer durations of follow-up.

Valproate maintenance treatment has been studied in two randomized, controlled trials (one vs. placebo,7 and one vs. olanzapine12) and two open-label comparison studies against lithium.13,14 In the placebo-controlled trial, which also included a lithium comparison group described earlier, there was no significant difference in the time to development of any mood episode among the three treatment groups.7 However, divalproex was superior to placebo on a number of other outcome measures, including rate of study termination for any mood episode, termination for depression, and termination for noncompliance.

Divalproex was superior to placebo in patients who received divalproex in the open-label treatment phase before randomization. This is clinically relevant, as this group reflects expected relapse rates in patients treated initially with divalproex for acute mania who then remain on the drug for maintenance therapy. Patients receiving divalproex had significantly lower rates of intolerance and noncompliance compared to those treated with lithium.

In the second comparison study, 167 patients initially randomized and responding to divalproex or olanzapine in a 3-week acute bipolar mania trial continued in a double-blind 44-week extension study.12 There were no significant differences in relapse into mania between the two groups (olanzapine 41%, divalproex 50%, p = 0.4) or time to manic relapse (olanzapine 270 days, divalproex 74 days, p = 0.4). There was no significant difference in tolerability between the two.

Two open-label studies compared valproate with lithium. In an 18-month study conducted in France, patients randomized to the valpromide formulation of valproate displayed a 20% lower relapse rate than those receiving lithium.13 The second open-label comparison trial found comparable efficacy between lithium and divalproex in a 1-year naturalistic pharmacoeconomic study that allowed additional medications as needed for recurrent symptoms.14

Carbamazepine has been evaluated in a limited number of studies for maintenance treatment of bipolar disorder. The results of many early randomized, controlled maintenance trials were criticized on methodologic grounds.15 But two recent comparison trials with lithium, as noted earlier, provided clinically important data regarding carbamazepine’s prophylactic efficacy.9,10 Several additional clinically relevant observations emerged from analyses of secondary outcome measures in these two trials.

In the first study, 52 patients with bipolar I or II disorder received lithium or carbamazepine for 1 year, crossed over to the alternate drug the second year, and received both drugs the third year.9 There was little difference in relapse rates during the first year between the lithium (31%) and carbamazepine (37%) groups. Similarly, in the overall trial, there was little difference in the percentage of patients who were rated as having a moderate or better response—33% on lithium, 31% on carbamazepine, and 55% on the combination. A higher proportion of patients receiving carbamazepine withdrew due to side effects. In the second trial, significantly more patients receiving carbamazepine required additional medications for breakthrough symptoms and experienced side effects requiring treatment discontinuation.10

Lamotrigine has been studied in two placebo-controlled, randomized maintenance studies in patients with bipolar disorder.8,16 The first evaluated bipolar I patients who had experienced a manic or hypomanic episode within 60 days of entry into an open-label treatment phase with lamotrigine.8 Patients who, in turn, improved or remained stable during the open-label treatment phase were then randomized to treatment with lamotrigine 200-400 mg/d, lithium, or placebo for up to 18 months. Both lamotrigine and lithium were superior to placebo on the primary outcome measure, which was time to need for additional medication for a mood episode. The median time until 25% of patients relapsed was 72 weeks for lamotrigine, 58 weeks for lithium, and 35 weeks for placebo.

Table 2

PUTATIVE PREDICTORS OF RESPONSE TO MAINTENANCE MEDICATIONS

MedicationPredictor of responseStrength of evidence
LithiumNonrap id cycling
Few episodes
Few depressive symptoms
Family history bipolar disorder
Episode sequence M-D-I
No substance/alcohol use disorder
◊◊◊




DivalproexEqual efficacy in rapid & non-rapid cycling, manic & mixed,
No personality disorder

CarbamazepineEqual efficacy in rapid & non-rapid cycling, manic & mixed
Mood-incongruent symptoms
◊◊
LamotrigineBipolar II > bipolar I
Depression > mania

◊◊
OlanzapineEqual efficacy in rapid & non-rapid cycling, manic & mixed, psychotic & nonpsychotic mania in acute studies; data pending in maintenance
ClozapineEfficacy in treatment-resistant mania rapid & nonrapid cycling, manic & mixed, psychotic & nonpsychotic in naturalistic studies◊◊◊
Key ◊ = reported in 1 study; ◊◊ = reported in 2 studies; ◊◊◊ = reported in > 3 studies.
 

 

On secondary outcome measures, lamotrigine, but not lithium, was superior to placebo in delaying time to depressive relapse. In contrast, lithium, but not lamotrigine, was superior to placebo in delaying time to manic relapse. Finally, lamotrigine, but not lithium, was superior to placebo in time to discontinuation for any reason.

From this study, it appears that lamotrigine is most effective in preventing depressive relapse, whereas lithium is most effective in preventing manic relapse. Thus, lithium and lamotrigine may complement each other in maintenance treatment by preventing manic and depressive episodes in combination.

The findings of the first trial were consistent with those of the second placebo-controlled lamotrigine (100-500 mg/d) maintenance study conducted specifically in patients with rapid cycling bipolar I and II disorders.16 In this 6-month trial, there was no significant difference in time to need for additional medications, the primary outcome measure, between the lamotrigine and placebo groups.

Median survival time was significantly greater for the lamotrigine group (18 weeks) than for the placebo group (12 weeks). The lamotrigine group had significantly longer time to need for additional medications in bipolar II, but not bipolar I, patients. Patients with bipolar II disorder also displayed significantly greater improvement with lamotrigine on global scales compared with bipolar I patients. Because bipolar II disorder is characterized predominantly by depressive episodes, these findings suggest that lamotrigine was again more beneficial in preventing recurrent depression, in this case, specifically, in rapid cycling bipolar II patients.

Taken together, the results of these studies are also consistent with the results of two placebo-controlled trials of lamotrigine in the treatment of acute bipolar depression.17,18 Its efficacy in acute bipolar I depression was first demonstrated in a 6-week, double-blind, randomized, parallel-group trial in which patients received lamotrigine 50 mg/d, 200 mg/d (after gradual titration over the first 4 weeks), and placebo.17

Both dosage groups of lamotrigine displayed significantly greater improvement in depressive symptoms as measured by the Montgomery-Asberg Depression Rating Scale (but not the Hamilton Depression Rating Scale) and by the Clinical Global Improvement Scale compared with the placebo group. There was a trend for the 200 mg/d group to have greater improvement than the 50 mg/d group, a trend that might have become significant had the study been longer, as the 200 mg/d group did not receive this dose for the full trial. There was no significant difference in the incidence of hypomanic or manic switches among the three study groups.

Frye and colleagues also found lamotrigine to be superior to placebo in global improvement in depressive (but not manic) symptoms in a crossover monotherapy trial in treatment-refractory bipolar I and II patients.18

Olanzapine, as described earlier, was comparable to divalproex in a head-to-head comparison 44-week extension trial in patients who responded to double-blind treatment in an acute mania study.12 These results were consistent with the preliminary findings of an open-label extension study of olanzapine.19 There are no placebo-controlled trials of olanzapine or any other atypical antipsychotic in the maintenance phase of bipolar disorder published to date.

Clozapine was more effective than “treatment as usual” (combinations of mood-stabilizers and typical antipsychotics) in patients with treatment-refractory bipolar and schizoaffective (bipolar subtype) disorders.20 Many patients with bipolar disorder now receive adjunctive maintenance treatment with typical antipsychotics, but the limited data from the few controlled trials of typical agents administered in combination with mood-stabilizers do not support the efficacy of this strategy.21 Moreover, some reports suggest that maintenance treatment incorporating typical antipsychotics may increase the frequency and severity of depressive episodes in patients with bipolar disorder.

Predicting response to pharmacologic treatment

Clinical experience and data from the randomized, controlled trials reviewed earlier have identified several tentative predictors of response—or nonresponse—to specific medications. Lithium, divalproex, and carbamazepine appear to have greater efficacy in the prevention of manic rather than depressive episodes.11 In contrast, lamotrigine appears to have greater efficacy in preventing depressive rather than manic episodes.

It is unclear whether olanzapine and perhaps other atypical antipsychotics may have a more favorable effect in preventing one pole of the illness over another (Table 2). The currently available atypicals (clozapine, risperidone, olanzapine, quetiapine, and ziprasidone) in general appear to differ from typical antipsychotics in having bidirectional (antimanic and antidepressant) effects on mood symptoms.

Atypicals may exert antidepressant effects via a number of different mechanisms, including 5HT2 receptor antagonism (a property shared by all atypicals and the antidepressants trazodone, nefazodone, and mirtazapine); alpha2-adrenergic antagonism (clozapine and risperidone); and 5HT1D antagonism, 5HT1A agonism, and 5HT and NE reuptake inhibition (ziprasidone).22

Patients with rapid-cycling bipolar disorder have a relatively poor response to lithium (response rates of approximately 20%).11 Patients with rapid-cycling bipolar I disorder may have a greater likelihood of response to combined treatment with lithium and carbamazepine,9 or divalproex.11 Alternately, patients with rapid-cycling bipolar II disorder have lower relapse rates on lamotrigine compared with placebo.16 Anecdotal reports and the results of acute treatment studies suggest that clozapine and olanzapine may be beneficial maintenance agents for rapid-cycling bipolar I patients.

 

 

Other predictors of favorable response to lithium prophylaxis include a family history of bipolar disorder, illness course characterized by maniadepression-euthymia episode sequence, and few prior mood episodes.21 Conversely, co-occurring alcohol or substance use disorder, multiple prior mood episodes, and familial negative affective style have been associated with poor response to lithium maintenance treatment.

Tentative predictors of response to divalproex maintenance treatment include mixed episodes, rapid cycling, and absence of co-occurring personality disorder. In one lithium comparison trial, patients with mixed episodes, bipolar II, and NOS disorders and mood-incongruent symptoms appeared to have a better response to carbamazepine.10

The dosage and relevant plasma concentrations of maintenance agents may also affect long-term efficacy. Gelenberg et al observed that the risk of relapse in bipolar patients maintained on low (0.4-0.6 mmol/L) serum concentrations was 2.6 times higher than for patients maintained on high (0.8-1.0 mmol/L) serum concentrations.23 However, there was a tradeoff in tolerability. Patients treated at the higher concentrations experienced significantly more and often treatment-limiting side effects.

No data are available regarding a possible plasma concentration-response relationship between maintenance treatment with divalproex or carbamazepine, but based on data from acute treatment trials, concentrations well within the therapeutic range of each drug appear essential.

The problem of subsyndromal symptoms

Evidence from a number of long-term outcome studies in bipolar disorder indicates that many patients spend protracted periods of time neither well nor in syndromal manic or depressive episodes, but rather experiencing chronic subsyndromal symptoms.24,25 In these studies, depressive symptoms were twice as prevalent as hypomanic symptoms between acute episodes of illness. Furthermore, persistent subsyndromal depressive symptoms were strongly associated with an increased risk for relapse and poor occupational functioning.

Keller et al26 found that patients maintained on low lithium serum concentrations (0.4-0.6 mmol/L) were more likely to experience subsyndromal symptoms and that their symptoms were more likely to worsen at any time than were symptoms of patients maintained at higher serum concentrations (0.8-10 mmol/L). The first occurrence of subsyndromal symptoms increased the risk of fullepisode relapse fourfold. These findings indicate that the optimal pharmacological maintenance treatment of bipolar disorder requires titration of mood-stabilizer medications to eradicate subsyndromal symptoms. Eliminating these residual or recurrent symptoms, in turn, substantially decreases the risk of relapse and of enduring functional impairment.

Conventional wisdom, in part supported by limited data from a small number of studies, suggests that antidepressants be used sparingly and for limited periods of time in conjunction with mood-stabilizers for bipolar depression. However, new data indicate that this strategy may significantly increase the risk of recurrence of depressive symptoms and episodes.27 Thus, combined treatment with mood-stabilizing agents and antidepressants for patients without rapid cycling and with recurrent depressive episodes may be indicated more often than previously thought.

In practice, perhaps the majority of patients with bipolar disorder require treatment with more than one mood-stabilizing medication to suppress subsyndromal symptoms as well as to prevent full episode recurrence. The use of combinations has been very poorly studied in randomized, controlled trials. In a pilot study of 12 patients with bipolar I disorder, Solomon et al compared the efficacy of lithium alone versus the combination of lithium and divalproex for 1 year.28 Not surprisingly, the combination significantly reduced the risk of recurrence of mania and depression but was also associated with more bothersome side effects.

Clinical practice has greatly outstripped the limited data available from formal studies. Recently reported as useful maintenance treatment strategies are combinations of:

  • lithium and divalproex
  • lithium and carbamazepine
  • divalproex and carbamazepine
  • lithium, divalproex, and carbamazepine
  • lithium and/or divalproex with atypical antipsychotics, antidepressants, and lamotrigine.

Related resources

  • Nathan PE, Gorman JM, eds. A Guide to Treatments that Work. 2nd ed. New York: Oxford University Press, 2001.
  • Buckley PF, Waddington JL, eds. Schizophrenia and Mood Disorders: the New Drug Therapies in Clinical Practice. Boston: Butterworth-Heiman, 2000.
  • Goldberg JF, Harrow M. Bipolar Disorders. Clinical Course and Outcome. Washington, DC: American Psychiatric Press, 1999.
  • Sachs GS, Thase ME. Bipolar disorder therapeutics: maintenance treatment. Biol Psychiatry 2000;48:573-81.
  • Hirschfeld RMA, Bowden CL, Gitlin MJ, et al. Practice guideline for the treatment of bipolar disorder (revision). Am J Psychiatry, in press.

Drug brand names

  • Clozapine • Clozaril
  • Divalproex • Depakote, Depakote Sprinkle
  • Lamotrigine • Lamictal
  • Mirtazapine • Remeron, Remeron Soltab
  • Nefazodone • Serzone
  • Olanzapine • Zyprexa
  • Quetiapine • Seroquel
  • Risperidone • Risperdal
  • Trazodone • Desyrel
  • Valproate sodium • Depacon
  • Ziprasidone • Geodon

Disclosure

Dr. Keck reports that he receives grant/research support from and serves as a consultant to Abbott Laboratories, AstraZeneca, Pfizer Inc., and Eli Lilly and Co. He also receives grant/research support from Merck and Co. and Otsuka America Pharmaceutical, and serves as a consultant to Bristol-Myers Squibb Co., GlaxoSmithKline, and Janssen Pharmaceutica.

 

 

Dr. McElroy reports that she receives grant/research support from and serves as a consultant to Abbott Laboratories, Elan Pharmaceuticals, Cephalon Inc. GlaxoSmithKline, and Eli Lilly and Co. She also receives grant/research support from Forest Pharmaceuticals and Solvay Pharmaceuticals, and serves as a consultant to Bristol-Myers Squibb Co., Ortho-McNeil Pharmaceutical, and Janssen Pharmaceutica.

Dr. Nelson reports no financial relationship with any company whose products are mentioned in this article.

What are we trying to accomplish in the maintenance treatment of patients with bipolar disorder? Given that the disorder recurs in more than 90% of patients who experience a manic episode,1 there are 5 important goals:

  1. Prevention of recurrent episodes;
  2. Amelioration of subsyndromal symptoms;
  3. Reduction of suicide risk;
  4. Compliance enhancement;
  5. Optimization of interpersonal, social, and vocational functioning.

There is a great premium on preventing mood episode recurrence in patients with bipolar disorder. Mood episodes themselves produce substantial morbidity, but morbidity is not confined to these episodes alone. Full recovery of functioning often lags many months behind remission of symptoms.2 Recurrent mood episodes also may lead to progressive loss of function between episodes. Mood episodes also carry risks of mortality from suicide, violence, and impulsive risk taking.

Clearly, mood-stabilizing medications form the cornerstone of maintenance treatment,3,4 along with a strong therapeutic alliance between patient and clinician and targeted psychosocial therapies. In the Expert Consensus Guidelines for medication treatment of bipolar I disorder, maintenance treatment was recommended for 1 year following an initial manic or mixed episode; longer (indefinite) treatment was recommended for patients with a family history of bipolar disorder or if 2 episodes occurred.3

Compared with clinical trials of agents for acute bipolar mania (and mixed episodes), there are relatively few randomized controlled trials of medications for the maintenance phase of bipolar disorder. Some naturalistic studies have provided data on relapse rates associated with treatment with a variety of different agents. Even fewer studies have examined psychosocial interventions designed specifically to reduce relapse rates.

But new data are beginning to emerge regarding the efficacy of divalproex, lamotrigine, and olanzapine as maintenance therapies. A number of clinical predictors of response to these agents have begun to be identified, as well as to lithium and carbamazepine. Eliciting these characteristics is important when making recommendations to patients about available drug therapies.

In addition, effective maintenance treatment often requires combinations of mood-stabilizing, antidepressant, and antipsychotic agents to control or eliminate subsyndromal and breakthrough symptoms.

In this review, we will cover the available new data on pharmacologic maintenance treatments of bipolar disorder and their clinical implications.

What the studies show

Lithium has been the mainstay of therapy for bipolar disorder for more than 35 years. Most randomized, controlled trials of lithium maintenance therapy were conducted in the 1960s and 1970s5 (Table 1). Unfortunately, these studies had several design limitations that inflated the expectations of lithium’s efficacy as a maintenance treatment.6 These included discontinuation designs in which patients stabilized on lithium were abruptly switched to placebo; exaggerated early placebo relapse rates; enrollment of both unipolar and bipolar patients; lack of specific diagnostic criteria; and reported results only for patients completing studies. Pooled data from these trials indicated that lithium reduced the risk of relapse fourfold compared with placebo at 6 months and 1 year.5

Two contemporary randomized, placebo-controlled maintenance studies utilizing more rigorous designs provided further evidence of lithium’s superiority over placebo in extending time to manic relapse.7,8 Both studies enrolled patients who were currently or had recently been manic and had been stabilized in open-label treatment that included study medications.

In the first study, comparing 1-year relapse rates among patients randomized to lithium, divalproex, or placebo, lithium extended time to recurrence of mania by 55% compared with placebo.7 In the second, an 18-month trial comparing lithium, lamotrigine, and placebo, lithium significantly increased the time to intervention for recurrence of mania compared with placebo.8 The overall manic relapse rates were 17% for lithium-treated patients and 41% for those on placebo. However, lithium did not significantly extend time to depressive relapse or intervention for depressive relapse, respectively, in either study. Moreover, in the first study, patients who received lithium tended to have greater subthreshold depressive symptoms.8

Table 1

What works in maintenance treatment of bipolar disorder (ranked by number of randomized, controlled trials)

Medication RCTs (n)Trial resultsStrong evidenceSome evidence
Lithium (15)Equal to divalproex and lamotrigine; equal to or better than carbamazepine; better than placebo4-fold reduction of relapse risk vs. placebo at 6, 12 months; more efficacious in mania than in depression; higher lithium level correlated with lower relapse rates, but more side effects 
Carbamazepine (7)No significant benefit vs. lithium; better than placebo May be more efficacious in mania than in depression; may be less tolerable than lithium; no data on serum levels
Divalproex (4)Equal to lithium and olanzapine; better than placebo May be more efficacious in mania than in depression; may be more tolerable than lithium; data on serum levels pending
Lamotrigine (2)Better than placeboMore efficacious in depression than in maniaDosage 200-400 mg/d
Olanzapine (1)Equal to divalproex Data on dosage and differential efficacy related to dosage pending
Clozapine (1)Better than treatment as usualEfficacy in treatment-resistant mania (bipolar schizoaffective disorder, bipolar type) 
 

 

These findings were consistent with earlier placebo-controlled studies of lithium maintenance treatment and a recent crossover comparison trial with carbamazepine.9 Lithium was also recently compared with carbamazepine in a 2.5-year maintenance multisite study in Europe.10 There was no significant difference in efficacy between the two in time to hospitalization, the primary outcome measure. On other outcome measures, including time to relapse or need for additional medication, lithium was superior to carbamazepine.

Pooled results from a number of naturalistic studies, which mirror clinical practice, indicated that approximately one-third of patients maintained on lithium had good functional outcomes without relapses and only minimal symptoms.11 In general, these studies found higher rates of relapse with longer durations of follow-up.

Valproate maintenance treatment has been studied in two randomized, controlled trials (one vs. placebo,7 and one vs. olanzapine12) and two open-label comparison studies against lithium.13,14 In the placebo-controlled trial, which also included a lithium comparison group described earlier, there was no significant difference in the time to development of any mood episode among the three treatment groups.7 However, divalproex was superior to placebo on a number of other outcome measures, including rate of study termination for any mood episode, termination for depression, and termination for noncompliance.

Divalproex was superior to placebo in patients who received divalproex in the open-label treatment phase before randomization. This is clinically relevant, as this group reflects expected relapse rates in patients treated initially with divalproex for acute mania who then remain on the drug for maintenance therapy. Patients receiving divalproex had significantly lower rates of intolerance and noncompliance compared to those treated with lithium.

In the second comparison study, 167 patients initially randomized and responding to divalproex or olanzapine in a 3-week acute bipolar mania trial continued in a double-blind 44-week extension study.12 There were no significant differences in relapse into mania between the two groups (olanzapine 41%, divalproex 50%, p = 0.4) or time to manic relapse (olanzapine 270 days, divalproex 74 days, p = 0.4). There was no significant difference in tolerability between the two.

Two open-label studies compared valproate with lithium. In an 18-month study conducted in France, patients randomized to the valpromide formulation of valproate displayed a 20% lower relapse rate than those receiving lithium.13 The second open-label comparison trial found comparable efficacy between lithium and divalproex in a 1-year naturalistic pharmacoeconomic study that allowed additional medications as needed for recurrent symptoms.14

Carbamazepine has been evaluated in a limited number of studies for maintenance treatment of bipolar disorder. The results of many early randomized, controlled maintenance trials were criticized on methodologic grounds.15 But two recent comparison trials with lithium, as noted earlier, provided clinically important data regarding carbamazepine’s prophylactic efficacy.9,10 Several additional clinically relevant observations emerged from analyses of secondary outcome measures in these two trials.

In the first study, 52 patients with bipolar I or II disorder received lithium or carbamazepine for 1 year, crossed over to the alternate drug the second year, and received both drugs the third year.9 There was little difference in relapse rates during the first year between the lithium (31%) and carbamazepine (37%) groups. Similarly, in the overall trial, there was little difference in the percentage of patients who were rated as having a moderate or better response—33% on lithium, 31% on carbamazepine, and 55% on the combination. A higher proportion of patients receiving carbamazepine withdrew due to side effects. In the second trial, significantly more patients receiving carbamazepine required additional medications for breakthrough symptoms and experienced side effects requiring treatment discontinuation.10

Lamotrigine has been studied in two placebo-controlled, randomized maintenance studies in patients with bipolar disorder.8,16 The first evaluated bipolar I patients who had experienced a manic or hypomanic episode within 60 days of entry into an open-label treatment phase with lamotrigine.8 Patients who, in turn, improved or remained stable during the open-label treatment phase were then randomized to treatment with lamotrigine 200-400 mg/d, lithium, or placebo for up to 18 months. Both lamotrigine and lithium were superior to placebo on the primary outcome measure, which was time to need for additional medication for a mood episode. The median time until 25% of patients relapsed was 72 weeks for lamotrigine, 58 weeks for lithium, and 35 weeks for placebo.

Table 2

PUTATIVE PREDICTORS OF RESPONSE TO MAINTENANCE MEDICATIONS

MedicationPredictor of responseStrength of evidence
LithiumNonrap id cycling
Few episodes
Few depressive symptoms
Family history bipolar disorder
Episode sequence M-D-I
No substance/alcohol use disorder
◊◊◊




DivalproexEqual efficacy in rapid & non-rapid cycling, manic & mixed,
No personality disorder

CarbamazepineEqual efficacy in rapid & non-rapid cycling, manic & mixed
Mood-incongruent symptoms
◊◊
LamotrigineBipolar II > bipolar I
Depression > mania

◊◊
OlanzapineEqual efficacy in rapid & non-rapid cycling, manic & mixed, psychotic & nonpsychotic mania in acute studies; data pending in maintenance
ClozapineEfficacy in treatment-resistant mania rapid & nonrapid cycling, manic & mixed, psychotic & nonpsychotic in naturalistic studies◊◊◊
Key ◊ = reported in 1 study; ◊◊ = reported in 2 studies; ◊◊◊ = reported in > 3 studies.
 

 

On secondary outcome measures, lamotrigine, but not lithium, was superior to placebo in delaying time to depressive relapse. In contrast, lithium, but not lamotrigine, was superior to placebo in delaying time to manic relapse. Finally, lamotrigine, but not lithium, was superior to placebo in time to discontinuation for any reason.

From this study, it appears that lamotrigine is most effective in preventing depressive relapse, whereas lithium is most effective in preventing manic relapse. Thus, lithium and lamotrigine may complement each other in maintenance treatment by preventing manic and depressive episodes in combination.

The findings of the first trial were consistent with those of the second placebo-controlled lamotrigine (100-500 mg/d) maintenance study conducted specifically in patients with rapid cycling bipolar I and II disorders.16 In this 6-month trial, there was no significant difference in time to need for additional medications, the primary outcome measure, between the lamotrigine and placebo groups.

Median survival time was significantly greater for the lamotrigine group (18 weeks) than for the placebo group (12 weeks). The lamotrigine group had significantly longer time to need for additional medications in bipolar II, but not bipolar I, patients. Patients with bipolar II disorder also displayed significantly greater improvement with lamotrigine on global scales compared with bipolar I patients. Because bipolar II disorder is characterized predominantly by depressive episodes, these findings suggest that lamotrigine was again more beneficial in preventing recurrent depression, in this case, specifically, in rapid cycling bipolar II patients.

Taken together, the results of these studies are also consistent with the results of two placebo-controlled trials of lamotrigine in the treatment of acute bipolar depression.17,18 Its efficacy in acute bipolar I depression was first demonstrated in a 6-week, double-blind, randomized, parallel-group trial in which patients received lamotrigine 50 mg/d, 200 mg/d (after gradual titration over the first 4 weeks), and placebo.17

Both dosage groups of lamotrigine displayed significantly greater improvement in depressive symptoms as measured by the Montgomery-Asberg Depression Rating Scale (but not the Hamilton Depression Rating Scale) and by the Clinical Global Improvement Scale compared with the placebo group. There was a trend for the 200 mg/d group to have greater improvement than the 50 mg/d group, a trend that might have become significant had the study been longer, as the 200 mg/d group did not receive this dose for the full trial. There was no significant difference in the incidence of hypomanic or manic switches among the three study groups.

Frye and colleagues also found lamotrigine to be superior to placebo in global improvement in depressive (but not manic) symptoms in a crossover monotherapy trial in treatment-refractory bipolar I and II patients.18

Olanzapine, as described earlier, was comparable to divalproex in a head-to-head comparison 44-week extension trial in patients who responded to double-blind treatment in an acute mania study.12 These results were consistent with the preliminary findings of an open-label extension study of olanzapine.19 There are no placebo-controlled trials of olanzapine or any other atypical antipsychotic in the maintenance phase of bipolar disorder published to date.

Clozapine was more effective than “treatment as usual” (combinations of mood-stabilizers and typical antipsychotics) in patients with treatment-refractory bipolar and schizoaffective (bipolar subtype) disorders.20 Many patients with bipolar disorder now receive adjunctive maintenance treatment with typical antipsychotics, but the limited data from the few controlled trials of typical agents administered in combination with mood-stabilizers do not support the efficacy of this strategy.21 Moreover, some reports suggest that maintenance treatment incorporating typical antipsychotics may increase the frequency and severity of depressive episodes in patients with bipolar disorder.

Predicting response to pharmacologic treatment

Clinical experience and data from the randomized, controlled trials reviewed earlier have identified several tentative predictors of response—or nonresponse—to specific medications. Lithium, divalproex, and carbamazepine appear to have greater efficacy in the prevention of manic rather than depressive episodes.11 In contrast, lamotrigine appears to have greater efficacy in preventing depressive rather than manic episodes.

It is unclear whether olanzapine and perhaps other atypical antipsychotics may have a more favorable effect in preventing one pole of the illness over another (Table 2). The currently available atypicals (clozapine, risperidone, olanzapine, quetiapine, and ziprasidone) in general appear to differ from typical antipsychotics in having bidirectional (antimanic and antidepressant) effects on mood symptoms.

Atypicals may exert antidepressant effects via a number of different mechanisms, including 5HT2 receptor antagonism (a property shared by all atypicals and the antidepressants trazodone, nefazodone, and mirtazapine); alpha2-adrenergic antagonism (clozapine and risperidone); and 5HT1D antagonism, 5HT1A agonism, and 5HT and NE reuptake inhibition (ziprasidone).22

Patients with rapid-cycling bipolar disorder have a relatively poor response to lithium (response rates of approximately 20%).11 Patients with rapid-cycling bipolar I disorder may have a greater likelihood of response to combined treatment with lithium and carbamazepine,9 or divalproex.11 Alternately, patients with rapid-cycling bipolar II disorder have lower relapse rates on lamotrigine compared with placebo.16 Anecdotal reports and the results of acute treatment studies suggest that clozapine and olanzapine may be beneficial maintenance agents for rapid-cycling bipolar I patients.

 

 

Other predictors of favorable response to lithium prophylaxis include a family history of bipolar disorder, illness course characterized by maniadepression-euthymia episode sequence, and few prior mood episodes.21 Conversely, co-occurring alcohol or substance use disorder, multiple prior mood episodes, and familial negative affective style have been associated with poor response to lithium maintenance treatment.

Tentative predictors of response to divalproex maintenance treatment include mixed episodes, rapid cycling, and absence of co-occurring personality disorder. In one lithium comparison trial, patients with mixed episodes, bipolar II, and NOS disorders and mood-incongruent symptoms appeared to have a better response to carbamazepine.10

The dosage and relevant plasma concentrations of maintenance agents may also affect long-term efficacy. Gelenberg et al observed that the risk of relapse in bipolar patients maintained on low (0.4-0.6 mmol/L) serum concentrations was 2.6 times higher than for patients maintained on high (0.8-1.0 mmol/L) serum concentrations.23 However, there was a tradeoff in tolerability. Patients treated at the higher concentrations experienced significantly more and often treatment-limiting side effects.

No data are available regarding a possible plasma concentration-response relationship between maintenance treatment with divalproex or carbamazepine, but based on data from acute treatment trials, concentrations well within the therapeutic range of each drug appear essential.

The problem of subsyndromal symptoms

Evidence from a number of long-term outcome studies in bipolar disorder indicates that many patients spend protracted periods of time neither well nor in syndromal manic or depressive episodes, but rather experiencing chronic subsyndromal symptoms.24,25 In these studies, depressive symptoms were twice as prevalent as hypomanic symptoms between acute episodes of illness. Furthermore, persistent subsyndromal depressive symptoms were strongly associated with an increased risk for relapse and poor occupational functioning.

Keller et al26 found that patients maintained on low lithium serum concentrations (0.4-0.6 mmol/L) were more likely to experience subsyndromal symptoms and that their symptoms were more likely to worsen at any time than were symptoms of patients maintained at higher serum concentrations (0.8-10 mmol/L). The first occurrence of subsyndromal symptoms increased the risk of fullepisode relapse fourfold. These findings indicate that the optimal pharmacological maintenance treatment of bipolar disorder requires titration of mood-stabilizer medications to eradicate subsyndromal symptoms. Eliminating these residual or recurrent symptoms, in turn, substantially decreases the risk of relapse and of enduring functional impairment.

Conventional wisdom, in part supported by limited data from a small number of studies, suggests that antidepressants be used sparingly and for limited periods of time in conjunction with mood-stabilizers for bipolar depression. However, new data indicate that this strategy may significantly increase the risk of recurrence of depressive symptoms and episodes.27 Thus, combined treatment with mood-stabilizing agents and antidepressants for patients without rapid cycling and with recurrent depressive episodes may be indicated more often than previously thought.

In practice, perhaps the majority of patients with bipolar disorder require treatment with more than one mood-stabilizing medication to suppress subsyndromal symptoms as well as to prevent full episode recurrence. The use of combinations has been very poorly studied in randomized, controlled trials. In a pilot study of 12 patients with bipolar I disorder, Solomon et al compared the efficacy of lithium alone versus the combination of lithium and divalproex for 1 year.28 Not surprisingly, the combination significantly reduced the risk of recurrence of mania and depression but was also associated with more bothersome side effects.

Clinical practice has greatly outstripped the limited data available from formal studies. Recently reported as useful maintenance treatment strategies are combinations of:

  • lithium and divalproex
  • lithium and carbamazepine
  • divalproex and carbamazepine
  • lithium, divalproex, and carbamazepine
  • lithium and/or divalproex with atypical antipsychotics, antidepressants, and lamotrigine.

Related resources

  • Nathan PE, Gorman JM, eds. A Guide to Treatments that Work. 2nd ed. New York: Oxford University Press, 2001.
  • Buckley PF, Waddington JL, eds. Schizophrenia and Mood Disorders: the New Drug Therapies in Clinical Practice. Boston: Butterworth-Heiman, 2000.
  • Goldberg JF, Harrow M. Bipolar Disorders. Clinical Course and Outcome. Washington, DC: American Psychiatric Press, 1999.
  • Sachs GS, Thase ME. Bipolar disorder therapeutics: maintenance treatment. Biol Psychiatry 2000;48:573-81.
  • Hirschfeld RMA, Bowden CL, Gitlin MJ, et al. Practice guideline for the treatment of bipolar disorder (revision). Am J Psychiatry, in press.

Drug brand names

  • Clozapine • Clozaril
  • Divalproex • Depakote, Depakote Sprinkle
  • Lamotrigine • Lamictal
  • Mirtazapine • Remeron, Remeron Soltab
  • Nefazodone • Serzone
  • Olanzapine • Zyprexa
  • Quetiapine • Seroquel
  • Risperidone • Risperdal
  • Trazodone • Desyrel
  • Valproate sodium • Depacon
  • Ziprasidone • Geodon

Disclosure

Dr. Keck reports that he receives grant/research support from and serves as a consultant to Abbott Laboratories, AstraZeneca, Pfizer Inc., and Eli Lilly and Co. He also receives grant/research support from Merck and Co. and Otsuka America Pharmaceutical, and serves as a consultant to Bristol-Myers Squibb Co., GlaxoSmithKline, and Janssen Pharmaceutica.

 

 

Dr. McElroy reports that she receives grant/research support from and serves as a consultant to Abbott Laboratories, Elan Pharmaceuticals, Cephalon Inc. GlaxoSmithKline, and Eli Lilly and Co. She also receives grant/research support from Forest Pharmaceuticals and Solvay Pharmaceuticals, and serves as a consultant to Bristol-Myers Squibb Co., Ortho-McNeil Pharmaceutical, and Janssen Pharmaceutica.

Dr. Nelson reports no financial relationship with any company whose products are mentioned in this article.

References

1. Solomon DA, Keitner GI, Miller IW, et al. Course of illness and maintenance treatments for patients with bipolar disorder. J Clin Psychiatry 1995;56:5-13.

2. Keck PE, Jr, McElroy SL, et al. Twelve-month outcome of bipolar patients following hospitalization for a manic or mixed episode. Am J Psychiatry 1998;155:646-52.

3. Sachs GS, Printz DJ, et al. The Expert Consensus Guideline Series: medication treatment of bipolar disorders 2000. Postgrad Med Special Report 2000;4:1-104.

4. Sachs GS, Thase ME. Bipolar disorder therapeutics: maintenance treatment. Biol Psychiatry 2000;48:573-81.

5. Keck PE, Jr, Welge JA, et al. Placebo effect in randomized, controlled maintenance studies of patients with bipolar disorder. Biol Psychiatry 2000;47:756-65.

6. Calabrese JR, Rapport DJ, Shelton MD, et al. Evolving methodologies in bipolar disorder maintenance research. Br J Psychiatry 2001;178 [Suppl 41]:157-63.

7. Bowden CL, Calabrese JR, McElroy SL, et al. A randomized, placebo-controlled 12-month trial of divalproex and lithium in treatment of outpatients with bipolar I disorder. Arch Gen Psychiatry 2000;57:481-9.

8. Calabrese JR, Bowden CL, DeVeaugh-Geiss J, et al. Lamotrigine demonstrates long term mood stabilization in recently manic patients. Lamictal 606 Study Group [Abstract]. Presented at the Annual Meeting of the American Psychiatric Association Meeting, New Orleans, LA, May 5-10, 2001.

9. Denicoff KD, et al. Comparative prophylactic efficacy of lithium, carbamazepine, and the combination in bipolar disorder. J Clin Psychiatry 1997;58:470-8.

10. Greil W, et al. Lithium versus carbamazepine in the maintenance treatment of bipolar disorders—a randomized study. J Affect Disord 1997;43:151-61.

11. Hirschfeld RMA, Bowden CL, Gitlin MJ, et al. Practice guideline for the treatment of patients with bipolar disorder (revision). Am J Psychiatry, in press.

12. Tohen M, Baker R. Olanzapine versus divalproex sodium for the treatment of acute mania and maintenance of remission: a 47-week study. Presented at the Annual Meeting of the American Psychiatric Association. New Orleans, La, May 5-10, 2001.

13. Lambert PA, Venaud G. Comparative study of valpromide versus lithium as prophylactic treatment in affective disorders. Nervure J Psychiatrie 1982;4:1-9.

14. Revicki DA, Hirschfeld RMA, Keck PE, Jr, et al. Cost-effectiveness of divalproex sodium vs. lithium in long-term therapy for bipolar disorder. Presented at the Annual Meeting of the American College of Neuropsychopharmacology. San Juan, Puerto Rico, Dec. 13-17, 1999.

15. Dardennnes R, Even C, et al. Comparison of carbamazepine and lithium in the prophylaxis of bipolar disorders. A meta-analysis. Br J Psychiatry 1995;166:375-81.

16. Calabrese JR, Suppes T, et al. A double-blind, placebo-controlled, prophylaxis study of lamotrigine in rapid-cycling bipolar disorder. J Clin Psychiatry 2000;61:841-50.

17. Calabrese JR, Bowden CL, Sachs GS, et al. A double-blind placebo-controlled study of lamotrigine monotherapy in outpatients with bipolar I depression. J Clin Psychiatry 1999;60:79-88.

18. Frye MA, Ketter TA, Kimbrell TA, et al. A double-blind, placebo-controlled study of lamotrigine and gabapentin monotherapy in refractory mood disorders. J Clin Psychopharmacol 2000;24:133-40.

19. Tohen M. Olanazpine versus placebo in the treatment of acute bipolar mania. Presented at the XI World Congress of Psychiatry, World Psychiatric Association. Hamburg, Germany, Aug. 7, 1999.

20. Suppes T, Webb A, Paul B, et al. Clinical outcome in a randomized 1-year trial of clozapine versus treatment as usual for patients with treatment-resistant illness and a history of mania. Am J Psychiatry 1999;156:1164-9.

21. Keck PE, Jr, McElroy SL. Pharmacological treatment of bipolar disorder. In: Nathan PE, Gorman JM, eds. A Guide to Treatments that Work. New York: Oxford University Press, 2001, in press.

22. Keck PE, Jr, Licht R. Antipsychotic medications in the treatment of mood disorders. In: Buckley PF, Waddington JL, eds. Schizophrenia and Mood Disorders: The New Drug Therapies in Clinical Practice. Boston: Butterworth-Heiman, 2000;199-211.

23. Gelenberg AJ, Kane JM, et al. Comparison of standard and low serum levels of lithium for maintenance treatment of bipolar disorder. N Engl J Med 1989;321:1489-93.

24. Judd LL, Akiskal HS. Delineating the longitudinal structure of depressive illness: beyond clinical subtypes and duration thresholds. Pharmacopsychiatry 2000;33:3-7.

25. Goodnick PJ, Fieve RR, Schlegel A, et al. Inter-episode major and subclinical symptoms in affective disorder. Acta Psychiatr Scand 1987;75:592-600.

26. Keller MB, Lavori PW, et al. Subsyndromal symptoms in bipolar disorder: a comparison of standard and low serum lithium levels. Arch Gen Psychiatry 1992;49:371-6.

27. Post RM, Altshuler LL, Frye MA, et al. Rate of switch in bipolar patients prospectively treated with second-generation antidepressants as augmentation to mood stabilizers. Bipolar Disorders 2001;3:259-65.

28. Solomon DA, Ryan CE, Keitner GI, et al. A pilot study of lithium carbonate plus divalproex sodium for the continuation and maintenance treatment of patients with bipolar I disorder. J Clin Psychiatry 1997;58:95-9.

References

1. Solomon DA, Keitner GI, Miller IW, et al. Course of illness and maintenance treatments for patients with bipolar disorder. J Clin Psychiatry 1995;56:5-13.

2. Keck PE, Jr, McElroy SL, et al. Twelve-month outcome of bipolar patients following hospitalization for a manic or mixed episode. Am J Psychiatry 1998;155:646-52.

3. Sachs GS, Printz DJ, et al. The Expert Consensus Guideline Series: medication treatment of bipolar disorders 2000. Postgrad Med Special Report 2000;4:1-104.

4. Sachs GS, Thase ME. Bipolar disorder therapeutics: maintenance treatment. Biol Psychiatry 2000;48:573-81.

5. Keck PE, Jr, Welge JA, et al. Placebo effect in randomized, controlled maintenance studies of patients with bipolar disorder. Biol Psychiatry 2000;47:756-65.

6. Calabrese JR, Rapport DJ, Shelton MD, et al. Evolving methodologies in bipolar disorder maintenance research. Br J Psychiatry 2001;178 [Suppl 41]:157-63.

7. Bowden CL, Calabrese JR, McElroy SL, et al. A randomized, placebo-controlled 12-month trial of divalproex and lithium in treatment of outpatients with bipolar I disorder. Arch Gen Psychiatry 2000;57:481-9.

8. Calabrese JR, Bowden CL, DeVeaugh-Geiss J, et al. Lamotrigine demonstrates long term mood stabilization in recently manic patients. Lamictal 606 Study Group [Abstract]. Presented at the Annual Meeting of the American Psychiatric Association Meeting, New Orleans, LA, May 5-10, 2001.

9. Denicoff KD, et al. Comparative prophylactic efficacy of lithium, carbamazepine, and the combination in bipolar disorder. J Clin Psychiatry 1997;58:470-8.

10. Greil W, et al. Lithium versus carbamazepine in the maintenance treatment of bipolar disorders—a randomized study. J Affect Disord 1997;43:151-61.

11. Hirschfeld RMA, Bowden CL, Gitlin MJ, et al. Practice guideline for the treatment of patients with bipolar disorder (revision). Am J Psychiatry, in press.

12. Tohen M, Baker R. Olanzapine versus divalproex sodium for the treatment of acute mania and maintenance of remission: a 47-week study. Presented at the Annual Meeting of the American Psychiatric Association. New Orleans, La, May 5-10, 2001.

13. Lambert PA, Venaud G. Comparative study of valpromide versus lithium as prophylactic treatment in affective disorders. Nervure J Psychiatrie 1982;4:1-9.

14. Revicki DA, Hirschfeld RMA, Keck PE, Jr, et al. Cost-effectiveness of divalproex sodium vs. lithium in long-term therapy for bipolar disorder. Presented at the Annual Meeting of the American College of Neuropsychopharmacology. San Juan, Puerto Rico, Dec. 13-17, 1999.

15. Dardennnes R, Even C, et al. Comparison of carbamazepine and lithium in the prophylaxis of bipolar disorders. A meta-analysis. Br J Psychiatry 1995;166:375-81.

16. Calabrese JR, Suppes T, et al. A double-blind, placebo-controlled, prophylaxis study of lamotrigine in rapid-cycling bipolar disorder. J Clin Psychiatry 2000;61:841-50.

17. Calabrese JR, Bowden CL, Sachs GS, et al. A double-blind placebo-controlled study of lamotrigine monotherapy in outpatients with bipolar I depression. J Clin Psychiatry 1999;60:79-88.

18. Frye MA, Ketter TA, Kimbrell TA, et al. A double-blind, placebo-controlled study of lamotrigine and gabapentin monotherapy in refractory mood disorders. J Clin Psychopharmacol 2000;24:133-40.

19. Tohen M. Olanazpine versus placebo in the treatment of acute bipolar mania. Presented at the XI World Congress of Psychiatry, World Psychiatric Association. Hamburg, Germany, Aug. 7, 1999.

20. Suppes T, Webb A, Paul B, et al. Clinical outcome in a randomized 1-year trial of clozapine versus treatment as usual for patients with treatment-resistant illness and a history of mania. Am J Psychiatry 1999;156:1164-9.

21. Keck PE, Jr, McElroy SL. Pharmacological treatment of bipolar disorder. In: Nathan PE, Gorman JM, eds. A Guide to Treatments that Work. New York: Oxford University Press, 2001, in press.

22. Keck PE, Jr, Licht R. Antipsychotic medications in the treatment of mood disorders. In: Buckley PF, Waddington JL, eds. Schizophrenia and Mood Disorders: The New Drug Therapies in Clinical Practice. Boston: Butterworth-Heiman, 2000;199-211.

23. Gelenberg AJ, Kane JM, et al. Comparison of standard and low serum levels of lithium for maintenance treatment of bipolar disorder. N Engl J Med 1989;321:1489-93.

24. Judd LL, Akiskal HS. Delineating the longitudinal structure of depressive illness: beyond clinical subtypes and duration thresholds. Pharmacopsychiatry 2000;33:3-7.

25. Goodnick PJ, Fieve RR, Schlegel A, et al. Inter-episode major and subclinical symptoms in affective disorder. Acta Psychiatr Scand 1987;75:592-600.

26. Keller MB, Lavori PW, et al. Subsyndromal symptoms in bipolar disorder: a comparison of standard and low serum lithium levels. Arch Gen Psychiatry 1992;49:371-6.

27. Post RM, Altshuler LL, Frye MA, et al. Rate of switch in bipolar patients prospectively treated with second-generation antidepressants as augmentation to mood stabilizers. Bipolar Disorders 2001;3:259-65.

28. Solomon DA, Ryan CE, Keitner GI, et al. A pilot study of lithium carbonate plus divalproex sodium for the continuation and maintenance treatment of patients with bipolar I disorder. J Clin Psychiatry 1997;58:95-9.

Issue
Current Psychiatry - 01(04)
Issue
Current Psychiatry - 01(04)
Page Number
40-47
Page Number
40-47
Publications
Publications
Topics
Article Type
Display Headline
Update on bipolar disorder: How to better predict response to maintenance therapy
Display Headline
Update on bipolar disorder: How to better predict response to maintenance therapy
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Antiepileptic drugs for bipolar disorder: Are there any clear winners?

Article Type
Changed
Tue, 12/11/2018 - 15:13
Display Headline
Antiepileptic drugs for bipolar disorder: Are there any clear winners?

The newer antiepileptic drugs pose a sometimes bewildering range of options for bipolar disorder treatment. Which work best for acute bipolar I mania? Which are best suited for maintenance in patients with mixed episodes, or for those with a history of rapid cycling? What about prevention of depressive episodes? And how do the antiepileptics compare with lithium?

For many patients with bipolar disorder, lithium is still the drug of choice. For others, however, an increasing body of evidence supports the efficacy of some antiepileptics and atypical antipsychotics.

The mood-stabilizing properties of two antiepileptic agents, carbamazepine and valproate, were demonstrated some years ago in randomized controlled trials in patients with bipolar disorder. Since then, there has been considerable interest in the potential thymoleptic properties of the new antiepileptic drugs.1 In recent years gabapentin, lamotrigine, topiramate, oxcarbazepine, zonisamide, tiagabine, and levetiracetam have been approved in the United States for the treatment of various types of epilepsy. These medications have diverse pharmacological properties that distinguish them from earlier agents and from one another.

Do these new agents have anything to offer patients? For the most part, the evidence is not yet in hand, but we will examine what’s available, starting with the most recent trial data regarding the efficacy of valproate and carbamazepine.

Carbamazepine for bipolar mania

Five randomized, controlled trials2 have shown the efficacy of carbamazepine in patients with acute bipolar I mania. Carbamazepine was superior to placebo and comparable to chlorpromazine and lithium. Pooled data reveal an overall response rate (defined as the proportion of patients experiencing > 50% reduction in manic symptoms) of 50% for carbamazepine, 56% for lithium, and 61% for chlorpromazine (differences in overall response rates are not significant).

Until recently, the efficacy of carbamazepine as a maintenance therapy for bipolar disorder was controversial.3 However, two recent large randomized, controlled maintenance studies that compared carbamazepine with lithium validated use of the agent for that purpose.4,5

In the first study, 144 patients received either drug and were followed for up to 2 1/2 years.4 The study showed no significant differences between the two groups in time-to-mood episode recurrence or hospitalization. However, significantly more patients receiving carbamazepine required treatment discontinuation for side effects and additional medications for breakthrough symptoms than did the patients receiving lithium.

Patients without both comorbid disorders and mood-incongruent delusions responded better to lithium, whereas those with mixed episodes and bipolar II disorder or NOS appeared to respond better to carbamazepine.

In the second trial, 52 patients with bipolar I or II disorders received either lithium or carbamazepine for one year, crossed over to the alternate drug the second year, and received both drugs during the third year.5 No significant differences were reported in relapse rates during the first year between lithium (31%) and carbamazepine (37%), or in the percentage of patients who experienced a moderate or better response: 33% on lithium, 31% on carbamazepine, and 55% on the combination. On a variety of other measures, lithium was superior to carbamazepine. But patients with a history of rapid cycling responded significantly better to the combination (56%) than to lithium (28%) or carbamazepine (19%) alone.

As in the previous study, a higher proportion of patients receiving carbamazepine withdrew after experiencing side effects. Both studies found that while carbamazepine is efficacious, lithium is superior overall. But since neither study included a placebo group, carbamazepine’s efficacy in maintenance treatment cannot be determined.

Carbamazepine also was evaluated in three randomized, controlled trials in patients with bipolar depression.2 These small trials suggest that carbamazepine’s antidepressant activity may be less robust than its antimanic effects. Response rates (>50% improvement in depressive symptoms) ranged from 32% to 34%, although many patients who participated had treatment-resistant bipolar depression.

Comparing the known efficacy of antiepileptic agents in bipolar disorder

DrugManiaDepressionMaintenanceComments
Valproate◊◊◊◊◊◊◊New Depakote ER formulation
Carbamazepine◊◊◊◊◊◊◊◊2 new maintenance studies v. lithium
Gabapentin2 negative placebo-controlled studies in mania
Lamotrigine×◊◊◊◊◊◊Antidepressant activity in several controlled trials
TopiramateDose-related weight loss
Oxcarbazepine◊◊Improved tolerability & pharmacokinetics
ZonisamideNDNDMay produce weight loss in some patients
Tiagabine×NDNDMore data needed regarding tolerability and efficacy
LevetiracetamNDNDNDData needed regarding efficacy and tolerability

Key

◊◊◊◊ efficacy demonstrated in ≥2 placebo-controlled trials

◊◊◊ efficacy demonstrated in one placebo-controlled or two large, active comparator trials

◊◊ efficacy in two small or one large active comparator trial

◊ efficacy only in open trials and case series

× conflicting evidence of efficacy in available studies

lack of efficacy demonstrated in randomized, controlled trials

ND no data presently available

Based on the studies reviewed above, carbamazepine is considered a second-line agent for bipolar mania and maintenance treatment with very limited data regarding its antidepressant effects.6

 

 

Valproate: For patients with mixed and mood episodes

The evidence seems to point to valproate as a suitable agent for patients with mixed and mood episodes.

Two earlier randomized, controlled studies7,8 showed that the divalproex sodium formulation of valproate was superior to a placebo, leading to the indication of divalproex for treatment of acute mania in patients with bipolar disorder. Additional analyses of data from the second controlled trial8 indicated that patients with prominent depressive symptoms during mania (mixed episodes) responded better to divalproex than lithium. Further, multiple prior mood episodes were associated with poor lithium response but not with divalproex response.

Two recent randomized, controlled trials compared the antimanic efficacy and tolerability of divalproex and olanzapine.9,10 The design of these two studies differed in two important ways: sample size and starting doses. This may explain the different results of the two trials.

In the first study, 248 patients received starting doses of either divalproex 750 mg/d with upward titration to therapeutic concentrations, or olanzapine 15 mg/d with titration if clinically indicated.9 Olanzapine was found to be superior in the mean reduction of manic symptoms and in the proportion of patients who either responded to treatment or were in remission.

In the second study, the number of patients (N=120) randomized was not sufficient to detect a statistically significant difference between treatment groups.10 Initial dosing consisted of rapid divalproex loading (20 mg/kg/d) versus olanzapine (10 mg/d) with upward titration as clinically indicated. This trial revealed no significant differences in efficacy on any outcome measure and the mean valproic acid plasma concentration was higher than in the first trial.

Differences in side effects between the two agents were similar in both studies. Olanzapine was associated with greater sedation, appetite stimulation, and weight gain, and divalproex with greater gastrointestinal symptoms. Taken together, these two studies indicate that olanzapine is at least as efficacious as divalproex in treating acute mania and that divalproex should be titrated to plasma concentrations well within the therapeutic range consistent with response and tolerability.

The efficacy of divalproex as a maintenance therapy for patients with bipolar disorder has been studied in four trials to date:

  1. A randomized, open comparison of lithium and divalproex found generally good efficacy for both drugs across 18 months.
  2. A second naturalistic, pharmacoeconomic, one-year open comparison also found both lithium and divalproex fairly equal in efficacy.
  3. A large, prospective, randomized one-year maintenance trial showed little difference in relapse rates among patients receiving divalproex (24%), lithium (31%), and a placebo (38%).11
  4. A recently completed one-year comparison of relapse rates between divalproex and olanzapine showed little difference between the two drugs.12

The overall results suggest that divalproex helps prevent mood episodes in patients with bipolar disorder, but the data are less substantial and conclusive than from placebo-controlled trials of lithium.

The antidepressant effects of divalproex in treating acute bipolar depression have not been studied in randomized controlled trials. Impressions from case reports and case series suggest that divalproex may exert some antidepressant activity, but that this action may be less robust than its antimanic effects.

Divalproex is now available in a once-daily extended release (ER) formulation. This appears to have improved tolerability, especially regarding gastrointestinal side effects. (Clinical experience suggests that the immediate release formulation of divalproex can also be given once daily.) The ER formulation is not bioequivalent to its immediate-release counterpart; it produces plasma concentrations of approximately 80% of those achieved with immediate release. Thus, switching a patient to the ER formulation might require a dose increase.

Lamotrigine for bipolar depression

Several recent randomized controlled trials indicate that lamotrigine has important thymoleptic properties.14-17

Three studies addressed the efficacy of lamotrigine in treating patients with acute bipolar mania. In two small trials, lamotrigine did not display superior efficacy over placebo in reducing manic symptoms.14 A third trial revealed differences between lithium and lamotrigine in reducing manic symptoms, but this study lacked sufficient power to detect potential differences in efficacy.

Two placebo-controlled maintenance studies of lamotrigine were recently reported.15,16 The first found no significant differences in relapse rates in patients with rapid cycling bipolar I and II disorders randomized to lamotrigine or placebo after initial stabilization on lamotrigine.15 However, in a post hoc analysis among bipolar II (but not bipolar I) patients, lamotrigine was significantly more efficacious than placebo in time-to-study dropout and considerably better (P=0.07) in time to need for additional medication.

In the second randomized, placebo-controlled trial, patients with bipolar I disorder who had recently experienced a manic episode were treated with lamotrigine 100-200 mg/d during an open-label phase (8-16 weeks) while other psychotropic agents were tapered and discontinued.16 Patients (N = 171) who remained stable were then randomized to lamotrigine 200-400 mg/d, lithium, or placebo for up to 18 months.

 

 

The lamotrigine-treated group showed significantly lower relapse rates than placebo-treated patients in time-to-study dropout, time to intervention for a mood episode, and time to intervention for depressive relapse. Differences between the lamotrigine and placebo groups in time to intervention for manic relapse were insignificant. Lithium was superior to a placebo in time to relapse for any mood episode and time to intervention for a manic episode. Lithium also outperformed lamotrigine in that manic symptoms did not worsen as quickly from baseline to endpoint.

The results here are consistent with those of the rapid cycling study: Lamotrigine helps prevent recurrence of depressive symptoms and episodes. Evidence of prophylaxis against manic recurrences was not compelling, however.

The findings of these two maintenance trials are consistent with the results of a placebo-controlled, randomized, 6-week acute treatment trial of lamotrigine (50 mg/d and 200 mg/d) in patients with bipolar depression.17 Seventeen lamotrigine-treated patients in both dosage groups saw more-significantly reduced depressive symptoms than did placebo-treated patients on the MADRS (but not the HAMD) total score and on the CGI. The 200 mg/d group tended to have greater improvement than the 50 mg/d group. There was little difference among the three treatment groups in the incidence of switching into hypomania or mania.

These studies indicate that lamotrigine has acute and prophylactic efficacy against bipolar depression. In contrast, evidence of the agent’s acute or prophylactic efficacy in mania is lacking.

Other new antiepileptics: More testing needed

Use of the six other newer antiepileptics in patients with bipolar disorder is still being explored. (See “Comparing the known efficacy of antiepileptic agents in bipolar disorder,”) Let’s look at what we know about these agents to this point.

Gabapentin A number of case reports and case series published in recent years suggest that gabapentin may have mood-stabilizing properties. In two randomized, controlled trials, however, gabapentin did not display significantly greater efficacy than a placebo in treating acute mania.13,14 Gabapentin has not been studied in controlled trials as a maintenance treatment or for bipolar depression.

In contrast to these negative studies, gabapentin was superior to placebo in studies of patients with panic disorder, social anxiety disorder, and neuropathic pain. Overall, these studies suggest that gabapentin is not a bona fide mood stabilizer for most patients.

Topiramate More than 10 case reports and case series suggest that topiramate may have mood-stabilizing properties. A number of open trials also have found significant dose-related weight loss in patients with weight gain associated with other psychotropic agents.18 These observations have sparked interest in topiramate’s potential role as an obesity treatment.

Only one randomized, controlled trial of topiramate for bipolar disorder has appeared to date.19 An interim analysis of a placebo-controlled, randomized trial of two doses of topiramate (approximately 250 mg/d and 500 mg/d) in 97 hospitalized patients with acute bipolar I mania revealed strong trends toward reduced manic symptoms for both topiramate groups over placebo. These differences were not significant by the time the study was concluded, however.

CHARACTERISTICS OF THE NEWER ANTIEPILEPTICS

Lamotrigine is a novel drug that blocks voltage-sensitive sodium channels, thereby indirectly inhibiting the release of excitatory neurotransmitters, particularly glutamate and aspartate.

Gabapentin was developed to mimic the synaptic effects of gamma-aminobutyric acid (GABA); it does not appear to appreciably interact with GABA receptors, however, and its mechanism of action in epilepsy remains unknown. It has several attractive pharmacokinetic properties, including lack of protein binding, renal clearance rather than hepatic metabolism, and few known drug-drug interactions.

Topiramate is a sulfamate-substituted monosaccharide with a number of possible mechanisms of action, including blockade of voltage-gated sodium channels, antagonism of the kainate/AMPA glutamate receptor subtype, enhancement of GABA activity at the GABA A receptor via interaction with a nonbenzodiazepine receptor site, and carbonic anhydrase inhibition.

Oxcarbazepine is the 10-keto analogue of carbamazepine, a chemical difference that translates into a number of safety advantages over carbamazepine. Oxcarbazepine is converted to an active 10-hydroxy metabolite rather than to the 10,11-epoxide metabolite of carbamazepine. The 10,11-epoxide metabolite of carbamazepine is associated with neurological side effects. Oxcarbazepine is a weak inducer of the CYP450 system, appears to have fewer drug-drug interactions, and offers better overall tolerability.

Zonisamide, a sulfonamide derivative, blocks voltage-sensitive sodium channels and T-type calcium currents, modulates GABAergic and dopaminergic neurotransmission, and is a free-radical scavenger.

Tiagabine is a selective GABA reuptake inhibitor.

Levetiracetam is the S-enantiomer of the ethyl analogue of the nootropic agent piracetam. Its mechanism of action in treating epilepsy is unknown. It does not appear to interact significantly with voltage-sensitive sodium channels or T-type calcium channels, nor does it significantly alter levels of GABA, glutamate, or glutamine in the central nervous system.

 

 

When patients with mania associated with recent antidepressant use were excluded from post hocanalysis, topiramate’s superiority over placebo was re-established. These tantalizing results require further study. Similarly, studies of topiramate’s potential efficacy in acute bipolar depression and as a maintenance treatment are needed.

Oxcarbazepine Oxcarbazepine is a relative newcomer to the United States, but has been available abroad for some time.

While numerous studies have tested carbamazepine in treating various aspects of bipolar disorder, few controlled trials have tested oxcarbazepine for this use.20

Oxcarbazepine as a treatment for acute bipolar mania was comparable to haloperidol in two small randomized, controlled trials and to lithium in one small trial. None of these studies had adequate power to detect potential differences in efficacy. Similarly, two very small maintenance studies (total N=25) found no differences in efficacy between oxcarbazepine and lithium.20

Additionally, to our knowledge oxcarbazepine has not been formally tested as a treatment for acute bipolar depression. So while oxcarbazepine is an attractive alternative to carbamazepine for pharmacokinetic and pharmacodynamic reasons, data supporting oxcarbazepine’s efficacy in bipolar disorder are far less substantial.

Zonisamide The only report of zonisamide as a bipolar disorder treatment so far is a case series of 15 patients who received the drug adjunctively for manic symptoms. Of these patients, 80% showed at least moderate improvement and 33% were rated as markedly improved. These preliminary observations require further study. Like topiramate, zonisamide may also be associated with weight loss in some patients.

Tiagabine A few case reports and case series describe tiagabine’s effects in patients with bipolar disorder. Conflicting results have been reported to date and some reports of poor tolerability have emerged. Fundamental safety and efficacy data for this agent in bipolar disorder are needed.

Levetiracetam Studies of levetiracetam for treatment of bipolar disorder are just under way. The Stanley Foundation Bipolar Treatment Network, a program of the National Alliance for the Mentally Ill Research Institute, is investigating its potential thymoleptic properties.

Related resources

  • Joffe RT, Calabrese JR. Anticonvulsants in Mood Disorders. New York: Marcel Dekker Inc., 1994.
  • Modigh K, Robak OH, Vestergaard P. Anticonvulsants in Psychiatry. Bristol, Pa: Wrightson Biomedical Publishing, Ltd., 1994.
  • Schatzberg AF, Nemeroff CB. Textbook of Psychopharmacology, 2 nd ed. Washington, DC: American Psychiatric Press, 1998.
  • Nathan P, Gorman J. A Guide to Treatments that Work, 2 nd ed. New York: Oxford University Press, 2001.
  • Janicak PG, Davis JM, Preskorn SH, Ayd FJ, Jr. Principles and Practice of Psychopharmacotherapy, 2 nd ed. Baltimore, MD: Williams & Wilkins, 1997.

Drug brand names

  • Carbamazepine • Tegretol, Epitol, Atretol
  • Gabapentin • Neurontin
  • Lamotrigine • Lamictal
  • Levetiracetam • Keppra
  • Oxcarbazepine • Trileptal
  • Tiagabine • Gabatril
  • Topiramate • Topamax
  • Valproate-divalproex sodium • Depakote, Depakote ER
  • Zonisamide • Zonegran

Disclosure

Dr. Keck reports that he receives grant/research support from and serves as a consultant to Abbott Laboratories, AstraZeneca, Pfizer Inc., and Eli Lilly and Co. He also receives grant/research support from Merck and Co. and Otsuka America Pharmaceutical, and serves as a consultant to Bristol-Myers Squibb Co., GlaxoSmithKline, and Janssen Pharmaceutica.

Dr. McElroy reports that she receives grant/research support from and serves as a consultant to Abbott Laboratories, Elan Pharmaceuticals, Cephalon Inc. GlaxoSmithKline, and Eli Lilly and Co. She also receives grant/research support from Forest Pharmaceuticals and Solvay Pharmaceuticals, and serves as a consultant to Bristol-Myers Squibb Co., Ortho-McNeil Pharmaceutical, and Janssen Pharmaceutica.

References

1. Keck PE, Jr, McElroy SL, Strakowski SM. Anticonvulsants and antipsychotics in the treatment of bipolar disorder. J Clin Psychiatry. 1998;59(Suppl 6):74-81.

2. McElroy SL, Keck PE, Jr. Pharmacologic agents for the treatment of acute bipolar mania. Biol Psychiatry. 2000;48:539-557.

3. Post RM, Denicoff KD, Frye MA, Leverich GS. Re-evaluating carbamazepine prophylaxis in bipolar disorder. Br J Psychiatry. 1997;170:202-204.

4. Greil W, Ludwig-Mayerhofer W, Erazon-Scheichlin C, Schmidt S, Engel RR, et al. Lithium versus carbamazepine in the maintenance treatment of bipolar disorders–a randomized study. J Affect Disorders. 1997;43:151-161.

5. Denicoff KD, Smith-Jackson EE, Disney ER, Ali SO, Leverich GS, Post RM. Comparative prophylactic efficacy of lithium, carbamazepine and the combination in bipolar disorder. J Clin Psychiatry. 1997;58:470-478.

6. Suppes T, Swann AC, Dennehy EB, Habermacher ED, Mason M, Crismon ML, Toprac MG, Rush AJ, Shon SP, Altshuler KZ. Texas Medication Algorithm Project: development and feasibility testing of a treatment algorithm for patients with bipolar disorder. J Clin Psychiatry. 2001;62:439-447.

7. Pope HG, Jr, McElroy SL, Keck PE, Jr, Hudson JI. Valproate in the treatment of acute mania: a placebo-controlled study. Arch Gen Psychiatry. 1991;48:62-68.

8. Bowden CL, Brugger AM, Swann AC, Calabrese JR, Janicak PG, Petty F, et al. Efficacy of divalproex sodium vs. lithium and placebo in the treatment of mania. JAMA. 1994;271:915-924.

9. Tohen M, Baker RW, Milton DR, Risser RC, Gilmore JA, Davis AR, et al. Olanzapine versus divalproex sodium for the treatment of acute mania. Bipolar Disorders. 2001;3(Suppl 1):60-61.

10. Zajecka J. Divalproex versus olanzapine in the treatment of acute mania. Presented at the 39 th Annual Meeting of the American College of Neuropsychopharmacology. San Juan, Puerto Rico, Dec. 10-14, 2000.

11. Bowden CL, Calabrese JR, McElroy SL, Hirschfeld RMA, Petty F, Gyulai LL, Pope HG, Jr, et al. Efficacy of divalproex versus lithium and placebo in maintenance treatment of bipolar disorder. Arch Gen Psychiatry. 2000;57:481-489.

12. Tohen M, Baker RW, Altshuler LL, Zarate CA, Suppes T, Ketter T, et al. Olanzapine versus divalproex for bipolar mania: a 47-week study. Presented at the European College of Neuropsychopharmacology Meeting, Istanbul, Turkey, Oct. 15, 2001.

13. Pande AD, Crockatt JG, Janney CA, Werth JL, Tsaaroucha G. and the Gabapentin Bipolar Disorder Study Group. Bipolar Disorders. 2000;2:249-255.

14. Frye MA, Ketter TA, Kimbrell TA, et al. A placebo controlled study of lamotrigine and gabapentin monotherapy in refractory mood disorders. J Clin Psychopharmacol. 2000;24:205-212.

15. Calabrese JR, Suppes T, Bowden CL, et al. A double-blind, placebo-controlled study of lamotrigine monotherapy in rapid-cycling bipolar disorder. J Clin Psychiatry. 2000;61:841-850.

16. Bowden CL, Calabrese JR, Akthar S, Olajossy M, Montgomery S, Ascher J, et al. Lamotrigine demonstrates long-term mood stabilization in manic patients. Bipolar Disorders. 2001;3(Suppl 1):27-28.

17. Calabrese JR, Bowden CL, Sachs GS, et al. A double-blind, placebo-controlled study of lamotrigine monotherapy in outpatients with bipolar I depression. J Clin Psychiatry. 1999;60:79-88.

18. McElroy SL, Suppes T, Keck PE, Jr, Frye MA, Denicoff KD, Altshuler LL, et al. Open-label adjunctive topiramate in the treatment of bipolar disorders. Biol Psychiatry. 2000;47:1025-1033.

19. Calabrese JR. A double-blind, placebo-controlled trial of topiramate monotherapy in the treatment of acute mania. Presented at the Annual Meeting of the European College of Neuropsychopharmacology. Munich, Germany, Sept. 9-13, 2000.

20. Emrich H, Schiwy W, Silverstone T. eds. Carbamazepine and Oxcarbazepine in Psychiatry. London: CNS Publishers, 1990.

Article PDF
Author and Disclosure Information

Paul E. Keck, Jr, MD
Susan L. McElroy, MD
Biological Psychiatry Program, Department of Psychiatry University of Cincinnati College of Medicine, Cincinnati, Ohio

Issue
Current Psychiatry - 01(01)
Publications
Topics
Page Number
18-24
Sections
Author and Disclosure Information

Paul E. Keck, Jr, MD
Susan L. McElroy, MD
Biological Psychiatry Program, Department of Psychiatry University of Cincinnati College of Medicine, Cincinnati, Ohio

Author and Disclosure Information

Paul E. Keck, Jr, MD
Susan L. McElroy, MD
Biological Psychiatry Program, Department of Psychiatry University of Cincinnati College of Medicine, Cincinnati, Ohio

Article PDF
Article PDF

The newer antiepileptic drugs pose a sometimes bewildering range of options for bipolar disorder treatment. Which work best for acute bipolar I mania? Which are best suited for maintenance in patients with mixed episodes, or for those with a history of rapid cycling? What about prevention of depressive episodes? And how do the antiepileptics compare with lithium?

For many patients with bipolar disorder, lithium is still the drug of choice. For others, however, an increasing body of evidence supports the efficacy of some antiepileptics and atypical antipsychotics.

The mood-stabilizing properties of two antiepileptic agents, carbamazepine and valproate, were demonstrated some years ago in randomized controlled trials in patients with bipolar disorder. Since then, there has been considerable interest in the potential thymoleptic properties of the new antiepileptic drugs.1 In recent years gabapentin, lamotrigine, topiramate, oxcarbazepine, zonisamide, tiagabine, and levetiracetam have been approved in the United States for the treatment of various types of epilepsy. These medications have diverse pharmacological properties that distinguish them from earlier agents and from one another.

Do these new agents have anything to offer patients? For the most part, the evidence is not yet in hand, but we will examine what’s available, starting with the most recent trial data regarding the efficacy of valproate and carbamazepine.

Carbamazepine for bipolar mania

Five randomized, controlled trials2 have shown the efficacy of carbamazepine in patients with acute bipolar I mania. Carbamazepine was superior to placebo and comparable to chlorpromazine and lithium. Pooled data reveal an overall response rate (defined as the proportion of patients experiencing > 50% reduction in manic symptoms) of 50% for carbamazepine, 56% for lithium, and 61% for chlorpromazine (differences in overall response rates are not significant).

Until recently, the efficacy of carbamazepine as a maintenance therapy for bipolar disorder was controversial.3 However, two recent large randomized, controlled maintenance studies that compared carbamazepine with lithium validated use of the agent for that purpose.4,5

In the first study, 144 patients received either drug and were followed for up to 2 1/2 years.4 The study showed no significant differences between the two groups in time-to-mood episode recurrence or hospitalization. However, significantly more patients receiving carbamazepine required treatment discontinuation for side effects and additional medications for breakthrough symptoms than did the patients receiving lithium.

Patients without both comorbid disorders and mood-incongruent delusions responded better to lithium, whereas those with mixed episodes and bipolar II disorder or NOS appeared to respond better to carbamazepine.

In the second trial, 52 patients with bipolar I or II disorders received either lithium or carbamazepine for one year, crossed over to the alternate drug the second year, and received both drugs during the third year.5 No significant differences were reported in relapse rates during the first year between lithium (31%) and carbamazepine (37%), or in the percentage of patients who experienced a moderate or better response: 33% on lithium, 31% on carbamazepine, and 55% on the combination. On a variety of other measures, lithium was superior to carbamazepine. But patients with a history of rapid cycling responded significantly better to the combination (56%) than to lithium (28%) or carbamazepine (19%) alone.

As in the previous study, a higher proportion of patients receiving carbamazepine withdrew after experiencing side effects. Both studies found that while carbamazepine is efficacious, lithium is superior overall. But since neither study included a placebo group, carbamazepine’s efficacy in maintenance treatment cannot be determined.

Carbamazepine also was evaluated in three randomized, controlled trials in patients with bipolar depression.2 These small trials suggest that carbamazepine’s antidepressant activity may be less robust than its antimanic effects. Response rates (>50% improvement in depressive symptoms) ranged from 32% to 34%, although many patients who participated had treatment-resistant bipolar depression.

Comparing the known efficacy of antiepileptic agents in bipolar disorder

DrugManiaDepressionMaintenanceComments
Valproate◊◊◊◊◊◊◊New Depakote ER formulation
Carbamazepine◊◊◊◊◊◊◊◊2 new maintenance studies v. lithium
Gabapentin2 negative placebo-controlled studies in mania
Lamotrigine×◊◊◊◊◊◊Antidepressant activity in several controlled trials
TopiramateDose-related weight loss
Oxcarbazepine◊◊Improved tolerability & pharmacokinetics
ZonisamideNDNDMay produce weight loss in some patients
Tiagabine×NDNDMore data needed regarding tolerability and efficacy
LevetiracetamNDNDNDData needed regarding efficacy and tolerability

Key

◊◊◊◊ efficacy demonstrated in ≥2 placebo-controlled trials

◊◊◊ efficacy demonstrated in one placebo-controlled or two large, active comparator trials

◊◊ efficacy in two small or one large active comparator trial

◊ efficacy only in open trials and case series

× conflicting evidence of efficacy in available studies

lack of efficacy demonstrated in randomized, controlled trials

ND no data presently available

Based on the studies reviewed above, carbamazepine is considered a second-line agent for bipolar mania and maintenance treatment with very limited data regarding its antidepressant effects.6

 

 

Valproate: For patients with mixed and mood episodes

The evidence seems to point to valproate as a suitable agent for patients with mixed and mood episodes.

Two earlier randomized, controlled studies7,8 showed that the divalproex sodium formulation of valproate was superior to a placebo, leading to the indication of divalproex for treatment of acute mania in patients with bipolar disorder. Additional analyses of data from the second controlled trial8 indicated that patients with prominent depressive symptoms during mania (mixed episodes) responded better to divalproex than lithium. Further, multiple prior mood episodes were associated with poor lithium response but not with divalproex response.

Two recent randomized, controlled trials compared the antimanic efficacy and tolerability of divalproex and olanzapine.9,10 The design of these two studies differed in two important ways: sample size and starting doses. This may explain the different results of the two trials.

In the first study, 248 patients received starting doses of either divalproex 750 mg/d with upward titration to therapeutic concentrations, or olanzapine 15 mg/d with titration if clinically indicated.9 Olanzapine was found to be superior in the mean reduction of manic symptoms and in the proportion of patients who either responded to treatment or were in remission.

In the second study, the number of patients (N=120) randomized was not sufficient to detect a statistically significant difference between treatment groups.10 Initial dosing consisted of rapid divalproex loading (20 mg/kg/d) versus olanzapine (10 mg/d) with upward titration as clinically indicated. This trial revealed no significant differences in efficacy on any outcome measure and the mean valproic acid plasma concentration was higher than in the first trial.

Differences in side effects between the two agents were similar in both studies. Olanzapine was associated with greater sedation, appetite stimulation, and weight gain, and divalproex with greater gastrointestinal symptoms. Taken together, these two studies indicate that olanzapine is at least as efficacious as divalproex in treating acute mania and that divalproex should be titrated to plasma concentrations well within the therapeutic range consistent with response and tolerability.

The efficacy of divalproex as a maintenance therapy for patients with bipolar disorder has been studied in four trials to date:

  1. A randomized, open comparison of lithium and divalproex found generally good efficacy for both drugs across 18 months.
  2. A second naturalistic, pharmacoeconomic, one-year open comparison also found both lithium and divalproex fairly equal in efficacy.
  3. A large, prospective, randomized one-year maintenance trial showed little difference in relapse rates among patients receiving divalproex (24%), lithium (31%), and a placebo (38%).11
  4. A recently completed one-year comparison of relapse rates between divalproex and olanzapine showed little difference between the two drugs.12

The overall results suggest that divalproex helps prevent mood episodes in patients with bipolar disorder, but the data are less substantial and conclusive than from placebo-controlled trials of lithium.

The antidepressant effects of divalproex in treating acute bipolar depression have not been studied in randomized controlled trials. Impressions from case reports and case series suggest that divalproex may exert some antidepressant activity, but that this action may be less robust than its antimanic effects.

Divalproex is now available in a once-daily extended release (ER) formulation. This appears to have improved tolerability, especially regarding gastrointestinal side effects. (Clinical experience suggests that the immediate release formulation of divalproex can also be given once daily.) The ER formulation is not bioequivalent to its immediate-release counterpart; it produces plasma concentrations of approximately 80% of those achieved with immediate release. Thus, switching a patient to the ER formulation might require a dose increase.

Lamotrigine for bipolar depression

Several recent randomized controlled trials indicate that lamotrigine has important thymoleptic properties.14-17

Three studies addressed the efficacy of lamotrigine in treating patients with acute bipolar mania. In two small trials, lamotrigine did not display superior efficacy over placebo in reducing manic symptoms.14 A third trial revealed differences between lithium and lamotrigine in reducing manic symptoms, but this study lacked sufficient power to detect potential differences in efficacy.

Two placebo-controlled maintenance studies of lamotrigine were recently reported.15,16 The first found no significant differences in relapse rates in patients with rapid cycling bipolar I and II disorders randomized to lamotrigine or placebo after initial stabilization on lamotrigine.15 However, in a post hoc analysis among bipolar II (but not bipolar I) patients, lamotrigine was significantly more efficacious than placebo in time-to-study dropout and considerably better (P=0.07) in time to need for additional medication.

In the second randomized, placebo-controlled trial, patients with bipolar I disorder who had recently experienced a manic episode were treated with lamotrigine 100-200 mg/d during an open-label phase (8-16 weeks) while other psychotropic agents were tapered and discontinued.16 Patients (N = 171) who remained stable were then randomized to lamotrigine 200-400 mg/d, lithium, or placebo for up to 18 months.

 

 

The lamotrigine-treated group showed significantly lower relapse rates than placebo-treated patients in time-to-study dropout, time to intervention for a mood episode, and time to intervention for depressive relapse. Differences between the lamotrigine and placebo groups in time to intervention for manic relapse were insignificant. Lithium was superior to a placebo in time to relapse for any mood episode and time to intervention for a manic episode. Lithium also outperformed lamotrigine in that manic symptoms did not worsen as quickly from baseline to endpoint.

The results here are consistent with those of the rapid cycling study: Lamotrigine helps prevent recurrence of depressive symptoms and episodes. Evidence of prophylaxis against manic recurrences was not compelling, however.

The findings of these two maintenance trials are consistent with the results of a placebo-controlled, randomized, 6-week acute treatment trial of lamotrigine (50 mg/d and 200 mg/d) in patients with bipolar depression.17 Seventeen lamotrigine-treated patients in both dosage groups saw more-significantly reduced depressive symptoms than did placebo-treated patients on the MADRS (but not the HAMD) total score and on the CGI. The 200 mg/d group tended to have greater improvement than the 50 mg/d group. There was little difference among the three treatment groups in the incidence of switching into hypomania or mania.

These studies indicate that lamotrigine has acute and prophylactic efficacy against bipolar depression. In contrast, evidence of the agent’s acute or prophylactic efficacy in mania is lacking.

Other new antiepileptics: More testing needed

Use of the six other newer antiepileptics in patients with bipolar disorder is still being explored. (See “Comparing the known efficacy of antiepileptic agents in bipolar disorder,”) Let’s look at what we know about these agents to this point.

Gabapentin A number of case reports and case series published in recent years suggest that gabapentin may have mood-stabilizing properties. In two randomized, controlled trials, however, gabapentin did not display significantly greater efficacy than a placebo in treating acute mania.13,14 Gabapentin has not been studied in controlled trials as a maintenance treatment or for bipolar depression.

In contrast to these negative studies, gabapentin was superior to placebo in studies of patients with panic disorder, social anxiety disorder, and neuropathic pain. Overall, these studies suggest that gabapentin is not a bona fide mood stabilizer for most patients.

Topiramate More than 10 case reports and case series suggest that topiramate may have mood-stabilizing properties. A number of open trials also have found significant dose-related weight loss in patients with weight gain associated with other psychotropic agents.18 These observations have sparked interest in topiramate’s potential role as an obesity treatment.

Only one randomized, controlled trial of topiramate for bipolar disorder has appeared to date.19 An interim analysis of a placebo-controlled, randomized trial of two doses of topiramate (approximately 250 mg/d and 500 mg/d) in 97 hospitalized patients with acute bipolar I mania revealed strong trends toward reduced manic symptoms for both topiramate groups over placebo. These differences were not significant by the time the study was concluded, however.

CHARACTERISTICS OF THE NEWER ANTIEPILEPTICS

Lamotrigine is a novel drug that blocks voltage-sensitive sodium channels, thereby indirectly inhibiting the release of excitatory neurotransmitters, particularly glutamate and aspartate.

Gabapentin was developed to mimic the synaptic effects of gamma-aminobutyric acid (GABA); it does not appear to appreciably interact with GABA receptors, however, and its mechanism of action in epilepsy remains unknown. It has several attractive pharmacokinetic properties, including lack of protein binding, renal clearance rather than hepatic metabolism, and few known drug-drug interactions.

Topiramate is a sulfamate-substituted monosaccharide with a number of possible mechanisms of action, including blockade of voltage-gated sodium channels, antagonism of the kainate/AMPA glutamate receptor subtype, enhancement of GABA activity at the GABA A receptor via interaction with a nonbenzodiazepine receptor site, and carbonic anhydrase inhibition.

Oxcarbazepine is the 10-keto analogue of carbamazepine, a chemical difference that translates into a number of safety advantages over carbamazepine. Oxcarbazepine is converted to an active 10-hydroxy metabolite rather than to the 10,11-epoxide metabolite of carbamazepine. The 10,11-epoxide metabolite of carbamazepine is associated with neurological side effects. Oxcarbazepine is a weak inducer of the CYP450 system, appears to have fewer drug-drug interactions, and offers better overall tolerability.

Zonisamide, a sulfonamide derivative, blocks voltage-sensitive sodium channels and T-type calcium currents, modulates GABAergic and dopaminergic neurotransmission, and is a free-radical scavenger.

Tiagabine is a selective GABA reuptake inhibitor.

Levetiracetam is the S-enantiomer of the ethyl analogue of the nootropic agent piracetam. Its mechanism of action in treating epilepsy is unknown. It does not appear to interact significantly with voltage-sensitive sodium channels or T-type calcium channels, nor does it significantly alter levels of GABA, glutamate, or glutamine in the central nervous system.

 

 

When patients with mania associated with recent antidepressant use were excluded from post hocanalysis, topiramate’s superiority over placebo was re-established. These tantalizing results require further study. Similarly, studies of topiramate’s potential efficacy in acute bipolar depression and as a maintenance treatment are needed.

Oxcarbazepine Oxcarbazepine is a relative newcomer to the United States, but has been available abroad for some time.

While numerous studies have tested carbamazepine in treating various aspects of bipolar disorder, few controlled trials have tested oxcarbazepine for this use.20

Oxcarbazepine as a treatment for acute bipolar mania was comparable to haloperidol in two small randomized, controlled trials and to lithium in one small trial. None of these studies had adequate power to detect potential differences in efficacy. Similarly, two very small maintenance studies (total N=25) found no differences in efficacy between oxcarbazepine and lithium.20

Additionally, to our knowledge oxcarbazepine has not been formally tested as a treatment for acute bipolar depression. So while oxcarbazepine is an attractive alternative to carbamazepine for pharmacokinetic and pharmacodynamic reasons, data supporting oxcarbazepine’s efficacy in bipolar disorder are far less substantial.

Zonisamide The only report of zonisamide as a bipolar disorder treatment so far is a case series of 15 patients who received the drug adjunctively for manic symptoms. Of these patients, 80% showed at least moderate improvement and 33% were rated as markedly improved. These preliminary observations require further study. Like topiramate, zonisamide may also be associated with weight loss in some patients.

Tiagabine A few case reports and case series describe tiagabine’s effects in patients with bipolar disorder. Conflicting results have been reported to date and some reports of poor tolerability have emerged. Fundamental safety and efficacy data for this agent in bipolar disorder are needed.

Levetiracetam Studies of levetiracetam for treatment of bipolar disorder are just under way. The Stanley Foundation Bipolar Treatment Network, a program of the National Alliance for the Mentally Ill Research Institute, is investigating its potential thymoleptic properties.

Related resources

  • Joffe RT, Calabrese JR. Anticonvulsants in Mood Disorders. New York: Marcel Dekker Inc., 1994.
  • Modigh K, Robak OH, Vestergaard P. Anticonvulsants in Psychiatry. Bristol, Pa: Wrightson Biomedical Publishing, Ltd., 1994.
  • Schatzberg AF, Nemeroff CB. Textbook of Psychopharmacology, 2 nd ed. Washington, DC: American Psychiatric Press, 1998.
  • Nathan P, Gorman J. A Guide to Treatments that Work, 2 nd ed. New York: Oxford University Press, 2001.
  • Janicak PG, Davis JM, Preskorn SH, Ayd FJ, Jr. Principles and Practice of Psychopharmacotherapy, 2 nd ed. Baltimore, MD: Williams & Wilkins, 1997.

Drug brand names

  • Carbamazepine • Tegretol, Epitol, Atretol
  • Gabapentin • Neurontin
  • Lamotrigine • Lamictal
  • Levetiracetam • Keppra
  • Oxcarbazepine • Trileptal
  • Tiagabine • Gabatril
  • Topiramate • Topamax
  • Valproate-divalproex sodium • Depakote, Depakote ER
  • Zonisamide • Zonegran

Disclosure

Dr. Keck reports that he receives grant/research support from and serves as a consultant to Abbott Laboratories, AstraZeneca, Pfizer Inc., and Eli Lilly and Co. He also receives grant/research support from Merck and Co. and Otsuka America Pharmaceutical, and serves as a consultant to Bristol-Myers Squibb Co., GlaxoSmithKline, and Janssen Pharmaceutica.

Dr. McElroy reports that she receives grant/research support from and serves as a consultant to Abbott Laboratories, Elan Pharmaceuticals, Cephalon Inc. GlaxoSmithKline, and Eli Lilly and Co. She also receives grant/research support from Forest Pharmaceuticals and Solvay Pharmaceuticals, and serves as a consultant to Bristol-Myers Squibb Co., Ortho-McNeil Pharmaceutical, and Janssen Pharmaceutica.

The newer antiepileptic drugs pose a sometimes bewildering range of options for bipolar disorder treatment. Which work best for acute bipolar I mania? Which are best suited for maintenance in patients with mixed episodes, or for those with a history of rapid cycling? What about prevention of depressive episodes? And how do the antiepileptics compare with lithium?

For many patients with bipolar disorder, lithium is still the drug of choice. For others, however, an increasing body of evidence supports the efficacy of some antiepileptics and atypical antipsychotics.

The mood-stabilizing properties of two antiepileptic agents, carbamazepine and valproate, were demonstrated some years ago in randomized controlled trials in patients with bipolar disorder. Since then, there has been considerable interest in the potential thymoleptic properties of the new antiepileptic drugs.1 In recent years gabapentin, lamotrigine, topiramate, oxcarbazepine, zonisamide, tiagabine, and levetiracetam have been approved in the United States for the treatment of various types of epilepsy. These medications have diverse pharmacological properties that distinguish them from earlier agents and from one another.

Do these new agents have anything to offer patients? For the most part, the evidence is not yet in hand, but we will examine what’s available, starting with the most recent trial data regarding the efficacy of valproate and carbamazepine.

Carbamazepine for bipolar mania

Five randomized, controlled trials2 have shown the efficacy of carbamazepine in patients with acute bipolar I mania. Carbamazepine was superior to placebo and comparable to chlorpromazine and lithium. Pooled data reveal an overall response rate (defined as the proportion of patients experiencing > 50% reduction in manic symptoms) of 50% for carbamazepine, 56% for lithium, and 61% for chlorpromazine (differences in overall response rates are not significant).

Until recently, the efficacy of carbamazepine as a maintenance therapy for bipolar disorder was controversial.3 However, two recent large randomized, controlled maintenance studies that compared carbamazepine with lithium validated use of the agent for that purpose.4,5

In the first study, 144 patients received either drug and were followed for up to 2 1/2 years.4 The study showed no significant differences between the two groups in time-to-mood episode recurrence or hospitalization. However, significantly more patients receiving carbamazepine required treatment discontinuation for side effects and additional medications for breakthrough symptoms than did the patients receiving lithium.

Patients without both comorbid disorders and mood-incongruent delusions responded better to lithium, whereas those with mixed episodes and bipolar II disorder or NOS appeared to respond better to carbamazepine.

In the second trial, 52 patients with bipolar I or II disorders received either lithium or carbamazepine for one year, crossed over to the alternate drug the second year, and received both drugs during the third year.5 No significant differences were reported in relapse rates during the first year between lithium (31%) and carbamazepine (37%), or in the percentage of patients who experienced a moderate or better response: 33% on lithium, 31% on carbamazepine, and 55% on the combination. On a variety of other measures, lithium was superior to carbamazepine. But patients with a history of rapid cycling responded significantly better to the combination (56%) than to lithium (28%) or carbamazepine (19%) alone.

As in the previous study, a higher proportion of patients receiving carbamazepine withdrew after experiencing side effects. Both studies found that while carbamazepine is efficacious, lithium is superior overall. But since neither study included a placebo group, carbamazepine’s efficacy in maintenance treatment cannot be determined.

Carbamazepine also was evaluated in three randomized, controlled trials in patients with bipolar depression.2 These small trials suggest that carbamazepine’s antidepressant activity may be less robust than its antimanic effects. Response rates (>50% improvement in depressive symptoms) ranged from 32% to 34%, although many patients who participated had treatment-resistant bipolar depression.

Comparing the known efficacy of antiepileptic agents in bipolar disorder

DrugManiaDepressionMaintenanceComments
Valproate◊◊◊◊◊◊◊New Depakote ER formulation
Carbamazepine◊◊◊◊◊◊◊◊2 new maintenance studies v. lithium
Gabapentin2 negative placebo-controlled studies in mania
Lamotrigine×◊◊◊◊◊◊Antidepressant activity in several controlled trials
TopiramateDose-related weight loss
Oxcarbazepine◊◊Improved tolerability & pharmacokinetics
ZonisamideNDNDMay produce weight loss in some patients
Tiagabine×NDNDMore data needed regarding tolerability and efficacy
LevetiracetamNDNDNDData needed regarding efficacy and tolerability

Key

◊◊◊◊ efficacy demonstrated in ≥2 placebo-controlled trials

◊◊◊ efficacy demonstrated in one placebo-controlled or two large, active comparator trials

◊◊ efficacy in two small or one large active comparator trial

◊ efficacy only in open trials and case series

× conflicting evidence of efficacy in available studies

lack of efficacy demonstrated in randomized, controlled trials

ND no data presently available

Based on the studies reviewed above, carbamazepine is considered a second-line agent for bipolar mania and maintenance treatment with very limited data regarding its antidepressant effects.6

 

 

Valproate: For patients with mixed and mood episodes

The evidence seems to point to valproate as a suitable agent for patients with mixed and mood episodes.

Two earlier randomized, controlled studies7,8 showed that the divalproex sodium formulation of valproate was superior to a placebo, leading to the indication of divalproex for treatment of acute mania in patients with bipolar disorder. Additional analyses of data from the second controlled trial8 indicated that patients with prominent depressive symptoms during mania (mixed episodes) responded better to divalproex than lithium. Further, multiple prior mood episodes were associated with poor lithium response but not with divalproex response.

Two recent randomized, controlled trials compared the antimanic efficacy and tolerability of divalproex and olanzapine.9,10 The design of these two studies differed in two important ways: sample size and starting doses. This may explain the different results of the two trials.

In the first study, 248 patients received starting doses of either divalproex 750 mg/d with upward titration to therapeutic concentrations, or olanzapine 15 mg/d with titration if clinically indicated.9 Olanzapine was found to be superior in the mean reduction of manic symptoms and in the proportion of patients who either responded to treatment or were in remission.

In the second study, the number of patients (N=120) randomized was not sufficient to detect a statistically significant difference between treatment groups.10 Initial dosing consisted of rapid divalproex loading (20 mg/kg/d) versus olanzapine (10 mg/d) with upward titration as clinically indicated. This trial revealed no significant differences in efficacy on any outcome measure and the mean valproic acid plasma concentration was higher than in the first trial.

Differences in side effects between the two agents were similar in both studies. Olanzapine was associated with greater sedation, appetite stimulation, and weight gain, and divalproex with greater gastrointestinal symptoms. Taken together, these two studies indicate that olanzapine is at least as efficacious as divalproex in treating acute mania and that divalproex should be titrated to plasma concentrations well within the therapeutic range consistent with response and tolerability.

The efficacy of divalproex as a maintenance therapy for patients with bipolar disorder has been studied in four trials to date:

  1. A randomized, open comparison of lithium and divalproex found generally good efficacy for both drugs across 18 months.
  2. A second naturalistic, pharmacoeconomic, one-year open comparison also found both lithium and divalproex fairly equal in efficacy.
  3. A large, prospective, randomized one-year maintenance trial showed little difference in relapse rates among patients receiving divalproex (24%), lithium (31%), and a placebo (38%).11
  4. A recently completed one-year comparison of relapse rates between divalproex and olanzapine showed little difference between the two drugs.12

The overall results suggest that divalproex helps prevent mood episodes in patients with bipolar disorder, but the data are less substantial and conclusive than from placebo-controlled trials of lithium.

The antidepressant effects of divalproex in treating acute bipolar depression have not been studied in randomized controlled trials. Impressions from case reports and case series suggest that divalproex may exert some antidepressant activity, but that this action may be less robust than its antimanic effects.

Divalproex is now available in a once-daily extended release (ER) formulation. This appears to have improved tolerability, especially regarding gastrointestinal side effects. (Clinical experience suggests that the immediate release formulation of divalproex can also be given once daily.) The ER formulation is not bioequivalent to its immediate-release counterpart; it produces plasma concentrations of approximately 80% of those achieved with immediate release. Thus, switching a patient to the ER formulation might require a dose increase.

Lamotrigine for bipolar depression

Several recent randomized controlled trials indicate that lamotrigine has important thymoleptic properties.14-17

Three studies addressed the efficacy of lamotrigine in treating patients with acute bipolar mania. In two small trials, lamotrigine did not display superior efficacy over placebo in reducing manic symptoms.14 A third trial revealed differences between lithium and lamotrigine in reducing manic symptoms, but this study lacked sufficient power to detect potential differences in efficacy.

Two placebo-controlled maintenance studies of lamotrigine were recently reported.15,16 The first found no significant differences in relapse rates in patients with rapid cycling bipolar I and II disorders randomized to lamotrigine or placebo after initial stabilization on lamotrigine.15 However, in a post hoc analysis among bipolar II (but not bipolar I) patients, lamotrigine was significantly more efficacious than placebo in time-to-study dropout and considerably better (P=0.07) in time to need for additional medication.

In the second randomized, placebo-controlled trial, patients with bipolar I disorder who had recently experienced a manic episode were treated with lamotrigine 100-200 mg/d during an open-label phase (8-16 weeks) while other psychotropic agents were tapered and discontinued.16 Patients (N = 171) who remained stable were then randomized to lamotrigine 200-400 mg/d, lithium, or placebo for up to 18 months.

 

 

The lamotrigine-treated group showed significantly lower relapse rates than placebo-treated patients in time-to-study dropout, time to intervention for a mood episode, and time to intervention for depressive relapse. Differences between the lamotrigine and placebo groups in time to intervention for manic relapse were insignificant. Lithium was superior to a placebo in time to relapse for any mood episode and time to intervention for a manic episode. Lithium also outperformed lamotrigine in that manic symptoms did not worsen as quickly from baseline to endpoint.

The results here are consistent with those of the rapid cycling study: Lamotrigine helps prevent recurrence of depressive symptoms and episodes. Evidence of prophylaxis against manic recurrences was not compelling, however.

The findings of these two maintenance trials are consistent with the results of a placebo-controlled, randomized, 6-week acute treatment trial of lamotrigine (50 mg/d and 200 mg/d) in patients with bipolar depression.17 Seventeen lamotrigine-treated patients in both dosage groups saw more-significantly reduced depressive symptoms than did placebo-treated patients on the MADRS (but not the HAMD) total score and on the CGI. The 200 mg/d group tended to have greater improvement than the 50 mg/d group. There was little difference among the three treatment groups in the incidence of switching into hypomania or mania.

These studies indicate that lamotrigine has acute and prophylactic efficacy against bipolar depression. In contrast, evidence of the agent’s acute or prophylactic efficacy in mania is lacking.

Other new antiepileptics: More testing needed

Use of the six other newer antiepileptics in patients with bipolar disorder is still being explored. (See “Comparing the known efficacy of antiepileptic agents in bipolar disorder,”) Let’s look at what we know about these agents to this point.

Gabapentin A number of case reports and case series published in recent years suggest that gabapentin may have mood-stabilizing properties. In two randomized, controlled trials, however, gabapentin did not display significantly greater efficacy than a placebo in treating acute mania.13,14 Gabapentin has not been studied in controlled trials as a maintenance treatment or for bipolar depression.

In contrast to these negative studies, gabapentin was superior to placebo in studies of patients with panic disorder, social anxiety disorder, and neuropathic pain. Overall, these studies suggest that gabapentin is not a bona fide mood stabilizer for most patients.

Topiramate More than 10 case reports and case series suggest that topiramate may have mood-stabilizing properties. A number of open trials also have found significant dose-related weight loss in patients with weight gain associated with other psychotropic agents.18 These observations have sparked interest in topiramate’s potential role as an obesity treatment.

Only one randomized, controlled trial of topiramate for bipolar disorder has appeared to date.19 An interim analysis of a placebo-controlled, randomized trial of two doses of topiramate (approximately 250 mg/d and 500 mg/d) in 97 hospitalized patients with acute bipolar I mania revealed strong trends toward reduced manic symptoms for both topiramate groups over placebo. These differences were not significant by the time the study was concluded, however.

CHARACTERISTICS OF THE NEWER ANTIEPILEPTICS

Lamotrigine is a novel drug that blocks voltage-sensitive sodium channels, thereby indirectly inhibiting the release of excitatory neurotransmitters, particularly glutamate and aspartate.

Gabapentin was developed to mimic the synaptic effects of gamma-aminobutyric acid (GABA); it does not appear to appreciably interact with GABA receptors, however, and its mechanism of action in epilepsy remains unknown. It has several attractive pharmacokinetic properties, including lack of protein binding, renal clearance rather than hepatic metabolism, and few known drug-drug interactions.

Topiramate is a sulfamate-substituted monosaccharide with a number of possible mechanisms of action, including blockade of voltage-gated sodium channels, antagonism of the kainate/AMPA glutamate receptor subtype, enhancement of GABA activity at the GABA A receptor via interaction with a nonbenzodiazepine receptor site, and carbonic anhydrase inhibition.

Oxcarbazepine is the 10-keto analogue of carbamazepine, a chemical difference that translates into a number of safety advantages over carbamazepine. Oxcarbazepine is converted to an active 10-hydroxy metabolite rather than to the 10,11-epoxide metabolite of carbamazepine. The 10,11-epoxide metabolite of carbamazepine is associated with neurological side effects. Oxcarbazepine is a weak inducer of the CYP450 system, appears to have fewer drug-drug interactions, and offers better overall tolerability.

Zonisamide, a sulfonamide derivative, blocks voltage-sensitive sodium channels and T-type calcium currents, modulates GABAergic and dopaminergic neurotransmission, and is a free-radical scavenger.

Tiagabine is a selective GABA reuptake inhibitor.

Levetiracetam is the S-enantiomer of the ethyl analogue of the nootropic agent piracetam. Its mechanism of action in treating epilepsy is unknown. It does not appear to interact significantly with voltage-sensitive sodium channels or T-type calcium channels, nor does it significantly alter levels of GABA, glutamate, or glutamine in the central nervous system.

 

 

When patients with mania associated with recent antidepressant use were excluded from post hocanalysis, topiramate’s superiority over placebo was re-established. These tantalizing results require further study. Similarly, studies of topiramate’s potential efficacy in acute bipolar depression and as a maintenance treatment are needed.

Oxcarbazepine Oxcarbazepine is a relative newcomer to the United States, but has been available abroad for some time.

While numerous studies have tested carbamazepine in treating various aspects of bipolar disorder, few controlled trials have tested oxcarbazepine for this use.20

Oxcarbazepine as a treatment for acute bipolar mania was comparable to haloperidol in two small randomized, controlled trials and to lithium in one small trial. None of these studies had adequate power to detect potential differences in efficacy. Similarly, two very small maintenance studies (total N=25) found no differences in efficacy between oxcarbazepine and lithium.20

Additionally, to our knowledge oxcarbazepine has not been formally tested as a treatment for acute bipolar depression. So while oxcarbazepine is an attractive alternative to carbamazepine for pharmacokinetic and pharmacodynamic reasons, data supporting oxcarbazepine’s efficacy in bipolar disorder are far less substantial.

Zonisamide The only report of zonisamide as a bipolar disorder treatment so far is a case series of 15 patients who received the drug adjunctively for manic symptoms. Of these patients, 80% showed at least moderate improvement and 33% were rated as markedly improved. These preliminary observations require further study. Like topiramate, zonisamide may also be associated with weight loss in some patients.

Tiagabine A few case reports and case series describe tiagabine’s effects in patients with bipolar disorder. Conflicting results have been reported to date and some reports of poor tolerability have emerged. Fundamental safety and efficacy data for this agent in bipolar disorder are needed.

Levetiracetam Studies of levetiracetam for treatment of bipolar disorder are just under way. The Stanley Foundation Bipolar Treatment Network, a program of the National Alliance for the Mentally Ill Research Institute, is investigating its potential thymoleptic properties.

Related resources

  • Joffe RT, Calabrese JR. Anticonvulsants in Mood Disorders. New York: Marcel Dekker Inc., 1994.
  • Modigh K, Robak OH, Vestergaard P. Anticonvulsants in Psychiatry. Bristol, Pa: Wrightson Biomedical Publishing, Ltd., 1994.
  • Schatzberg AF, Nemeroff CB. Textbook of Psychopharmacology, 2 nd ed. Washington, DC: American Psychiatric Press, 1998.
  • Nathan P, Gorman J. A Guide to Treatments that Work, 2 nd ed. New York: Oxford University Press, 2001.
  • Janicak PG, Davis JM, Preskorn SH, Ayd FJ, Jr. Principles and Practice of Psychopharmacotherapy, 2 nd ed. Baltimore, MD: Williams & Wilkins, 1997.

Drug brand names

  • Carbamazepine • Tegretol, Epitol, Atretol
  • Gabapentin • Neurontin
  • Lamotrigine • Lamictal
  • Levetiracetam • Keppra
  • Oxcarbazepine • Trileptal
  • Tiagabine • Gabatril
  • Topiramate • Topamax
  • Valproate-divalproex sodium • Depakote, Depakote ER
  • Zonisamide • Zonegran

Disclosure

Dr. Keck reports that he receives grant/research support from and serves as a consultant to Abbott Laboratories, AstraZeneca, Pfizer Inc., and Eli Lilly and Co. He also receives grant/research support from Merck and Co. and Otsuka America Pharmaceutical, and serves as a consultant to Bristol-Myers Squibb Co., GlaxoSmithKline, and Janssen Pharmaceutica.

Dr. McElroy reports that she receives grant/research support from and serves as a consultant to Abbott Laboratories, Elan Pharmaceuticals, Cephalon Inc. GlaxoSmithKline, and Eli Lilly and Co. She also receives grant/research support from Forest Pharmaceuticals and Solvay Pharmaceuticals, and serves as a consultant to Bristol-Myers Squibb Co., Ortho-McNeil Pharmaceutical, and Janssen Pharmaceutica.

References

1. Keck PE, Jr, McElroy SL, Strakowski SM. Anticonvulsants and antipsychotics in the treatment of bipolar disorder. J Clin Psychiatry. 1998;59(Suppl 6):74-81.

2. McElroy SL, Keck PE, Jr. Pharmacologic agents for the treatment of acute bipolar mania. Biol Psychiatry. 2000;48:539-557.

3. Post RM, Denicoff KD, Frye MA, Leverich GS. Re-evaluating carbamazepine prophylaxis in bipolar disorder. Br J Psychiatry. 1997;170:202-204.

4. Greil W, Ludwig-Mayerhofer W, Erazon-Scheichlin C, Schmidt S, Engel RR, et al. Lithium versus carbamazepine in the maintenance treatment of bipolar disorders–a randomized study. J Affect Disorders. 1997;43:151-161.

5. Denicoff KD, Smith-Jackson EE, Disney ER, Ali SO, Leverich GS, Post RM. Comparative prophylactic efficacy of lithium, carbamazepine and the combination in bipolar disorder. J Clin Psychiatry. 1997;58:470-478.

6. Suppes T, Swann AC, Dennehy EB, Habermacher ED, Mason M, Crismon ML, Toprac MG, Rush AJ, Shon SP, Altshuler KZ. Texas Medication Algorithm Project: development and feasibility testing of a treatment algorithm for patients with bipolar disorder. J Clin Psychiatry. 2001;62:439-447.

7. Pope HG, Jr, McElroy SL, Keck PE, Jr, Hudson JI. Valproate in the treatment of acute mania: a placebo-controlled study. Arch Gen Psychiatry. 1991;48:62-68.

8. Bowden CL, Brugger AM, Swann AC, Calabrese JR, Janicak PG, Petty F, et al. Efficacy of divalproex sodium vs. lithium and placebo in the treatment of mania. JAMA. 1994;271:915-924.

9. Tohen M, Baker RW, Milton DR, Risser RC, Gilmore JA, Davis AR, et al. Olanzapine versus divalproex sodium for the treatment of acute mania. Bipolar Disorders. 2001;3(Suppl 1):60-61.

10. Zajecka J. Divalproex versus olanzapine in the treatment of acute mania. Presented at the 39 th Annual Meeting of the American College of Neuropsychopharmacology. San Juan, Puerto Rico, Dec. 10-14, 2000.

11. Bowden CL, Calabrese JR, McElroy SL, Hirschfeld RMA, Petty F, Gyulai LL, Pope HG, Jr, et al. Efficacy of divalproex versus lithium and placebo in maintenance treatment of bipolar disorder. Arch Gen Psychiatry. 2000;57:481-489.

12. Tohen M, Baker RW, Altshuler LL, Zarate CA, Suppes T, Ketter T, et al. Olanzapine versus divalproex for bipolar mania: a 47-week study. Presented at the European College of Neuropsychopharmacology Meeting, Istanbul, Turkey, Oct. 15, 2001.

13. Pande AD, Crockatt JG, Janney CA, Werth JL, Tsaaroucha G. and the Gabapentin Bipolar Disorder Study Group. Bipolar Disorders. 2000;2:249-255.

14. Frye MA, Ketter TA, Kimbrell TA, et al. A placebo controlled study of lamotrigine and gabapentin monotherapy in refractory mood disorders. J Clin Psychopharmacol. 2000;24:205-212.

15. Calabrese JR, Suppes T, Bowden CL, et al. A double-blind, placebo-controlled study of lamotrigine monotherapy in rapid-cycling bipolar disorder. J Clin Psychiatry. 2000;61:841-850.

16. Bowden CL, Calabrese JR, Akthar S, Olajossy M, Montgomery S, Ascher J, et al. Lamotrigine demonstrates long-term mood stabilization in manic patients. Bipolar Disorders. 2001;3(Suppl 1):27-28.

17. Calabrese JR, Bowden CL, Sachs GS, et al. A double-blind, placebo-controlled study of lamotrigine monotherapy in outpatients with bipolar I depression. J Clin Psychiatry. 1999;60:79-88.

18. McElroy SL, Suppes T, Keck PE, Jr, Frye MA, Denicoff KD, Altshuler LL, et al. Open-label adjunctive topiramate in the treatment of bipolar disorders. Biol Psychiatry. 2000;47:1025-1033.

19. Calabrese JR. A double-blind, placebo-controlled trial of topiramate monotherapy in the treatment of acute mania. Presented at the Annual Meeting of the European College of Neuropsychopharmacology. Munich, Germany, Sept. 9-13, 2000.

20. Emrich H, Schiwy W, Silverstone T. eds. Carbamazepine and Oxcarbazepine in Psychiatry. London: CNS Publishers, 1990.

References

1. Keck PE, Jr, McElroy SL, Strakowski SM. Anticonvulsants and antipsychotics in the treatment of bipolar disorder. J Clin Psychiatry. 1998;59(Suppl 6):74-81.

2. McElroy SL, Keck PE, Jr. Pharmacologic agents for the treatment of acute bipolar mania. Biol Psychiatry. 2000;48:539-557.

3. Post RM, Denicoff KD, Frye MA, Leverich GS. Re-evaluating carbamazepine prophylaxis in bipolar disorder. Br J Psychiatry. 1997;170:202-204.

4. Greil W, Ludwig-Mayerhofer W, Erazon-Scheichlin C, Schmidt S, Engel RR, et al. Lithium versus carbamazepine in the maintenance treatment of bipolar disorders–a randomized study. J Affect Disorders. 1997;43:151-161.

5. Denicoff KD, Smith-Jackson EE, Disney ER, Ali SO, Leverich GS, Post RM. Comparative prophylactic efficacy of lithium, carbamazepine and the combination in bipolar disorder. J Clin Psychiatry. 1997;58:470-478.

6. Suppes T, Swann AC, Dennehy EB, Habermacher ED, Mason M, Crismon ML, Toprac MG, Rush AJ, Shon SP, Altshuler KZ. Texas Medication Algorithm Project: development and feasibility testing of a treatment algorithm for patients with bipolar disorder. J Clin Psychiatry. 2001;62:439-447.

7. Pope HG, Jr, McElroy SL, Keck PE, Jr, Hudson JI. Valproate in the treatment of acute mania: a placebo-controlled study. Arch Gen Psychiatry. 1991;48:62-68.

8. Bowden CL, Brugger AM, Swann AC, Calabrese JR, Janicak PG, Petty F, et al. Efficacy of divalproex sodium vs. lithium and placebo in the treatment of mania. JAMA. 1994;271:915-924.

9. Tohen M, Baker RW, Milton DR, Risser RC, Gilmore JA, Davis AR, et al. Olanzapine versus divalproex sodium for the treatment of acute mania. Bipolar Disorders. 2001;3(Suppl 1):60-61.

10. Zajecka J. Divalproex versus olanzapine in the treatment of acute mania. Presented at the 39 th Annual Meeting of the American College of Neuropsychopharmacology. San Juan, Puerto Rico, Dec. 10-14, 2000.

11. Bowden CL, Calabrese JR, McElroy SL, Hirschfeld RMA, Petty F, Gyulai LL, Pope HG, Jr, et al. Efficacy of divalproex versus lithium and placebo in maintenance treatment of bipolar disorder. Arch Gen Psychiatry. 2000;57:481-489.

12. Tohen M, Baker RW, Altshuler LL, Zarate CA, Suppes T, Ketter T, et al. Olanzapine versus divalproex for bipolar mania: a 47-week study. Presented at the European College of Neuropsychopharmacology Meeting, Istanbul, Turkey, Oct. 15, 2001.

13. Pande AD, Crockatt JG, Janney CA, Werth JL, Tsaaroucha G. and the Gabapentin Bipolar Disorder Study Group. Bipolar Disorders. 2000;2:249-255.

14. Frye MA, Ketter TA, Kimbrell TA, et al. A placebo controlled study of lamotrigine and gabapentin monotherapy in refractory mood disorders. J Clin Psychopharmacol. 2000;24:205-212.

15. Calabrese JR, Suppes T, Bowden CL, et al. A double-blind, placebo-controlled study of lamotrigine monotherapy in rapid-cycling bipolar disorder. J Clin Psychiatry. 2000;61:841-850.

16. Bowden CL, Calabrese JR, Akthar S, Olajossy M, Montgomery S, Ascher J, et al. Lamotrigine demonstrates long-term mood stabilization in manic patients. Bipolar Disorders. 2001;3(Suppl 1):27-28.

17. Calabrese JR, Bowden CL, Sachs GS, et al. A double-blind, placebo-controlled study of lamotrigine monotherapy in outpatients with bipolar I depression. J Clin Psychiatry. 1999;60:79-88.

18. McElroy SL, Suppes T, Keck PE, Jr, Frye MA, Denicoff KD, Altshuler LL, et al. Open-label adjunctive topiramate in the treatment of bipolar disorders. Biol Psychiatry. 2000;47:1025-1033.

19. Calabrese JR. A double-blind, placebo-controlled trial of topiramate monotherapy in the treatment of acute mania. Presented at the Annual Meeting of the European College of Neuropsychopharmacology. Munich, Germany, Sept. 9-13, 2000.

20. Emrich H, Schiwy W, Silverstone T. eds. Carbamazepine and Oxcarbazepine in Psychiatry. London: CNS Publishers, 1990.

Issue
Current Psychiatry - 01(01)
Issue
Current Psychiatry - 01(01)
Page Number
18-24
Page Number
18-24
Publications
Publications
Topics
Article Type
Display Headline
Antiepileptic drugs for bipolar disorder: Are there any clear winners?
Display Headline
Antiepileptic drugs for bipolar disorder: Are there any clear winners?
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media