User login
Schizophrenia, but not bipolar disorder, linked to social cognition deficits
Significant deficits in social cognition were found in patients with schizophrenia spectrum disorders (SSDs), but not in patients with bipolar disorder (BD), Dr. George C. Nitzburg and his associates reported.
In a study of 537 SSD patients, 85 BD patients with psychotic features, 37 BD patients without psychotic features, and 309 controls, SSD patients had significant social cognition deficits, compared with controls. Bipolar patients did not have these deficits, the investigators reported (F (2,964) = 24.85, P < .001). Social cognition was assessed using scores on the Mayer-Salovey-Caruso Emotional Intelligence Test (MSCEIT).
The results of this study “highlight the importance of developing standardized social-cognitive batteries for use across BD and SSD and emphasize the need for future work on social brain development in clinical populations,” Dr. Nitzburg and his coauthors wrote in the report.
Read the full article here: doi:10.1016/j.scog.2014.12.003.
Significant deficits in social cognition were found in patients with schizophrenia spectrum disorders (SSDs), but not in patients with bipolar disorder (BD), Dr. George C. Nitzburg and his associates reported.
In a study of 537 SSD patients, 85 BD patients with psychotic features, 37 BD patients without psychotic features, and 309 controls, SSD patients had significant social cognition deficits, compared with controls. Bipolar patients did not have these deficits, the investigators reported (F (2,964) = 24.85, P < .001). Social cognition was assessed using scores on the Mayer-Salovey-Caruso Emotional Intelligence Test (MSCEIT).
The results of this study “highlight the importance of developing standardized social-cognitive batteries for use across BD and SSD and emphasize the need for future work on social brain development in clinical populations,” Dr. Nitzburg and his coauthors wrote in the report.
Read the full article here: doi:10.1016/j.scog.2014.12.003.
Significant deficits in social cognition were found in patients with schizophrenia spectrum disorders (SSDs), but not in patients with bipolar disorder (BD), Dr. George C. Nitzburg and his associates reported.
In a study of 537 SSD patients, 85 BD patients with psychotic features, 37 BD patients without psychotic features, and 309 controls, SSD patients had significant social cognition deficits, compared with controls. Bipolar patients did not have these deficits, the investigators reported (F (2,964) = 24.85, P < .001). Social cognition was assessed using scores on the Mayer-Salovey-Caruso Emotional Intelligence Test (MSCEIT).
The results of this study “highlight the importance of developing standardized social-cognitive batteries for use across BD and SSD and emphasize the need for future work on social brain development in clinical populations,” Dr. Nitzburg and his coauthors wrote in the report.
Read the full article here: doi:10.1016/j.scog.2014.12.003.
Delusional and aggressive, while playing the lottery
CASE Delusional and aggressive
Mr. P, age 78, of Filipino heritage, is brought to the psychiatric hospital because he has been verbally aggressive toward his wife for several weeks. He has no history of a psychiatric diagnosis or inpatient psychiatric hospitalization, and no history of taking any psychotropic medications.
According to his wife, Mr. P has been ruminating about his father, who died in World War II, saying that “the Japanese never gave his body back” to him. Also, his wife describes 3 weeks of physically aggressive behavior, such as throwing punches; the last episode was 2 days before admission.
Mr. P is not bathing, eating, taking his medications, and attending to his activities of daily living. He sleeps for only 1 to 2 hours a night; is irritable and easily distractible; and experiences flight of ideas. Mr. P has been buying lottery tickets, telling his daughter that he will become a millionaire and then buy a house in the Philippines.
Mr. P reports depressed mood, but no other depressive symptoms are present. He reports no suicidal or homicidal ideations, auditory or visual hallucinations, or anxiety symptoms. He has no history of substance abuse.
What diagnosis would you give Mr. P?
a) late-onset bipolar disorder
b) Alzheimer’s disease
c) major depressive disorder
d) frontotemporal dementia
The authors’ observations
Bipolar disorder in later life is a complex and confounding neuropsychiatric syndrome with diagnostic and therapeutic challenges. The disorder can affect people of all ages and is not uncommon among geriatric patients, with a 1-year prevalence in United States of 0.4%.1 In one study, 10% of new bipolar disorder cases were found to occur after age 50.2 As the American population grows older, the number of bipolar disorder cases among seniors is expected to increase.3
It was once thought that symptoms of bipolar disorder disappear with age; newer research has disproved this theory, and proposes that untreated bipolar disorder worsens over time.4 Persons who are given the diagnosis later in life could have had bipolar disorder for decades, but symptoms became more noticeable and problematic with age.5
Common symptoms in geriatric patients can differ from what we might expect in younger patients: agitation, hyperactivity, irritability, confusion, and psychosis.6 When the disorder presents in patients age >60, it can be severe, with significant changes in cognitive function, including difficulties with memory, perception, judgment, and problem-solving.7,8
HISTORY Medical comorbidities
Mr. P emigrated from the Philippines 20 years ago, is married, and lives with his wife. He has 3 brothers; his parents were divorced, and his mother remarried. Mr. P completed high school.
Mr. P has an extensive medical history: diabetes mellitus, hypertension, dyslipidemia, and recent double coronary artery bypass grafting. He is taking several medications: sitagliptin, 25 mg/d; pantoprazole, 5 mg/d; metformin, 1,000 mg/d; rivaroxaban, 20 mg/d; amiodarone, 200 mg/d; metoprolol, 12.5 mg/d; olmesartan medoxomil, 40 mg/d; aspirin, 81 mg/d; simvastatin, 10 mg/d; eszopiclone, 3 mg at bedtime; and amlodipine, 5 mg at bedtime.
Mr. P was following up with his primary care physician for his medical conditions and was adherent with treatment until 1 week before he was admitted to our facility.
The authors’ observations
Always rule out medical causes in a case of new-onset mania, which is particularly important in geriatric patients. Older patients with new-onset mania are more than twice as likely to have a comorbid neurologic disorder.9 Neurologic causes of late-onset mania include:
• stroke
• tumor
• epilepsy
• Huntington’s disease and other movement disorders
• multiple sclerosis and other white-matter diseases
• head trauma
• infection (such as neurosyphilis)
• Creutzfeldt-Jakob disease
• frontotemporal dementia.10
Mr. P’s presentation of psychomotor agitation, impaired functioning, decreased need for sleep, increased energy, hyperverbal speech, and complex paranoid delusions meets DSM-5 criteria for bipolar disorder, manic phase. In addition, older manic patients frequently present with confusion, disorientation, and distractibility. Younger patients with mania often present with euphoric moods and grandiosity; in contrast, geriatric patients are more likely to show a mixture of depressed affect and manic symptoms (pressured speech and a decreased need for sleep).11-15
We considered an emerging neurodegenerative process, because dementia can present early with disinhibition, lability, and other behavioral disturbances, including classic manic syndromes.16 Although we could not fully rule out a neurodegenerative process in the initial phase of treatment, Mr. P’s longitudinal course demonstrated no change in baseline cognitive function and no evidence of subsequent decline, making dementia unlikely.17
Patients with frontotemporal dementia are more likely to present initially to a psychiatrist than to a neurologist.18
Frontotemporal dementia is a progressive neurodegenerative disease that affects the frontal and temporal cortices; it is a common cause of dementia in patients age <65.19 Frontotemporal dementia is characterized by insidious behavioral and personality changes; often, the initial presentation lacks any clear neurologic signs or symptoms. Key features include apathy, disinhibition, loss of sympathy and empathy, repetitive motor behaviors, and overeating.20
Mr. P’s symptoms stabilized with divalproex sprinkles and risperidone. There was no evidence of decline in memory, social interaction, or behavior.
EVALUATION Paranoia
On mental status exam, Mr. P has an appropriate appearance; he is clean and shaven, with good eye contact. Muscular tone and gait are within normal limits. Level of activity is increased; he exhibits psychomotor agitation. Speech is rapid, over-productive, and loud; thought process shows flight of ideas, and thought associations are circumstantial.
Mr. P has paranoid delusions about the staff trying to hurt him. His judgment is poor, evidenced by an inability to take care of himself. Insight is minimal, as seen by noncompliance with treatment. Mr. P is oriented only to person and place. His mood is anxious; affect is labile.
Complete blood count, comprehensive metabolic profile, blood alcohol level, urine analysis, urine toxicology, electrocardiogram, and CT scan of the head are within normal limits.
Mr. P is given a diagnosis of mood disorder due to general medical condition, psychotic disorder due to general medical condition. The team rules out acute delirium, bipolar I disorder, and neurodegenerative disorders such as frontotemporal dementia.
Mr. P is maintained on pre-admission medications for his medical conditions. A mood stabilizer, divalproex sprinkles, 250 mg/d, is added.
Once on the unit, Mr. P is re-evaluated. Divalproex is increased to 500 mg/d; risperidone, 0.5 mg/d, is added to address paranoia. Mr. P also receives group and individual psychotherapy. He does not participate in neuropsychological testing, and no single-photon emission CT analysis is done. Mr. P remains in the hospital for 2 weeks. After a family meeting, his daughter says she feels comfortable taking Mr. P home. He follows up in the outpatient clinic and is doing well.
The authors’ observations
Treating geriatric patients with bipolar disorder requires attention to several factors (Table). Older patients might tolerate or metabolize medications differently than younger adults, and therefore may need a different dosage. Older patients are more likely to have comorbid medical conditions and to be taking medications for those ailments. Treatment is much more complicated for this age group because physicians need to account for possible drug-drug interactions.21
A number of medications can be helpful in treating older patients who have bipolar disorder.11 Ongoing research compares lithium with anticonvulsants in older bipolar disorder patients to determine which drug has the greatest benefit with the lowest risk of side effects.
Psychotherapy can be a valuable addition to pharmacotherapy in older adults. Some psychotherapy programs are specifically geared to older bipolar disorder patients.22,23
Use of divalproex sodium in older patients
First, perform baseline laboratory tests: complete blood count, liver function, and electrocardiogram. Initiate divalproex sodium, 250 mg at bedtime, increasing the dosage every 3 to 5 days by 250 mg, with a target dose of 500 to 2,000 mg/d (divided into 2 or 3 doses). Monitor serum levels; levels of 29 to 100 μg/mL are effective and well tolerated. Common side effects include excess sedation, ataxia, tremor, nausea, and, rarely, hepatotoxicity, leukopenia, and thrombocytopenia.24
Use of lithium in geriatric patients
First, perform baseline laboratory tests: electrolytes, creatinine, blood urea nitrogen, urine, thyroid stimulating hormone, and electrocardiogram. Starting dosage is 300 mg at bedtime (150 mg for frail cachectic patients). Monitor serum levels 12 hours after last dose, adjusting dosage every 5 days until a target serum level of 0.5 to 0.8 mEq/L is reached. Common dosages for geriatric patients are 300 to 600 mg/d, which often can be given as a single bedtime dose. Cautions: When using lithium with a thiazide diuretic or nonsteroidal anti-inflammatory drug, watch for dehydration, vomiting, and diarrhea, which will elevate the serum lithium level. Side effects include ataxia, tremor, urinary frequency, thirst, nausea, diarrhea, hypothyroidism, and exacerbation of psoriasis. Once stabilized, monitor the serum lithium level, thyroid-stimulating hormone, and kidney function every 3 to 6 months.24
Bottom Line
In geriatric patients, bipolar disorder can present with agitation, irritability, confusion, and psychosis, rather than euphoric mood and grandiosity. When you suspect bipolar disorder in an older patient, first rule out medical causes of symptoms. When selecting treatment, consider comorbid medical conditions and possible drug-drug interactions.
Related Resources
• Sajatovic M, Forester BP, Gildengers A, et al. Aging changes and medical complexity in late-life bipolar disorder: emerging research findings that may help advance care. Neuropsychiatry (London). 2013;3(6):621-633.
• Dols A, Rhebergen D, Beekman A, et al. Psychiatric and medical comorbidities: results from a bipolar elderly cohort study. Am J Geriatr Psychiatry. 2014;22(11):1066-1074.
Drug Brand Names
Amiodarone • Cordarone Olanzapine • Zyprexa
Amlodipine • Norvasc Olmesartan medoxomil • Benicar
Divalproex sodium • Depakote Pantoprazole • Protonix
Eszopiclone • Lunesta Risperidone • Risperdal
Lithium • Eskalith, Lithobid Rivaroxaban • Xarelto
Lorazepam • Ativan Simvastatin • Zocor
Metformin • Glucophage Sitagliptin • Januvia
Metoprolol • Lopressor
Disclosures
The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Weissman MM, Leaf PJ, Tischler GL, et al. Affective disorders in five United States communities. Psychol Med. 1988;18(1):141-153.
2. Yassa R, Nair NP, Iskandar H. Late-onset bipolar disorder. Psychiatr Clin North Am. 1988;11(1):117-131.
3. Verdoux H, Bourgeois M. Secondary mania caused by cerebral organic pathology [in French]. Ann Med Psychol (Paris). 1995;153(3):161-168.
4. Fadden G, Bebbington P, Kuipers L. The burden of care: the impact of functional psychiatric illness in the patient’s family. Br J Psychiatry. 1987;150:285-292.
5. Yassa R, Nair V, Nastase C, et al. Prevalence of bipolar disorder in a psychogeriatric population. J Affect Disord. 1988;14(3):197-201.
6. Robinson RG, Boston JD, Starkstein SE, et al. Comparison of mania with depression following brain injury: casual factors. Am J Psychiatry. 1988;145(2):172-178.
7. Starkstein SE, Boston JD, Robinson RG. Mechanisms of mania after brain injury: 12 case reports and review of the literature. J Nerv Ment Dis. 1988;176(2):87-100.
8. Herrmann N, Bremner KE, Naranjo CA. Pharmacotherapy of late life mood disorders. Clin Neurosci. 1997;4(1):41-47.
9. Tohen M, Shulman KI, Satlin A. First-episode mania in late life. Am J Psychiatry. 1994;151(1):130-132.
10. Mendez MF. Mania in neurologic disorders. Curr Psychiatry Rep. 2000;2(5):440-445.
11. Eagles JM, Whalley LJ. Aging and affective disorders: the age at first onset of affective disorders in Scotland, 1969- 1978. Br J Psychiatry. 1985;147:180-187.
12. Snowdon J. A retrospective case-note study of bipolar disorder in old age. Br J Psychiatry. 1991;158:485-490.
13. Winokur G. The Iowa 500: heterogeneity and course in manic-depressive illness (bipolar). Compr Psychiatry. 1975;16(2):125-131.
14. Shulman K, Post F. Bipolar affective disorder in old age. Br J Psychiatry. 1980;136:26-32.
15. Young RC, Falk JR. Age, manic psychopathology, and treatment response. Int J Geriatr Psychiatry. 1989;4(2):73-78.
16. Almeida OP. Bipolar disorder with late onset: an organic variety of mood disorder [in Portuguese]? Rev Bras Psiquiatr. 2004;26(suppl 3):27-30.
17. Carlino AR, Stinnett JL, Kim DR. New onset of bipolar disorder in late life. Psychosomatics. 2013;54(1):94-97.
18. Woolley JD, Wilson MR, Hung E, et al. Frontotemporal dementia and mania. Am J Psychiatry. 2007;164(12):1811-1816.
19. Ratnavalli E, Brayne C, Dawson K, et al. The prevalence of frontotemporal dementia. Neurology. 2002;58(11):1615-1621.
20. Gregory CA, Hodges JR. Clinical features of frontal lobe dementia in comparison to Alzheimer’s disease. J Neural Transm Suppl. 1996;47:103-123.
21. Broadhead J, Jacoby R. Mania in old age: a first prospective study. Int J Geriatr Psychiatry. 1990;5(4):215-222.
22. Dhingra U, Rabins PV. Mania in the elderly: a 5-7 year follow-up. J Am Geriatr Soc. 1991;39(6):581-583.
23. Shulman KI. Neurologic comorbidity and mania in old age. Clin Neurosci. 1997;4(1):37-40.
24. Shulman KI, Herrmann N. Bipolar disorder in old age. Can Fam Physician. 1999;45:1229-1237.
CASE Delusional and aggressive
Mr. P, age 78, of Filipino heritage, is brought to the psychiatric hospital because he has been verbally aggressive toward his wife for several weeks. He has no history of a psychiatric diagnosis or inpatient psychiatric hospitalization, and no history of taking any psychotropic medications.
According to his wife, Mr. P has been ruminating about his father, who died in World War II, saying that “the Japanese never gave his body back” to him. Also, his wife describes 3 weeks of physically aggressive behavior, such as throwing punches; the last episode was 2 days before admission.
Mr. P is not bathing, eating, taking his medications, and attending to his activities of daily living. He sleeps for only 1 to 2 hours a night; is irritable and easily distractible; and experiences flight of ideas. Mr. P has been buying lottery tickets, telling his daughter that he will become a millionaire and then buy a house in the Philippines.
Mr. P reports depressed mood, but no other depressive symptoms are present. He reports no suicidal or homicidal ideations, auditory or visual hallucinations, or anxiety symptoms. He has no history of substance abuse.
What diagnosis would you give Mr. P?
a) late-onset bipolar disorder
b) Alzheimer’s disease
c) major depressive disorder
d) frontotemporal dementia
The authors’ observations
Bipolar disorder in later life is a complex and confounding neuropsychiatric syndrome with diagnostic and therapeutic challenges. The disorder can affect people of all ages and is not uncommon among geriatric patients, with a 1-year prevalence in United States of 0.4%.1 In one study, 10% of new bipolar disorder cases were found to occur after age 50.2 As the American population grows older, the number of bipolar disorder cases among seniors is expected to increase.3
It was once thought that symptoms of bipolar disorder disappear with age; newer research has disproved this theory, and proposes that untreated bipolar disorder worsens over time.4 Persons who are given the diagnosis later in life could have had bipolar disorder for decades, but symptoms became more noticeable and problematic with age.5
Common symptoms in geriatric patients can differ from what we might expect in younger patients: agitation, hyperactivity, irritability, confusion, and psychosis.6 When the disorder presents in patients age >60, it can be severe, with significant changes in cognitive function, including difficulties with memory, perception, judgment, and problem-solving.7,8
HISTORY Medical comorbidities
Mr. P emigrated from the Philippines 20 years ago, is married, and lives with his wife. He has 3 brothers; his parents were divorced, and his mother remarried. Mr. P completed high school.
Mr. P has an extensive medical history: diabetes mellitus, hypertension, dyslipidemia, and recent double coronary artery bypass grafting. He is taking several medications: sitagliptin, 25 mg/d; pantoprazole, 5 mg/d; metformin, 1,000 mg/d; rivaroxaban, 20 mg/d; amiodarone, 200 mg/d; metoprolol, 12.5 mg/d; olmesartan medoxomil, 40 mg/d; aspirin, 81 mg/d; simvastatin, 10 mg/d; eszopiclone, 3 mg at bedtime; and amlodipine, 5 mg at bedtime.
Mr. P was following up with his primary care physician for his medical conditions and was adherent with treatment until 1 week before he was admitted to our facility.
The authors’ observations
Always rule out medical causes in a case of new-onset mania, which is particularly important in geriatric patients. Older patients with new-onset mania are more than twice as likely to have a comorbid neurologic disorder.9 Neurologic causes of late-onset mania include:
• stroke
• tumor
• epilepsy
• Huntington’s disease and other movement disorders
• multiple sclerosis and other white-matter diseases
• head trauma
• infection (such as neurosyphilis)
• Creutzfeldt-Jakob disease
• frontotemporal dementia.10
Mr. P’s presentation of psychomotor agitation, impaired functioning, decreased need for sleep, increased energy, hyperverbal speech, and complex paranoid delusions meets DSM-5 criteria for bipolar disorder, manic phase. In addition, older manic patients frequently present with confusion, disorientation, and distractibility. Younger patients with mania often present with euphoric moods and grandiosity; in contrast, geriatric patients are more likely to show a mixture of depressed affect and manic symptoms (pressured speech and a decreased need for sleep).11-15
We considered an emerging neurodegenerative process, because dementia can present early with disinhibition, lability, and other behavioral disturbances, including classic manic syndromes.16 Although we could not fully rule out a neurodegenerative process in the initial phase of treatment, Mr. P’s longitudinal course demonstrated no change in baseline cognitive function and no evidence of subsequent decline, making dementia unlikely.17
Patients with frontotemporal dementia are more likely to present initially to a psychiatrist than to a neurologist.18
Frontotemporal dementia is a progressive neurodegenerative disease that affects the frontal and temporal cortices; it is a common cause of dementia in patients age <65.19 Frontotemporal dementia is characterized by insidious behavioral and personality changes; often, the initial presentation lacks any clear neurologic signs or symptoms. Key features include apathy, disinhibition, loss of sympathy and empathy, repetitive motor behaviors, and overeating.20
Mr. P’s symptoms stabilized with divalproex sprinkles and risperidone. There was no evidence of decline in memory, social interaction, or behavior.
EVALUATION Paranoia
On mental status exam, Mr. P has an appropriate appearance; he is clean and shaven, with good eye contact. Muscular tone and gait are within normal limits. Level of activity is increased; he exhibits psychomotor agitation. Speech is rapid, over-productive, and loud; thought process shows flight of ideas, and thought associations are circumstantial.
Mr. P has paranoid delusions about the staff trying to hurt him. His judgment is poor, evidenced by an inability to take care of himself. Insight is minimal, as seen by noncompliance with treatment. Mr. P is oriented only to person and place. His mood is anxious; affect is labile.
Complete blood count, comprehensive metabolic profile, blood alcohol level, urine analysis, urine toxicology, electrocardiogram, and CT scan of the head are within normal limits.
Mr. P is given a diagnosis of mood disorder due to general medical condition, psychotic disorder due to general medical condition. The team rules out acute delirium, bipolar I disorder, and neurodegenerative disorders such as frontotemporal dementia.
Mr. P is maintained on pre-admission medications for his medical conditions. A mood stabilizer, divalproex sprinkles, 250 mg/d, is added.
Once on the unit, Mr. P is re-evaluated. Divalproex is increased to 500 mg/d; risperidone, 0.5 mg/d, is added to address paranoia. Mr. P also receives group and individual psychotherapy. He does not participate in neuropsychological testing, and no single-photon emission CT analysis is done. Mr. P remains in the hospital for 2 weeks. After a family meeting, his daughter says she feels comfortable taking Mr. P home. He follows up in the outpatient clinic and is doing well.
The authors’ observations
Treating geriatric patients with bipolar disorder requires attention to several factors (Table). Older patients might tolerate or metabolize medications differently than younger adults, and therefore may need a different dosage. Older patients are more likely to have comorbid medical conditions and to be taking medications for those ailments. Treatment is much more complicated for this age group because physicians need to account for possible drug-drug interactions.21
A number of medications can be helpful in treating older patients who have bipolar disorder.11 Ongoing research compares lithium with anticonvulsants in older bipolar disorder patients to determine which drug has the greatest benefit with the lowest risk of side effects.
Psychotherapy can be a valuable addition to pharmacotherapy in older adults. Some psychotherapy programs are specifically geared to older bipolar disorder patients.22,23
Use of divalproex sodium in older patients
First, perform baseline laboratory tests: complete blood count, liver function, and electrocardiogram. Initiate divalproex sodium, 250 mg at bedtime, increasing the dosage every 3 to 5 days by 250 mg, with a target dose of 500 to 2,000 mg/d (divided into 2 or 3 doses). Monitor serum levels; levels of 29 to 100 μg/mL are effective and well tolerated. Common side effects include excess sedation, ataxia, tremor, nausea, and, rarely, hepatotoxicity, leukopenia, and thrombocytopenia.24
Use of lithium in geriatric patients
First, perform baseline laboratory tests: electrolytes, creatinine, blood urea nitrogen, urine, thyroid stimulating hormone, and electrocardiogram. Starting dosage is 300 mg at bedtime (150 mg for frail cachectic patients). Monitor serum levels 12 hours after last dose, adjusting dosage every 5 days until a target serum level of 0.5 to 0.8 mEq/L is reached. Common dosages for geriatric patients are 300 to 600 mg/d, which often can be given as a single bedtime dose. Cautions: When using lithium with a thiazide diuretic or nonsteroidal anti-inflammatory drug, watch for dehydration, vomiting, and diarrhea, which will elevate the serum lithium level. Side effects include ataxia, tremor, urinary frequency, thirst, nausea, diarrhea, hypothyroidism, and exacerbation of psoriasis. Once stabilized, monitor the serum lithium level, thyroid-stimulating hormone, and kidney function every 3 to 6 months.24
Bottom Line
In geriatric patients, bipolar disorder can present with agitation, irritability, confusion, and psychosis, rather than euphoric mood and grandiosity. When you suspect bipolar disorder in an older patient, first rule out medical causes of symptoms. When selecting treatment, consider comorbid medical conditions and possible drug-drug interactions.
Related Resources
• Sajatovic M, Forester BP, Gildengers A, et al. Aging changes and medical complexity in late-life bipolar disorder: emerging research findings that may help advance care. Neuropsychiatry (London). 2013;3(6):621-633.
• Dols A, Rhebergen D, Beekman A, et al. Psychiatric and medical comorbidities: results from a bipolar elderly cohort study. Am J Geriatr Psychiatry. 2014;22(11):1066-1074.
Drug Brand Names
Amiodarone • Cordarone Olanzapine • Zyprexa
Amlodipine • Norvasc Olmesartan medoxomil • Benicar
Divalproex sodium • Depakote Pantoprazole • Protonix
Eszopiclone • Lunesta Risperidone • Risperdal
Lithium • Eskalith, Lithobid Rivaroxaban • Xarelto
Lorazepam • Ativan Simvastatin • Zocor
Metformin • Glucophage Sitagliptin • Januvia
Metoprolol • Lopressor
Disclosures
The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
CASE Delusional and aggressive
Mr. P, age 78, of Filipino heritage, is brought to the psychiatric hospital because he has been verbally aggressive toward his wife for several weeks. He has no history of a psychiatric diagnosis or inpatient psychiatric hospitalization, and no history of taking any psychotropic medications.
According to his wife, Mr. P has been ruminating about his father, who died in World War II, saying that “the Japanese never gave his body back” to him. Also, his wife describes 3 weeks of physically aggressive behavior, such as throwing punches; the last episode was 2 days before admission.
Mr. P is not bathing, eating, taking his medications, and attending to his activities of daily living. He sleeps for only 1 to 2 hours a night; is irritable and easily distractible; and experiences flight of ideas. Mr. P has been buying lottery tickets, telling his daughter that he will become a millionaire and then buy a house in the Philippines.
Mr. P reports depressed mood, but no other depressive symptoms are present. He reports no suicidal or homicidal ideations, auditory or visual hallucinations, or anxiety symptoms. He has no history of substance abuse.
What diagnosis would you give Mr. P?
a) late-onset bipolar disorder
b) Alzheimer’s disease
c) major depressive disorder
d) frontotemporal dementia
The authors’ observations
Bipolar disorder in later life is a complex and confounding neuropsychiatric syndrome with diagnostic and therapeutic challenges. The disorder can affect people of all ages and is not uncommon among geriatric patients, with a 1-year prevalence in United States of 0.4%.1 In one study, 10% of new bipolar disorder cases were found to occur after age 50.2 As the American population grows older, the number of bipolar disorder cases among seniors is expected to increase.3
It was once thought that symptoms of bipolar disorder disappear with age; newer research has disproved this theory, and proposes that untreated bipolar disorder worsens over time.4 Persons who are given the diagnosis later in life could have had bipolar disorder for decades, but symptoms became more noticeable and problematic with age.5
Common symptoms in geriatric patients can differ from what we might expect in younger patients: agitation, hyperactivity, irritability, confusion, and psychosis.6 When the disorder presents in patients age >60, it can be severe, with significant changes in cognitive function, including difficulties with memory, perception, judgment, and problem-solving.7,8
HISTORY Medical comorbidities
Mr. P emigrated from the Philippines 20 years ago, is married, and lives with his wife. He has 3 brothers; his parents were divorced, and his mother remarried. Mr. P completed high school.
Mr. P has an extensive medical history: diabetes mellitus, hypertension, dyslipidemia, and recent double coronary artery bypass grafting. He is taking several medications: sitagliptin, 25 mg/d; pantoprazole, 5 mg/d; metformin, 1,000 mg/d; rivaroxaban, 20 mg/d; amiodarone, 200 mg/d; metoprolol, 12.5 mg/d; olmesartan medoxomil, 40 mg/d; aspirin, 81 mg/d; simvastatin, 10 mg/d; eszopiclone, 3 mg at bedtime; and amlodipine, 5 mg at bedtime.
Mr. P was following up with his primary care physician for his medical conditions and was adherent with treatment until 1 week before he was admitted to our facility.
The authors’ observations
Always rule out medical causes in a case of new-onset mania, which is particularly important in geriatric patients. Older patients with new-onset mania are more than twice as likely to have a comorbid neurologic disorder.9 Neurologic causes of late-onset mania include:
• stroke
• tumor
• epilepsy
• Huntington’s disease and other movement disorders
• multiple sclerosis and other white-matter diseases
• head trauma
• infection (such as neurosyphilis)
• Creutzfeldt-Jakob disease
• frontotemporal dementia.10
Mr. P’s presentation of psychomotor agitation, impaired functioning, decreased need for sleep, increased energy, hyperverbal speech, and complex paranoid delusions meets DSM-5 criteria for bipolar disorder, manic phase. In addition, older manic patients frequently present with confusion, disorientation, and distractibility. Younger patients with mania often present with euphoric moods and grandiosity; in contrast, geriatric patients are more likely to show a mixture of depressed affect and manic symptoms (pressured speech and a decreased need for sleep).11-15
We considered an emerging neurodegenerative process, because dementia can present early with disinhibition, lability, and other behavioral disturbances, including classic manic syndromes.16 Although we could not fully rule out a neurodegenerative process in the initial phase of treatment, Mr. P’s longitudinal course demonstrated no change in baseline cognitive function and no evidence of subsequent decline, making dementia unlikely.17
Patients with frontotemporal dementia are more likely to present initially to a psychiatrist than to a neurologist.18
Frontotemporal dementia is a progressive neurodegenerative disease that affects the frontal and temporal cortices; it is a common cause of dementia in patients age <65.19 Frontotemporal dementia is characterized by insidious behavioral and personality changes; often, the initial presentation lacks any clear neurologic signs or symptoms. Key features include apathy, disinhibition, loss of sympathy and empathy, repetitive motor behaviors, and overeating.20
Mr. P’s symptoms stabilized with divalproex sprinkles and risperidone. There was no evidence of decline in memory, social interaction, or behavior.
EVALUATION Paranoia
On mental status exam, Mr. P has an appropriate appearance; he is clean and shaven, with good eye contact. Muscular tone and gait are within normal limits. Level of activity is increased; he exhibits psychomotor agitation. Speech is rapid, over-productive, and loud; thought process shows flight of ideas, and thought associations are circumstantial.
Mr. P has paranoid delusions about the staff trying to hurt him. His judgment is poor, evidenced by an inability to take care of himself. Insight is minimal, as seen by noncompliance with treatment. Mr. P is oriented only to person and place. His mood is anxious; affect is labile.
Complete blood count, comprehensive metabolic profile, blood alcohol level, urine analysis, urine toxicology, electrocardiogram, and CT scan of the head are within normal limits.
Mr. P is given a diagnosis of mood disorder due to general medical condition, psychotic disorder due to general medical condition. The team rules out acute delirium, bipolar I disorder, and neurodegenerative disorders such as frontotemporal dementia.
Mr. P is maintained on pre-admission medications for his medical conditions. A mood stabilizer, divalproex sprinkles, 250 mg/d, is added.
Once on the unit, Mr. P is re-evaluated. Divalproex is increased to 500 mg/d; risperidone, 0.5 mg/d, is added to address paranoia. Mr. P also receives group and individual psychotherapy. He does not participate in neuropsychological testing, and no single-photon emission CT analysis is done. Mr. P remains in the hospital for 2 weeks. After a family meeting, his daughter says she feels comfortable taking Mr. P home. He follows up in the outpatient clinic and is doing well.
The authors’ observations
Treating geriatric patients with bipolar disorder requires attention to several factors (Table). Older patients might tolerate or metabolize medications differently than younger adults, and therefore may need a different dosage. Older patients are more likely to have comorbid medical conditions and to be taking medications for those ailments. Treatment is much more complicated for this age group because physicians need to account for possible drug-drug interactions.21
A number of medications can be helpful in treating older patients who have bipolar disorder.11 Ongoing research compares lithium with anticonvulsants in older bipolar disorder patients to determine which drug has the greatest benefit with the lowest risk of side effects.
Psychotherapy can be a valuable addition to pharmacotherapy in older adults. Some psychotherapy programs are specifically geared to older bipolar disorder patients.22,23
Use of divalproex sodium in older patients
First, perform baseline laboratory tests: complete blood count, liver function, and electrocardiogram. Initiate divalproex sodium, 250 mg at bedtime, increasing the dosage every 3 to 5 days by 250 mg, with a target dose of 500 to 2,000 mg/d (divided into 2 or 3 doses). Monitor serum levels; levels of 29 to 100 μg/mL are effective and well tolerated. Common side effects include excess sedation, ataxia, tremor, nausea, and, rarely, hepatotoxicity, leukopenia, and thrombocytopenia.24
Use of lithium in geriatric patients
First, perform baseline laboratory tests: electrolytes, creatinine, blood urea nitrogen, urine, thyroid stimulating hormone, and electrocardiogram. Starting dosage is 300 mg at bedtime (150 mg for frail cachectic patients). Monitor serum levels 12 hours after last dose, adjusting dosage every 5 days until a target serum level of 0.5 to 0.8 mEq/L is reached. Common dosages for geriatric patients are 300 to 600 mg/d, which often can be given as a single bedtime dose. Cautions: When using lithium with a thiazide diuretic or nonsteroidal anti-inflammatory drug, watch for dehydration, vomiting, and diarrhea, which will elevate the serum lithium level. Side effects include ataxia, tremor, urinary frequency, thirst, nausea, diarrhea, hypothyroidism, and exacerbation of psoriasis. Once stabilized, monitor the serum lithium level, thyroid-stimulating hormone, and kidney function every 3 to 6 months.24
Bottom Line
In geriatric patients, bipolar disorder can present with agitation, irritability, confusion, and psychosis, rather than euphoric mood and grandiosity. When you suspect bipolar disorder in an older patient, first rule out medical causes of symptoms. When selecting treatment, consider comorbid medical conditions and possible drug-drug interactions.
Related Resources
• Sajatovic M, Forester BP, Gildengers A, et al. Aging changes and medical complexity in late-life bipolar disorder: emerging research findings that may help advance care. Neuropsychiatry (London). 2013;3(6):621-633.
• Dols A, Rhebergen D, Beekman A, et al. Psychiatric and medical comorbidities: results from a bipolar elderly cohort study. Am J Geriatr Psychiatry. 2014;22(11):1066-1074.
Drug Brand Names
Amiodarone • Cordarone Olanzapine • Zyprexa
Amlodipine • Norvasc Olmesartan medoxomil • Benicar
Divalproex sodium • Depakote Pantoprazole • Protonix
Eszopiclone • Lunesta Risperidone • Risperdal
Lithium • Eskalith, Lithobid Rivaroxaban • Xarelto
Lorazepam • Ativan Simvastatin • Zocor
Metformin • Glucophage Sitagliptin • Januvia
Metoprolol • Lopressor
Disclosures
The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Weissman MM, Leaf PJ, Tischler GL, et al. Affective disorders in five United States communities. Psychol Med. 1988;18(1):141-153.
2. Yassa R, Nair NP, Iskandar H. Late-onset bipolar disorder. Psychiatr Clin North Am. 1988;11(1):117-131.
3. Verdoux H, Bourgeois M. Secondary mania caused by cerebral organic pathology [in French]. Ann Med Psychol (Paris). 1995;153(3):161-168.
4. Fadden G, Bebbington P, Kuipers L. The burden of care: the impact of functional psychiatric illness in the patient’s family. Br J Psychiatry. 1987;150:285-292.
5. Yassa R, Nair V, Nastase C, et al. Prevalence of bipolar disorder in a psychogeriatric population. J Affect Disord. 1988;14(3):197-201.
6. Robinson RG, Boston JD, Starkstein SE, et al. Comparison of mania with depression following brain injury: casual factors. Am J Psychiatry. 1988;145(2):172-178.
7. Starkstein SE, Boston JD, Robinson RG. Mechanisms of mania after brain injury: 12 case reports and review of the literature. J Nerv Ment Dis. 1988;176(2):87-100.
8. Herrmann N, Bremner KE, Naranjo CA. Pharmacotherapy of late life mood disorders. Clin Neurosci. 1997;4(1):41-47.
9. Tohen M, Shulman KI, Satlin A. First-episode mania in late life. Am J Psychiatry. 1994;151(1):130-132.
10. Mendez MF. Mania in neurologic disorders. Curr Psychiatry Rep. 2000;2(5):440-445.
11. Eagles JM, Whalley LJ. Aging and affective disorders: the age at first onset of affective disorders in Scotland, 1969- 1978. Br J Psychiatry. 1985;147:180-187.
12. Snowdon J. A retrospective case-note study of bipolar disorder in old age. Br J Psychiatry. 1991;158:485-490.
13. Winokur G. The Iowa 500: heterogeneity and course in manic-depressive illness (bipolar). Compr Psychiatry. 1975;16(2):125-131.
14. Shulman K, Post F. Bipolar affective disorder in old age. Br J Psychiatry. 1980;136:26-32.
15. Young RC, Falk JR. Age, manic psychopathology, and treatment response. Int J Geriatr Psychiatry. 1989;4(2):73-78.
16. Almeida OP. Bipolar disorder with late onset: an organic variety of mood disorder [in Portuguese]? Rev Bras Psiquiatr. 2004;26(suppl 3):27-30.
17. Carlino AR, Stinnett JL, Kim DR. New onset of bipolar disorder in late life. Psychosomatics. 2013;54(1):94-97.
18. Woolley JD, Wilson MR, Hung E, et al. Frontotemporal dementia and mania. Am J Psychiatry. 2007;164(12):1811-1816.
19. Ratnavalli E, Brayne C, Dawson K, et al. The prevalence of frontotemporal dementia. Neurology. 2002;58(11):1615-1621.
20. Gregory CA, Hodges JR. Clinical features of frontal lobe dementia in comparison to Alzheimer’s disease. J Neural Transm Suppl. 1996;47:103-123.
21. Broadhead J, Jacoby R. Mania in old age: a first prospective study. Int J Geriatr Psychiatry. 1990;5(4):215-222.
22. Dhingra U, Rabins PV. Mania in the elderly: a 5-7 year follow-up. J Am Geriatr Soc. 1991;39(6):581-583.
23. Shulman KI. Neurologic comorbidity and mania in old age. Clin Neurosci. 1997;4(1):37-40.
24. Shulman KI, Herrmann N. Bipolar disorder in old age. Can Fam Physician. 1999;45:1229-1237.
1. Weissman MM, Leaf PJ, Tischler GL, et al. Affective disorders in five United States communities. Psychol Med. 1988;18(1):141-153.
2. Yassa R, Nair NP, Iskandar H. Late-onset bipolar disorder. Psychiatr Clin North Am. 1988;11(1):117-131.
3. Verdoux H, Bourgeois M. Secondary mania caused by cerebral organic pathology [in French]. Ann Med Psychol (Paris). 1995;153(3):161-168.
4. Fadden G, Bebbington P, Kuipers L. The burden of care: the impact of functional psychiatric illness in the patient’s family. Br J Psychiatry. 1987;150:285-292.
5. Yassa R, Nair V, Nastase C, et al. Prevalence of bipolar disorder in a psychogeriatric population. J Affect Disord. 1988;14(3):197-201.
6. Robinson RG, Boston JD, Starkstein SE, et al. Comparison of mania with depression following brain injury: casual factors. Am J Psychiatry. 1988;145(2):172-178.
7. Starkstein SE, Boston JD, Robinson RG. Mechanisms of mania after brain injury: 12 case reports and review of the literature. J Nerv Ment Dis. 1988;176(2):87-100.
8. Herrmann N, Bremner KE, Naranjo CA. Pharmacotherapy of late life mood disorders. Clin Neurosci. 1997;4(1):41-47.
9. Tohen M, Shulman KI, Satlin A. First-episode mania in late life. Am J Psychiatry. 1994;151(1):130-132.
10. Mendez MF. Mania in neurologic disorders. Curr Psychiatry Rep. 2000;2(5):440-445.
11. Eagles JM, Whalley LJ. Aging and affective disorders: the age at first onset of affective disorders in Scotland, 1969- 1978. Br J Psychiatry. 1985;147:180-187.
12. Snowdon J. A retrospective case-note study of bipolar disorder in old age. Br J Psychiatry. 1991;158:485-490.
13. Winokur G. The Iowa 500: heterogeneity and course in manic-depressive illness (bipolar). Compr Psychiatry. 1975;16(2):125-131.
14. Shulman K, Post F. Bipolar affective disorder in old age. Br J Psychiatry. 1980;136:26-32.
15. Young RC, Falk JR. Age, manic psychopathology, and treatment response. Int J Geriatr Psychiatry. 1989;4(2):73-78.
16. Almeida OP. Bipolar disorder with late onset: an organic variety of mood disorder [in Portuguese]? Rev Bras Psiquiatr. 2004;26(suppl 3):27-30.
17. Carlino AR, Stinnett JL, Kim DR. New onset of bipolar disorder in late life. Psychosomatics. 2013;54(1):94-97.
18. Woolley JD, Wilson MR, Hung E, et al. Frontotemporal dementia and mania. Am J Psychiatry. 2007;164(12):1811-1816.
19. Ratnavalli E, Brayne C, Dawson K, et al. The prevalence of frontotemporal dementia. Neurology. 2002;58(11):1615-1621.
20. Gregory CA, Hodges JR. Clinical features of frontal lobe dementia in comparison to Alzheimer’s disease. J Neural Transm Suppl. 1996;47:103-123.
21. Broadhead J, Jacoby R. Mania in old age: a first prospective study. Int J Geriatr Psychiatry. 1990;5(4):215-222.
22. Dhingra U, Rabins PV. Mania in the elderly: a 5-7 year follow-up. J Am Geriatr Soc. 1991;39(6):581-583.
23. Shulman KI. Neurologic comorbidity and mania in old age. Clin Neurosci. 1997;4(1):37-40.
24. Shulman KI, Herrmann N. Bipolar disorder in old age. Can Fam Physician. 1999;45:1229-1237.
Outpatient mania
Treating bipolar mania in the outpatient setting: Risk vs reward
Manic episodes, by definition, are associated with significant social or occupational impairment.1 Some manic patients are violent or engage in reckless behaviors that can harm themselves or others, such as speeding, disrupting traffic, or playing with fire. When these patients present to a psychiatrist’s outpatient practice, involuntary hospitalization might be justified.
However, some manic patients, in spite of their elevated, expansive, or irritable mood state, never behave dangerously and might not meet legal criteria for involuntary hospitalization, although these criteria differ from state to state. These patients might see a psychiatrist because manic symptoms such as irritability, talkativeness, and impulsivity are bothersome to their family members but pose no serious danger (Box). In this situation, the psychiatrist can strongly encourage the patient to seek voluntary hospitalization or attend a partial hospitalization program. If the patient declines, the psychiatrist is left with 2 choices: initiate treatment in the outpatient setting or refuse to treat the patient and refer to another provider.
Treating “non-dangerous” mania in the outpatient setting is fraught with challenges:
• the possibility that the patient’s condition will progress to dangerousness
• poor adherence to treatment because of the patient’s limited insight
• the large amount of time required from the psychiatrist and care team to adequately manage the manic episode (eg, time spent with family members, frequent patient visits, and managing communications from the patient).
There are no guidelines to assist the office-based practitioner in treating mania in the outpatient setting. When considering dosing and optimal medication combinations for treating mania, clinical trials may be of limited value because most of these studies only included hospitalized manic patients.
Because of this dearth of knowledge, we provide recommendations based on our review of the literature and from our experience working with manic patients who refuse voluntary hospitalization and could not be hospitalized against their will. These recommendations are organized into 3 sections: diagnostic approach, treatment strategy, and family involvement.
Diagnostic approach
Making a diagnosis of mania might seem straightforward for clinicians who work in inpatient settings; however, mania might not present with classic florid symptoms among outpatients. Patients might have a chief concern of irritability, dysphoria, anxiety, or “insomnia,” which may lead clinicians to focus initially on non-bipolar conditions.2
During the interview, it is important to assess for any current DSM-5 symptoms of a manic episode, while being careful not to accept a patient’s denial of symptoms. Patients with mania often have poor insight and are unaware of changes from their baseline state when manic.3 Alternatively, manic patients may want you to believe that they are well and could minimize or deny all symptoms. Therefore, it is important to pay attention to mental status examination findings, such as hyperverbal speech, elated affect, psychomotor agitation, a tangential thought process, or flight of ideas.
Countertransference feelings of diagnostic confusion or frustration after long patient monologues or multiple interruptions by the patient should be incorporated into the diagnostic assessment. Family members or friends often can provide objective observations of behavioral changes necessary to secure the diagnosis.
Treatment strategy
Decision points. When treating manic outpatients, assess the need for hospitalization at each visit. Advantages of the inpatient setting include:
• the possibility of rapid medication adjustments
• continuous observation to ensure the patient’s safety
• keeping the patient temporarily removed from his community to prevent irreversible social and economic harms.
However, a challenge with hospitalization is third-party payers’ influence on a patient’s length of stay, which may lead to rapid medication changes that may not be clinically ideal.
At each outpatient visit, explore with the patient and family emerging symptoms that could justify involuntary hospitalization. Document whether you recommended inpatient hospitalization, the patient’s response to the recommendation, that you are aware and have considered the risks associated with outpatient care, and that you have discussed these risks with the patient and family.
For patients well-known to the psychiatrist, a history of dangerous mania may lead him (her) to strongly recommend hospitalization, whereas a pre-existing therapeutic alliance and no current or distant history of dangerous mania may lead the clinician to look for alternatives to inpatient care. Concomitant drug or alcohol use may increase the likelihood of mania becoming dangerous, making outpatient treatment ill-advised and riskier for everyone involved.
In exchange for agreeing to provide outpatient care for mania, it often is helpful to negotiate with the patient and family a threshold level of symptoms or behavior that will result in the patient agreeing to voluntary hospitalization (Table 1). Such an agreement can include stopping outpatient treatment if the patient does not improve significantly after 2 or 3 weeks or develops psychotic symptoms. The negotiation also can include partial hospitalization as an option, so long as the patient’s mania continues to be non-dangerous.
Obtaining pretreatment blood work can help a clinician determine whether a medication is safe to prescribe and establish causality if laboratory abnormalities arise after treatment begins. Ideally, the psychiatrist should follow consensus guidelines developed by the International Society for Bipolar Disorders4 or the American Psychiatric Association (APA)5 and order appropriate laboratory tests before prescribing anti-manic medications. Determine the pregnancy status of female patients of child-bearing age before prescribing a potentially teratogenic medication, especially because mania is associated with increased libido.6
Manic patients might be too disorganized to follow up with recommendations for laboratory testing, or could wait several days before completing blood work. Although not ideal, to avoid delaying treatment, a clinician might need to prescribe medication at the initial office visit, without pretreatment laboratory results. When the patient is more organized, complete the blood work. Keeping home pregnancy tests in the office can help rule out pregnancy before prescribing medication.
Medication. Meta-analyses have established the efficacy of mood stabilizers and antipsychotics for treating mania,7,8 and several consensus guidelines have incorporated these findings into treatment algorithms.9
For a patient already taking medications recommended by the guidelines, assess treatment adherence during the initial interview by questioning the patient and family. When the logistics of phlebotomy permit, obtaining the blood level of psychotropics can show the presence of any detectable drug concentration, which demonstrates that the patient has taken the medication recently.
If there is no evidence of nonadherence, an initial step might be to increase the dosage of the antipsychotic or mood stabilizer that the patient is already taking, ensuring that the dosage is optimized based on FDA indications and clinical trials data. The recommended rate of dosage adjustments differs among medications; however, optimal dosing should be reached quickly because a World Federation of Societies of Biological Psychiatry task force recommends that a mania treatment trial not exceed 2 weeks.10
Dosage increases can be made at weekly visits or sooner, based on treatment response and tolerability. If there is no benefit after optimizing the dosage, the next step would be to add a mood stabilizer to a second-generation antipsychotic (SGA), or vice versa to promote additive or synergistic medication effects.11 Switching one medication for the other should be avoided unless there are tolerability concerns.
For a patient who is not taking any medications, select a treatment that balances rapid stabilization with long-term efficacy and tolerability. Table 2 lists FDA-approved treatments for mania. Lamotrigine provides prophylactic efficacy with few associated risks, but it has no anti-manic effects and would be a poor choice for most actively manic patients. Most studies indicate that antipsychotics work faster than lithium at the 1-week mark; however, this may be a function of the lithium titration schedule followed in the protocols, the severity of mania among enrolled patients, the inclusion of typically non-responsive manic patients (eg, mixed) in the analysis, and the antipsychotic’s sedative potential relative to lithium. Although the anti-manic and prophylactic potential of lithium and valproate might make them an ideal first-line option, antipsychotics could stabilize a manic patient faster, especially if agitation is present.12,13
Breaking mania quickly is important when treating patients in the outpatient setting. In these situations, a reasonable choice is to prescribe a SGA, because of their rapid onset of effect, low potential for switch to depression, and utility in treating classic, mixed, or psychotic mania.10 Oral loading of valproate (20 mg/kg) is another option. An inpatient study that used an oral-loading strategy demonstrated a similar time to response as olanzapine,14 in contrast to an inpatient15 and an outpatient study16 that employed a standard starting dosage for each patient and led to slower improvement compared with olanzapine.
SGAs should be dosed moderately and lower than if the patient were hospitalized, to avoid alienating the patient from treatment by causing intolerable side effects. In particular, patients and their families should be warned about immediate risks, such as orthostasis or extrapyramidal symptoms. Although treatment guidelines recommend combination therapy as a possible first-line option,9 in the outpatient setting, monotherapy with an optimally dosed, rapid-acting agent is preferred to promote medication adherence and avoid potentially dangerous sedation. Manic patients experience increased distractibility and verbal memory and executive function impairments that can interfere with medication adherence.17 Therefore, patients are more likely to follow a simpler regimen. If SGA or valproate monotherapy does not control mania, begin combination treatment with a mood stabilizer and SGA. If the patient experiences remission with SGA monotherapy, the risks and benefits of maintaining the SGA vs switching to a mood stabilizer can be discussed.
Provide medication “as needed” for agitation—additional SGA dosing or a benzodiazepine—and explain to family members when their use is warranted. Benzodiazepines can provide short-term benefits for manic patients: anxiety relief, sedation, and anti-manic efficacy as monotherapy18-20 and in combination with other medications.21 Studies showing monotherapy efficacy employed high dosages of benzodiazepines (lorazepam mean dosage, 14 mg/d; clonazepam mean dosage, 13 mg/d)19 and high dosages of antipsychotics as needed,18,20 and often were associated with excessive sedation and ataxia.18,19 This makes benzodiazepine monotherapy a potentially dangerous approach for outpatient treatment of mania. IM lorazepam treated manic agitation less quickly than IM olanzapine, suggesting that SGAs are preferable in the outpatient setting because rapid control of agitation is crucial.22 If prescribed, a trusted family member should dispense benzodiazepines to the patient to minimize misuse because of impulsivity, distractibility, desperation to sleep, or pleasure seeking.
SGAs have the benefit of sedation but occasionally additional sleep medications are required. Benzodiazepine receptor agonists (BzRAs), such as zolpidem, eszopiclone, and zaleplon, should be used with caution. Although these medicines are effective in treating insomnia in individuals with primary insomnia23 and major depression,24 they have not been studied in manic patients. The decreased need for sleep in mania is phenomenologically25 and perhaps biologically different than insomnia in major depression.26 Therefore, mania-associated sleep disturbance might not respond to BZRAs. BzRAs also might induce somnambulism and other parasomnias,27 especially when used in combination with psychotropics, such as valproate28; it is unclear if the manic state itself increases this risk further. Sedating antihistamines with anticholinergic blockade, such as diphenhydramine and low dosages (<100 mg/d) of quetiapine, are best used only in combination with anti-manic medications because of putative link between anticholinergic blockade and manic induction.29 Less studied but safer options include novel anticonvulsants (gabapentin, pregabalin), melatonin, and melatonin receptor agonists. Sedating antidepressants, such as mirtazapine and trazodone, should be avoided.25
Important adjunctive treatment steps include discontinuing all pro-manic agents, including antidepressants, stimulants, and steroids, and discouraging use of caffeine, energy drinks, illicit drugs, and alcohol. The patient should return for office visits at least weekly, and possibly more frequently, depending on severity. Telephone check-in calls between scheduled visits may be necessary until the mania is broken.
Psychotherapy. Other than supportive therapy and psychoeducation, other forms of psychotherapy during mania are not indicated. Psychotherapy trials in bipolar disorder do not inform anti-manic efficacy because few have enrolled acutely manic patients and most report long-term benefits rather than short-term efficacy for the index manic episode.30 Educate patients about the importance of maintaining regular social rhythms and taking medication as prescribed. Manic patients might not be aware that they are acting differently during manic episodes, therefore efforts to improve the patient’s insight are unlikely to succeed. More time should be spent emphasizing the importance of adherence to treatment and taking anti-manic medications as prescribed. This discussion can be enhanced by focusing on the medication’s potential to reduce the unpleasant symptoms of mania, including irritability, insomnia, anxiety, and racing thoughts. At the first visit, discuss setting boundaries with the patient to reduce mania-driven, intrusive phone calls. A patient might develop insight after mania has resolved and he (she) can appreciate social or economic harm that occurred while manic. This discussion might foster adherence to maintenance treatment. Advise your patient to limit activities that may increase stimulation and perpetuate the mania, such as exercise, parties, concerts, or crowded shopping malls. Also, recommend that your patient stop working temporarily, to reduce stress and prevent any manic-driven interactions that could result in job loss.
If your patient has an established relationship with a psychotherapist, discuss with the therapist the plan to initiate mania treatment in the outpatient setting and work as a collaborative team, assuming that the patient has granted permission to share information. Encourage the therapist to increase the frequency of sessions with the patient to enable greater monitoring of changes in the patient’s manic symptoms.
Family involvement
Family support is crucial when treating mania in the outpatient setting. Lacking insight and organization, manic patients require the “auxiliary” judgment of trusted family members to ensure treatment success. The family should identify a single person to act as the liaison between the family and the psychiatrist. The psychiatrist should instruct this individual to accompany the patient to each clinic visit and provide regular updates on the patient’s adherence to treatment, changes in symptoms, and any new behaviors that would justify involuntary hospitalization. The treatment plan should be clearly communicated to this individual to ensure that it is implemented correctly. Ideally, this individual would be someone who understands that bipolar disorder is a mental illness, who can tolerate the patient’s potential resentment of them for taking on this role, and who can influence the patient and the other family members to adhere to the treatment plan.
This family member also should watch the patient take medication to rule out nonadherence if the patient’s condition does not improve.
Provide extensive psychoeducation to the family (Table 3). Discuss these teaching points and their implications at length during the first visit and reinforce them at subsequent visits. Advise spouses that the acute manic period is not the time to make major decisions about their marriage or to engage in couple’s therapy. These options are better explored after the patient recovers from the manic episode.
Encourage the family to engage in mania harm-reduction techniques to the extent that the patient will allow (Table 4). In particular, they should hold onto their loved one’s credit cards and checkbook, and discourage the patient from making any major financial decisions until the mania has resolved. Additionally, patients should be relieved of childcare responsibilities during this period. If there are any child welfare safety concerns, the clinician will need to report this to authorities as required by local laws.
Advise family members or roommates to call emergency services and request a crisis intervention team, or to take the patient to an emergency room if he (she) makes verbal threats to harm themselves or others, is violent, or demonstrates behaviors that indicate that he is no longer able to care for himself. The psychiatrist should assist with completing Family and Medical Leave Act paperwork for family members who will monitor the patient at home, a work-excuse letter for the patient so he does not lose his job, and short-term disability paperwork to ensure income for the patient during the manic period.
These interventions can be challenging for the entire family system because they place family members in a paternalistic role and reduce the patient’s autonomy within the family. This is problematic when these role changes occur between spouses or between a patient-parent and his (her) children. Such changes typically need to be reversed over time and may require the help of a family or couple’s therapist. To support the psychological health of the patient’s family, refer them to the National Alliance on Mental Illness for family support groups or to individual psychotherapists.
Outpatient management can be rewarding
For “non-dangerous” manic patients who cannot be hospitalized involuntarily and refuse full or partial hospitalization, a psychiatrist must choose between beginning treatment in the clinic and referring the patient to another provider. The latter option is consistent with the APA’s ethical guidelines,31 but must be done appropriately to avoid legal liability.32 This decision may disappoint a family desperate to see their loved one recover quickly and may leave them feeling betrayed by the mental health system. On the other hand, choosing to treat mania in the outpatient setting can be rewarding when resolution of mania restores the family’s homeostasis.
To achieve this outcome, the outpatient psychiatrist must engage the patient’s family to ensure that the patient adheres to the treatment plan and monitor for potentially dangerous behavior. The psychiatrist also must use his knowledge of mood symptoms, cognitive impairments, and the psychological experience of manic patients to create a safe and effective treatment strategy that the patient and family can implement.
Because of mania’s unpredictability and destructive potential, psychiatrists who agree to treat manic patients as outpatients should be familiar with their state’s statutes and case law that pertain to the refusal to accept a new patient, patient abandonment, involuntary hospitalization, confidentiality, and mandatory reporting. They also should seek clinical or legal consultation if they feel overwhelmed or uncertain about the safest and most legally sound approach.
Bottom Line
Treating mania in the outpatient setting is risky but can be accomplished in select patients with the help of the patient’s family and a strategy that integrates evidence-based pharmacotherapeutic and psychotherapeutic strategies. Because manic patients could display dangerous behavior, be familiar with your state’s laws regarding involuntary commitment, patient abandonment, and mandatory reporting.
Related Resources
• National Alliance on Mental Illness. www.NAMI.org.
• Depression and Bipolar Support Alliance. www.DBSAlliance.org.
Drug Brand Names
Aripiprazole • Abilify Mirtazapine • Remeron
Asenapine • Saphris Olanzapine • Zyprexa
Carbamazepine • Equetro, Tegretol Pregabalin • Lyrica
Chlorpromazine • Thorazine Quetiapine • Seroquel
Clonazepam • Klonopin Risperidone • Risperdal
Diphrenhydramine • Benadryl Trazodone • Desyrel
Eszopiclone • Lunesta Valproate • Divalproex
Gabapentin • Neurontin Zaleplon • Sonata
Lamotrigine • Lamictal Ziprasidone • Geodon
Lithium • Eskalith, Lithobid Zolpidem • Ambien
Lorazepam • Ativan
Acknowledgement
The authors thank Peter Ash, MD, for carefully reviewing this manuscript and providing feedback.
Disclosures
Dr. Rakofsky receives research or grant support from Takeda. Dr. Dunlop receives research or grant support from Forest, GlaxoSmithKline, and Otsuka.
1. Diagnostic and statistical manual of mental disorders. 5th ed. Washington, DC: American Psychiatric Association; 2013.
2. Cassidy F, Murry E, Forest K, et al. Signs and symptoms of mania in pure and mixed episodes. J Affect Disord. 1998;50(2-3):187-201.
3. Yen CF, Chen CS, Ko CH, et al. Changes in insight among patients with bipolar I disorder: a 2-year prospective study. Bipolar Disord. 2007;9(3):238-242.
4. Ng F, Mammen OK, Wilting I, et al. The International Society for Bipolar Disorders (ISBD) consensus guidelines for the safety monitoring of bipolar disorder treatments. Bipolar Disord. 2009;11(6):559-595.
5. American Psychiatric Association. Practice guideline for the treatment of patients with bipolar disorder (revision). Am J Psychiatry. 2002;159(suppl 4):1-50.
6. Allison JB, Wilson WP. Sexual behavior of manic patients: a preliminary report. South Med J. 1960;53:870-874.
7. Cipriani A, Barbui C, Salanti G, et al. Comparative efficacy and acceptability of antimanic drugs in acute mania: a multiple-treatments meta-analysis. Lancet. 2011; 378(9799):1306-1315.
8. Yildiz A, Vieta E, Leucht S, et al. Efficacy of antimanic treatments: meta-analysis of randomized, controlled trials. Neuropsychopharmacology. 2011;36(2):375-389.
9. Nivoli AM, Murru A, Goikolea JM, et al. New treatment guidelines for acute bipolar mania: a critical review. J Affect Disord. 2012;140(2):125-141.
10. Grunze H, Vieta E, Goodwin GM, et al. The World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the biological treatment of bipolar disorders: update 2009 on the treatment of acute mania. World J Biol Psychiatry. 2009;10(2):85-116.
11. Tohen M, Chengappa KN, Suppes T, et al. Efficacy of olanzapine in combination with valproate or lithium in the treatment of mania in patients partially nonresponsive to valproate or lithium monotherapy. Arch Gen Psychiatry. 2002;59(1):62-69.
12. Tohen M, Jacobs TG, Feldman PD. Onset of action of antipsychotics in the treatment of mania. Bipolar Disord. 2000;2(3 pt 2):261-268.
13. Goikolea JM, Colom F, Capapey J, et al. Faster onset of antimanic action with haloperidol compared to second-generation antipsychotics. A meta-analysis of randomized clinical trials in acute mania. Eur Neuropsychopharmacol. 2013;23(4):305-316.
14. Zajecka JM, Weisler R, Sachs G, et al. A comparison of the efficacy, safety, and tolerability of divalproex sodium and olanzapine in the treatment of bipolar disorder. J Clin Psychiatry. 2002;63(12):1148-1155.
15. Tohen M, Baker RW, Altshuler LL, et al. Olanzapine versus divalproex in the treatment of acute mania. Am J Psychiatry. 2002;159(6):1011-1017.
16. Tohen M, Vieta E, Goodwin GM, et al. Olanzapine versus divalproex versus placebo in the treatment of mild to moderate mania: a randomized, 12-week, double-blind study. J Clin Psychiatry. 2008;69(11):1776-1789.
17. Martínez-Arán A, Vieta E, Reinares M, et al. Cognitive function across manic or hypomanic, depressed, and euthymic states in bipolar disorder. Am J Psychiatry. 2004; 161(2):262-270.
18. Edwards R, Stephenson U, Flewett T. Clonazepam in acute mania: a double blind trial. Aust N Z J Psychiatry. 1991;25(2):238-242.
19. Bradwejn J, Shriqui C, Koszycki D, et al. Double-blind comparison of the effects of clonazepam and lorazepam in acute mania. J Clin Psychopharmacol. 1990;10(6):403-408.
20. Clark HM, Berk M, Brook S. A randomized controlled single blind study of the efficacy of clonazepam and lithium in the treatment of acute mania. Human Psychopharmacology: Clinical and Experimental. 1997;12(4):325-328.
21. Lenox RH, Newhouse PA, Creelman WL, et al. Adjunctive treatment of manic agitation with lorazepam versus haloperidol: a double-blind study. J Clin Psychiatry. 1992;53(2):47-52.
22. Meehan K, Zhang F, David S, et al. A double-blind, randomized comparison of the efficacy and safety of intramuscular injections of olanzapine, lorazepam, or placebo in treating acutely agitated patients diagnosed with bipolar mania. J Clin Psychopharmacol. 2001;21(4):389-397.
23. Huedo-Medina TB, Kirsch I, Middlemass J, et al. Effectiveness of non-benzodiazepine hypnotics in treatment of adult insomnia: meta-analysis of data submitted to the Food and Drug Administration. BMJ. 2012;345:e8343. doi: 10.1136/bmj.e8343.
24. Fava M, Asnis GM, Shrivastava RK, et al. Improved insomnia symptoms and sleep-related next-day functioning in patients with comorbid major depressive disorder and insomnia following concomitant zolpidem extended-release 12.5 mg and escitalopram treatment: a randomized controlled trial. J Clin Psychiatry. 2011;72(7):914-928.
25. Plante DT, Winkelman JW. Sleep disturbance in bipolar disorder: therapeutic implications. Am J Psychiatry. 2008;165(7):830-843.
26. Linkowski P, Kerkhofs M, Rielaert C, et al. Sleep during mania in manic-depressive males. Eur Arch Psychiatry Neurol Sci. 1986;235(6):339-341.
27. Poceta JS. Zolpidem ingestion, automatisms, and sleep driving: a clinical and legal case series. J Clin Sleep Med. 2011;7(6):632-638.
28. Sattar SP, Ramaswamy S, Bhatia SC, et al. Somnambulism due to probable interaction of valproic acid and zolpidem. Ann Pharmacother. 2003;37(10):1429-1433.
29. Rybakowski JK, Koszewska I, Puzynski S. Anticholinergic mechanisms: a forgotten cause of the switch process in bipolar disorder [Comment on: The neurolobiology of the switch process in bipolar disorder: a review. J Clin Psychiatry. 2010]. J Clin Psychiatry. 2010;71(12):1698-1699; author reply 1699-1700.
30. Miklowitz DJ. Adjunctive psychotherapy for bipolar disorder: state of the evidence. Am J Psychiatry. 2008;165(11):1408-1419.
31. American Psychiatric Association. The principles of medical ethics with annotations especially applicable to psychiatry. Arlington, VA: American Psychiatric Association; 2013.
32. Simon RI, Shuman DW. Clinical manual of psychiatry and law. Arlington, VA: American Psychiatric Publishing; 2007:17-36.
Manic episodes, by definition, are associated with significant social or occupational impairment.1 Some manic patients are violent or engage in reckless behaviors that can harm themselves or others, such as speeding, disrupting traffic, or playing with fire. When these patients present to a psychiatrist’s outpatient practice, involuntary hospitalization might be justified.
However, some manic patients, in spite of their elevated, expansive, or irritable mood state, never behave dangerously and might not meet legal criteria for involuntary hospitalization, although these criteria differ from state to state. These patients might see a psychiatrist because manic symptoms such as irritability, talkativeness, and impulsivity are bothersome to their family members but pose no serious danger (Box). In this situation, the psychiatrist can strongly encourage the patient to seek voluntary hospitalization or attend a partial hospitalization program. If the patient declines, the psychiatrist is left with 2 choices: initiate treatment in the outpatient setting or refuse to treat the patient and refer to another provider.
Treating “non-dangerous” mania in the outpatient setting is fraught with challenges:
• the possibility that the patient’s condition will progress to dangerousness
• poor adherence to treatment because of the patient’s limited insight
• the large amount of time required from the psychiatrist and care team to adequately manage the manic episode (eg, time spent with family members, frequent patient visits, and managing communications from the patient).
There are no guidelines to assist the office-based practitioner in treating mania in the outpatient setting. When considering dosing and optimal medication combinations for treating mania, clinical trials may be of limited value because most of these studies only included hospitalized manic patients.
Because of this dearth of knowledge, we provide recommendations based on our review of the literature and from our experience working with manic patients who refuse voluntary hospitalization and could not be hospitalized against their will. These recommendations are organized into 3 sections: diagnostic approach, treatment strategy, and family involvement.
Diagnostic approach
Making a diagnosis of mania might seem straightforward for clinicians who work in inpatient settings; however, mania might not present with classic florid symptoms among outpatients. Patients might have a chief concern of irritability, dysphoria, anxiety, or “insomnia,” which may lead clinicians to focus initially on non-bipolar conditions.2
During the interview, it is important to assess for any current DSM-5 symptoms of a manic episode, while being careful not to accept a patient’s denial of symptoms. Patients with mania often have poor insight and are unaware of changes from their baseline state when manic.3 Alternatively, manic patients may want you to believe that they are well and could minimize or deny all symptoms. Therefore, it is important to pay attention to mental status examination findings, such as hyperverbal speech, elated affect, psychomotor agitation, a tangential thought process, or flight of ideas.
Countertransference feelings of diagnostic confusion or frustration after long patient monologues or multiple interruptions by the patient should be incorporated into the diagnostic assessment. Family members or friends often can provide objective observations of behavioral changes necessary to secure the diagnosis.
Treatment strategy
Decision points. When treating manic outpatients, assess the need for hospitalization at each visit. Advantages of the inpatient setting include:
• the possibility of rapid medication adjustments
• continuous observation to ensure the patient’s safety
• keeping the patient temporarily removed from his community to prevent irreversible social and economic harms.
However, a challenge with hospitalization is third-party payers’ influence on a patient’s length of stay, which may lead to rapid medication changes that may not be clinically ideal.
At each outpatient visit, explore with the patient and family emerging symptoms that could justify involuntary hospitalization. Document whether you recommended inpatient hospitalization, the patient’s response to the recommendation, that you are aware and have considered the risks associated with outpatient care, and that you have discussed these risks with the patient and family.
For patients well-known to the psychiatrist, a history of dangerous mania may lead him (her) to strongly recommend hospitalization, whereas a pre-existing therapeutic alliance and no current or distant history of dangerous mania may lead the clinician to look for alternatives to inpatient care. Concomitant drug or alcohol use may increase the likelihood of mania becoming dangerous, making outpatient treatment ill-advised and riskier for everyone involved.
In exchange for agreeing to provide outpatient care for mania, it often is helpful to negotiate with the patient and family a threshold level of symptoms or behavior that will result in the patient agreeing to voluntary hospitalization (Table 1). Such an agreement can include stopping outpatient treatment if the patient does not improve significantly after 2 or 3 weeks or develops psychotic symptoms. The negotiation also can include partial hospitalization as an option, so long as the patient’s mania continues to be non-dangerous.
Obtaining pretreatment blood work can help a clinician determine whether a medication is safe to prescribe and establish causality if laboratory abnormalities arise after treatment begins. Ideally, the psychiatrist should follow consensus guidelines developed by the International Society for Bipolar Disorders4 or the American Psychiatric Association (APA)5 and order appropriate laboratory tests before prescribing anti-manic medications. Determine the pregnancy status of female patients of child-bearing age before prescribing a potentially teratogenic medication, especially because mania is associated with increased libido.6
Manic patients might be too disorganized to follow up with recommendations for laboratory testing, or could wait several days before completing blood work. Although not ideal, to avoid delaying treatment, a clinician might need to prescribe medication at the initial office visit, without pretreatment laboratory results. When the patient is more organized, complete the blood work. Keeping home pregnancy tests in the office can help rule out pregnancy before prescribing medication.
Medication. Meta-analyses have established the efficacy of mood stabilizers and antipsychotics for treating mania,7,8 and several consensus guidelines have incorporated these findings into treatment algorithms.9
For a patient already taking medications recommended by the guidelines, assess treatment adherence during the initial interview by questioning the patient and family. When the logistics of phlebotomy permit, obtaining the blood level of psychotropics can show the presence of any detectable drug concentration, which demonstrates that the patient has taken the medication recently.
If there is no evidence of nonadherence, an initial step might be to increase the dosage of the antipsychotic or mood stabilizer that the patient is already taking, ensuring that the dosage is optimized based on FDA indications and clinical trials data. The recommended rate of dosage adjustments differs among medications; however, optimal dosing should be reached quickly because a World Federation of Societies of Biological Psychiatry task force recommends that a mania treatment trial not exceed 2 weeks.10
Dosage increases can be made at weekly visits or sooner, based on treatment response and tolerability. If there is no benefit after optimizing the dosage, the next step would be to add a mood stabilizer to a second-generation antipsychotic (SGA), or vice versa to promote additive or synergistic medication effects.11 Switching one medication for the other should be avoided unless there are tolerability concerns.
For a patient who is not taking any medications, select a treatment that balances rapid stabilization with long-term efficacy and tolerability. Table 2 lists FDA-approved treatments for mania. Lamotrigine provides prophylactic efficacy with few associated risks, but it has no anti-manic effects and would be a poor choice for most actively manic patients. Most studies indicate that antipsychotics work faster than lithium at the 1-week mark; however, this may be a function of the lithium titration schedule followed in the protocols, the severity of mania among enrolled patients, the inclusion of typically non-responsive manic patients (eg, mixed) in the analysis, and the antipsychotic’s sedative potential relative to lithium. Although the anti-manic and prophylactic potential of lithium and valproate might make them an ideal first-line option, antipsychotics could stabilize a manic patient faster, especially if agitation is present.12,13
Breaking mania quickly is important when treating patients in the outpatient setting. In these situations, a reasonable choice is to prescribe a SGA, because of their rapid onset of effect, low potential for switch to depression, and utility in treating classic, mixed, or psychotic mania.10 Oral loading of valproate (20 mg/kg) is another option. An inpatient study that used an oral-loading strategy demonstrated a similar time to response as olanzapine,14 in contrast to an inpatient15 and an outpatient study16 that employed a standard starting dosage for each patient and led to slower improvement compared with olanzapine.
SGAs should be dosed moderately and lower than if the patient were hospitalized, to avoid alienating the patient from treatment by causing intolerable side effects. In particular, patients and their families should be warned about immediate risks, such as orthostasis or extrapyramidal symptoms. Although treatment guidelines recommend combination therapy as a possible first-line option,9 in the outpatient setting, monotherapy with an optimally dosed, rapid-acting agent is preferred to promote medication adherence and avoid potentially dangerous sedation. Manic patients experience increased distractibility and verbal memory and executive function impairments that can interfere with medication adherence.17 Therefore, patients are more likely to follow a simpler regimen. If SGA or valproate monotherapy does not control mania, begin combination treatment with a mood stabilizer and SGA. If the patient experiences remission with SGA monotherapy, the risks and benefits of maintaining the SGA vs switching to a mood stabilizer can be discussed.
Provide medication “as needed” for agitation—additional SGA dosing or a benzodiazepine—and explain to family members when their use is warranted. Benzodiazepines can provide short-term benefits for manic patients: anxiety relief, sedation, and anti-manic efficacy as monotherapy18-20 and in combination with other medications.21 Studies showing monotherapy efficacy employed high dosages of benzodiazepines (lorazepam mean dosage, 14 mg/d; clonazepam mean dosage, 13 mg/d)19 and high dosages of antipsychotics as needed,18,20 and often were associated with excessive sedation and ataxia.18,19 This makes benzodiazepine monotherapy a potentially dangerous approach for outpatient treatment of mania. IM lorazepam treated manic agitation less quickly than IM olanzapine, suggesting that SGAs are preferable in the outpatient setting because rapid control of agitation is crucial.22 If prescribed, a trusted family member should dispense benzodiazepines to the patient to minimize misuse because of impulsivity, distractibility, desperation to sleep, or pleasure seeking.
SGAs have the benefit of sedation but occasionally additional sleep medications are required. Benzodiazepine receptor agonists (BzRAs), such as zolpidem, eszopiclone, and zaleplon, should be used with caution. Although these medicines are effective in treating insomnia in individuals with primary insomnia23 and major depression,24 they have not been studied in manic patients. The decreased need for sleep in mania is phenomenologically25 and perhaps biologically different than insomnia in major depression.26 Therefore, mania-associated sleep disturbance might not respond to BZRAs. BzRAs also might induce somnambulism and other parasomnias,27 especially when used in combination with psychotropics, such as valproate28; it is unclear if the manic state itself increases this risk further. Sedating antihistamines with anticholinergic blockade, such as diphenhydramine and low dosages (<100 mg/d) of quetiapine, are best used only in combination with anti-manic medications because of putative link between anticholinergic blockade and manic induction.29 Less studied but safer options include novel anticonvulsants (gabapentin, pregabalin), melatonin, and melatonin receptor agonists. Sedating antidepressants, such as mirtazapine and trazodone, should be avoided.25
Important adjunctive treatment steps include discontinuing all pro-manic agents, including antidepressants, stimulants, and steroids, and discouraging use of caffeine, energy drinks, illicit drugs, and alcohol. The patient should return for office visits at least weekly, and possibly more frequently, depending on severity. Telephone check-in calls between scheduled visits may be necessary until the mania is broken.
Psychotherapy. Other than supportive therapy and psychoeducation, other forms of psychotherapy during mania are not indicated. Psychotherapy trials in bipolar disorder do not inform anti-manic efficacy because few have enrolled acutely manic patients and most report long-term benefits rather than short-term efficacy for the index manic episode.30 Educate patients about the importance of maintaining regular social rhythms and taking medication as prescribed. Manic patients might not be aware that they are acting differently during manic episodes, therefore efforts to improve the patient’s insight are unlikely to succeed. More time should be spent emphasizing the importance of adherence to treatment and taking anti-manic medications as prescribed. This discussion can be enhanced by focusing on the medication’s potential to reduce the unpleasant symptoms of mania, including irritability, insomnia, anxiety, and racing thoughts. At the first visit, discuss setting boundaries with the patient to reduce mania-driven, intrusive phone calls. A patient might develop insight after mania has resolved and he (she) can appreciate social or economic harm that occurred while manic. This discussion might foster adherence to maintenance treatment. Advise your patient to limit activities that may increase stimulation and perpetuate the mania, such as exercise, parties, concerts, or crowded shopping malls. Also, recommend that your patient stop working temporarily, to reduce stress and prevent any manic-driven interactions that could result in job loss.
If your patient has an established relationship with a psychotherapist, discuss with the therapist the plan to initiate mania treatment in the outpatient setting and work as a collaborative team, assuming that the patient has granted permission to share information. Encourage the therapist to increase the frequency of sessions with the patient to enable greater monitoring of changes in the patient’s manic symptoms.
Family involvement
Family support is crucial when treating mania in the outpatient setting. Lacking insight and organization, manic patients require the “auxiliary” judgment of trusted family members to ensure treatment success. The family should identify a single person to act as the liaison between the family and the psychiatrist. The psychiatrist should instruct this individual to accompany the patient to each clinic visit and provide regular updates on the patient’s adherence to treatment, changes in symptoms, and any new behaviors that would justify involuntary hospitalization. The treatment plan should be clearly communicated to this individual to ensure that it is implemented correctly. Ideally, this individual would be someone who understands that bipolar disorder is a mental illness, who can tolerate the patient’s potential resentment of them for taking on this role, and who can influence the patient and the other family members to adhere to the treatment plan.
This family member also should watch the patient take medication to rule out nonadherence if the patient’s condition does not improve.
Provide extensive psychoeducation to the family (Table 3). Discuss these teaching points and their implications at length during the first visit and reinforce them at subsequent visits. Advise spouses that the acute manic period is not the time to make major decisions about their marriage or to engage in couple’s therapy. These options are better explored after the patient recovers from the manic episode.
Encourage the family to engage in mania harm-reduction techniques to the extent that the patient will allow (Table 4). In particular, they should hold onto their loved one’s credit cards and checkbook, and discourage the patient from making any major financial decisions until the mania has resolved. Additionally, patients should be relieved of childcare responsibilities during this period. If there are any child welfare safety concerns, the clinician will need to report this to authorities as required by local laws.
Advise family members or roommates to call emergency services and request a crisis intervention team, or to take the patient to an emergency room if he (she) makes verbal threats to harm themselves or others, is violent, or demonstrates behaviors that indicate that he is no longer able to care for himself. The psychiatrist should assist with completing Family and Medical Leave Act paperwork for family members who will monitor the patient at home, a work-excuse letter for the patient so he does not lose his job, and short-term disability paperwork to ensure income for the patient during the manic period.
These interventions can be challenging for the entire family system because they place family members in a paternalistic role and reduce the patient’s autonomy within the family. This is problematic when these role changes occur between spouses or between a patient-parent and his (her) children. Such changes typically need to be reversed over time and may require the help of a family or couple’s therapist. To support the psychological health of the patient’s family, refer them to the National Alliance on Mental Illness for family support groups or to individual psychotherapists.
Outpatient management can be rewarding
For “non-dangerous” manic patients who cannot be hospitalized involuntarily and refuse full or partial hospitalization, a psychiatrist must choose between beginning treatment in the clinic and referring the patient to another provider. The latter option is consistent with the APA’s ethical guidelines,31 but must be done appropriately to avoid legal liability.32 This decision may disappoint a family desperate to see their loved one recover quickly and may leave them feeling betrayed by the mental health system. On the other hand, choosing to treat mania in the outpatient setting can be rewarding when resolution of mania restores the family’s homeostasis.
To achieve this outcome, the outpatient psychiatrist must engage the patient’s family to ensure that the patient adheres to the treatment plan and monitor for potentially dangerous behavior. The psychiatrist also must use his knowledge of mood symptoms, cognitive impairments, and the psychological experience of manic patients to create a safe and effective treatment strategy that the patient and family can implement.
Because of mania’s unpredictability and destructive potential, psychiatrists who agree to treat manic patients as outpatients should be familiar with their state’s statutes and case law that pertain to the refusal to accept a new patient, patient abandonment, involuntary hospitalization, confidentiality, and mandatory reporting. They also should seek clinical or legal consultation if they feel overwhelmed or uncertain about the safest and most legally sound approach.
Bottom Line
Treating mania in the outpatient setting is risky but can be accomplished in select patients with the help of the patient’s family and a strategy that integrates evidence-based pharmacotherapeutic and psychotherapeutic strategies. Because manic patients could display dangerous behavior, be familiar with your state’s laws regarding involuntary commitment, patient abandonment, and mandatory reporting.
Related Resources
• National Alliance on Mental Illness. www.NAMI.org.
• Depression and Bipolar Support Alliance. www.DBSAlliance.org.
Drug Brand Names
Aripiprazole • Abilify Mirtazapine • Remeron
Asenapine • Saphris Olanzapine • Zyprexa
Carbamazepine • Equetro, Tegretol Pregabalin • Lyrica
Chlorpromazine • Thorazine Quetiapine • Seroquel
Clonazepam • Klonopin Risperidone • Risperdal
Diphrenhydramine • Benadryl Trazodone • Desyrel
Eszopiclone • Lunesta Valproate • Divalproex
Gabapentin • Neurontin Zaleplon • Sonata
Lamotrigine • Lamictal Ziprasidone • Geodon
Lithium • Eskalith, Lithobid Zolpidem • Ambien
Lorazepam • Ativan
Acknowledgement
The authors thank Peter Ash, MD, for carefully reviewing this manuscript and providing feedback.
Disclosures
Dr. Rakofsky receives research or grant support from Takeda. Dr. Dunlop receives research or grant support from Forest, GlaxoSmithKline, and Otsuka.
Manic episodes, by definition, are associated with significant social or occupational impairment.1 Some manic patients are violent or engage in reckless behaviors that can harm themselves or others, such as speeding, disrupting traffic, or playing with fire. When these patients present to a psychiatrist’s outpatient practice, involuntary hospitalization might be justified.
However, some manic patients, in spite of their elevated, expansive, or irritable mood state, never behave dangerously and might not meet legal criteria for involuntary hospitalization, although these criteria differ from state to state. These patients might see a psychiatrist because manic symptoms such as irritability, talkativeness, and impulsivity are bothersome to their family members but pose no serious danger (Box). In this situation, the psychiatrist can strongly encourage the patient to seek voluntary hospitalization or attend a partial hospitalization program. If the patient declines, the psychiatrist is left with 2 choices: initiate treatment in the outpatient setting or refuse to treat the patient and refer to another provider.
Treating “non-dangerous” mania in the outpatient setting is fraught with challenges:
• the possibility that the patient’s condition will progress to dangerousness
• poor adherence to treatment because of the patient’s limited insight
• the large amount of time required from the psychiatrist and care team to adequately manage the manic episode (eg, time spent with family members, frequent patient visits, and managing communications from the patient).
There are no guidelines to assist the office-based practitioner in treating mania in the outpatient setting. When considering dosing and optimal medication combinations for treating mania, clinical trials may be of limited value because most of these studies only included hospitalized manic patients.
Because of this dearth of knowledge, we provide recommendations based on our review of the literature and from our experience working with manic patients who refuse voluntary hospitalization and could not be hospitalized against their will. These recommendations are organized into 3 sections: diagnostic approach, treatment strategy, and family involvement.
Diagnostic approach
Making a diagnosis of mania might seem straightforward for clinicians who work in inpatient settings; however, mania might not present with classic florid symptoms among outpatients. Patients might have a chief concern of irritability, dysphoria, anxiety, or “insomnia,” which may lead clinicians to focus initially on non-bipolar conditions.2
During the interview, it is important to assess for any current DSM-5 symptoms of a manic episode, while being careful not to accept a patient’s denial of symptoms. Patients with mania often have poor insight and are unaware of changes from their baseline state when manic.3 Alternatively, manic patients may want you to believe that they are well and could minimize or deny all symptoms. Therefore, it is important to pay attention to mental status examination findings, such as hyperverbal speech, elated affect, psychomotor agitation, a tangential thought process, or flight of ideas.
Countertransference feelings of diagnostic confusion or frustration after long patient monologues or multiple interruptions by the patient should be incorporated into the diagnostic assessment. Family members or friends often can provide objective observations of behavioral changes necessary to secure the diagnosis.
Treatment strategy
Decision points. When treating manic outpatients, assess the need for hospitalization at each visit. Advantages of the inpatient setting include:
• the possibility of rapid medication adjustments
• continuous observation to ensure the patient’s safety
• keeping the patient temporarily removed from his community to prevent irreversible social and economic harms.
However, a challenge with hospitalization is third-party payers’ influence on a patient’s length of stay, which may lead to rapid medication changes that may not be clinically ideal.
At each outpatient visit, explore with the patient and family emerging symptoms that could justify involuntary hospitalization. Document whether you recommended inpatient hospitalization, the patient’s response to the recommendation, that you are aware and have considered the risks associated with outpatient care, and that you have discussed these risks with the patient and family.
For patients well-known to the psychiatrist, a history of dangerous mania may lead him (her) to strongly recommend hospitalization, whereas a pre-existing therapeutic alliance and no current or distant history of dangerous mania may lead the clinician to look for alternatives to inpatient care. Concomitant drug or alcohol use may increase the likelihood of mania becoming dangerous, making outpatient treatment ill-advised and riskier for everyone involved.
In exchange for agreeing to provide outpatient care for mania, it often is helpful to negotiate with the patient and family a threshold level of symptoms or behavior that will result in the patient agreeing to voluntary hospitalization (Table 1). Such an agreement can include stopping outpatient treatment if the patient does not improve significantly after 2 or 3 weeks or develops psychotic symptoms. The negotiation also can include partial hospitalization as an option, so long as the patient’s mania continues to be non-dangerous.
Obtaining pretreatment blood work can help a clinician determine whether a medication is safe to prescribe and establish causality if laboratory abnormalities arise after treatment begins. Ideally, the psychiatrist should follow consensus guidelines developed by the International Society for Bipolar Disorders4 or the American Psychiatric Association (APA)5 and order appropriate laboratory tests before prescribing anti-manic medications. Determine the pregnancy status of female patients of child-bearing age before prescribing a potentially teratogenic medication, especially because mania is associated with increased libido.6
Manic patients might be too disorganized to follow up with recommendations for laboratory testing, or could wait several days before completing blood work. Although not ideal, to avoid delaying treatment, a clinician might need to prescribe medication at the initial office visit, without pretreatment laboratory results. When the patient is more organized, complete the blood work. Keeping home pregnancy tests in the office can help rule out pregnancy before prescribing medication.
Medication. Meta-analyses have established the efficacy of mood stabilizers and antipsychotics for treating mania,7,8 and several consensus guidelines have incorporated these findings into treatment algorithms.9
For a patient already taking medications recommended by the guidelines, assess treatment adherence during the initial interview by questioning the patient and family. When the logistics of phlebotomy permit, obtaining the blood level of psychotropics can show the presence of any detectable drug concentration, which demonstrates that the patient has taken the medication recently.
If there is no evidence of nonadherence, an initial step might be to increase the dosage of the antipsychotic or mood stabilizer that the patient is already taking, ensuring that the dosage is optimized based on FDA indications and clinical trials data. The recommended rate of dosage adjustments differs among medications; however, optimal dosing should be reached quickly because a World Federation of Societies of Biological Psychiatry task force recommends that a mania treatment trial not exceed 2 weeks.10
Dosage increases can be made at weekly visits or sooner, based on treatment response and tolerability. If there is no benefit after optimizing the dosage, the next step would be to add a mood stabilizer to a second-generation antipsychotic (SGA), or vice versa to promote additive or synergistic medication effects.11 Switching one medication for the other should be avoided unless there are tolerability concerns.
For a patient who is not taking any medications, select a treatment that balances rapid stabilization with long-term efficacy and tolerability. Table 2 lists FDA-approved treatments for mania. Lamotrigine provides prophylactic efficacy with few associated risks, but it has no anti-manic effects and would be a poor choice for most actively manic patients. Most studies indicate that antipsychotics work faster than lithium at the 1-week mark; however, this may be a function of the lithium titration schedule followed in the protocols, the severity of mania among enrolled patients, the inclusion of typically non-responsive manic patients (eg, mixed) in the analysis, and the antipsychotic’s sedative potential relative to lithium. Although the anti-manic and prophylactic potential of lithium and valproate might make them an ideal first-line option, antipsychotics could stabilize a manic patient faster, especially if agitation is present.12,13
Breaking mania quickly is important when treating patients in the outpatient setting. In these situations, a reasonable choice is to prescribe a SGA, because of their rapid onset of effect, low potential for switch to depression, and utility in treating classic, mixed, or psychotic mania.10 Oral loading of valproate (20 mg/kg) is another option. An inpatient study that used an oral-loading strategy demonstrated a similar time to response as olanzapine,14 in contrast to an inpatient15 and an outpatient study16 that employed a standard starting dosage for each patient and led to slower improvement compared with olanzapine.
SGAs should be dosed moderately and lower than if the patient were hospitalized, to avoid alienating the patient from treatment by causing intolerable side effects. In particular, patients and their families should be warned about immediate risks, such as orthostasis or extrapyramidal symptoms. Although treatment guidelines recommend combination therapy as a possible first-line option,9 in the outpatient setting, monotherapy with an optimally dosed, rapid-acting agent is preferred to promote medication adherence and avoid potentially dangerous sedation. Manic patients experience increased distractibility and verbal memory and executive function impairments that can interfere with medication adherence.17 Therefore, patients are more likely to follow a simpler regimen. If SGA or valproate monotherapy does not control mania, begin combination treatment with a mood stabilizer and SGA. If the patient experiences remission with SGA monotherapy, the risks and benefits of maintaining the SGA vs switching to a mood stabilizer can be discussed.
Provide medication “as needed” for agitation—additional SGA dosing or a benzodiazepine—and explain to family members when their use is warranted. Benzodiazepines can provide short-term benefits for manic patients: anxiety relief, sedation, and anti-manic efficacy as monotherapy18-20 and in combination with other medications.21 Studies showing monotherapy efficacy employed high dosages of benzodiazepines (lorazepam mean dosage, 14 mg/d; clonazepam mean dosage, 13 mg/d)19 and high dosages of antipsychotics as needed,18,20 and often were associated with excessive sedation and ataxia.18,19 This makes benzodiazepine monotherapy a potentially dangerous approach for outpatient treatment of mania. IM lorazepam treated manic agitation less quickly than IM olanzapine, suggesting that SGAs are preferable in the outpatient setting because rapid control of agitation is crucial.22 If prescribed, a trusted family member should dispense benzodiazepines to the patient to minimize misuse because of impulsivity, distractibility, desperation to sleep, or pleasure seeking.
SGAs have the benefit of sedation but occasionally additional sleep medications are required. Benzodiazepine receptor agonists (BzRAs), such as zolpidem, eszopiclone, and zaleplon, should be used with caution. Although these medicines are effective in treating insomnia in individuals with primary insomnia23 and major depression,24 they have not been studied in manic patients. The decreased need for sleep in mania is phenomenologically25 and perhaps biologically different than insomnia in major depression.26 Therefore, mania-associated sleep disturbance might not respond to BZRAs. BzRAs also might induce somnambulism and other parasomnias,27 especially when used in combination with psychotropics, such as valproate28; it is unclear if the manic state itself increases this risk further. Sedating antihistamines with anticholinergic blockade, such as diphenhydramine and low dosages (<100 mg/d) of quetiapine, are best used only in combination with anti-manic medications because of putative link between anticholinergic blockade and manic induction.29 Less studied but safer options include novel anticonvulsants (gabapentin, pregabalin), melatonin, and melatonin receptor agonists. Sedating antidepressants, such as mirtazapine and trazodone, should be avoided.25
Important adjunctive treatment steps include discontinuing all pro-manic agents, including antidepressants, stimulants, and steroids, and discouraging use of caffeine, energy drinks, illicit drugs, and alcohol. The patient should return for office visits at least weekly, and possibly more frequently, depending on severity. Telephone check-in calls between scheduled visits may be necessary until the mania is broken.
Psychotherapy. Other than supportive therapy and psychoeducation, other forms of psychotherapy during mania are not indicated. Psychotherapy trials in bipolar disorder do not inform anti-manic efficacy because few have enrolled acutely manic patients and most report long-term benefits rather than short-term efficacy for the index manic episode.30 Educate patients about the importance of maintaining regular social rhythms and taking medication as prescribed. Manic patients might not be aware that they are acting differently during manic episodes, therefore efforts to improve the patient’s insight are unlikely to succeed. More time should be spent emphasizing the importance of adherence to treatment and taking anti-manic medications as prescribed. This discussion can be enhanced by focusing on the medication’s potential to reduce the unpleasant symptoms of mania, including irritability, insomnia, anxiety, and racing thoughts. At the first visit, discuss setting boundaries with the patient to reduce mania-driven, intrusive phone calls. A patient might develop insight after mania has resolved and he (she) can appreciate social or economic harm that occurred while manic. This discussion might foster adherence to maintenance treatment. Advise your patient to limit activities that may increase stimulation and perpetuate the mania, such as exercise, parties, concerts, or crowded shopping malls. Also, recommend that your patient stop working temporarily, to reduce stress and prevent any manic-driven interactions that could result in job loss.
If your patient has an established relationship with a psychotherapist, discuss with the therapist the plan to initiate mania treatment in the outpatient setting and work as a collaborative team, assuming that the patient has granted permission to share information. Encourage the therapist to increase the frequency of sessions with the patient to enable greater monitoring of changes in the patient’s manic symptoms.
Family involvement
Family support is crucial when treating mania in the outpatient setting. Lacking insight and organization, manic patients require the “auxiliary” judgment of trusted family members to ensure treatment success. The family should identify a single person to act as the liaison between the family and the psychiatrist. The psychiatrist should instruct this individual to accompany the patient to each clinic visit and provide regular updates on the patient’s adherence to treatment, changes in symptoms, and any new behaviors that would justify involuntary hospitalization. The treatment plan should be clearly communicated to this individual to ensure that it is implemented correctly. Ideally, this individual would be someone who understands that bipolar disorder is a mental illness, who can tolerate the patient’s potential resentment of them for taking on this role, and who can influence the patient and the other family members to adhere to the treatment plan.
This family member also should watch the patient take medication to rule out nonadherence if the patient’s condition does not improve.
Provide extensive psychoeducation to the family (Table 3). Discuss these teaching points and their implications at length during the first visit and reinforce them at subsequent visits. Advise spouses that the acute manic period is not the time to make major decisions about their marriage or to engage in couple’s therapy. These options are better explored after the patient recovers from the manic episode.
Encourage the family to engage in mania harm-reduction techniques to the extent that the patient will allow (Table 4). In particular, they should hold onto their loved one’s credit cards and checkbook, and discourage the patient from making any major financial decisions until the mania has resolved. Additionally, patients should be relieved of childcare responsibilities during this period. If there are any child welfare safety concerns, the clinician will need to report this to authorities as required by local laws.
Advise family members or roommates to call emergency services and request a crisis intervention team, or to take the patient to an emergency room if he (she) makes verbal threats to harm themselves or others, is violent, or demonstrates behaviors that indicate that he is no longer able to care for himself. The psychiatrist should assist with completing Family and Medical Leave Act paperwork for family members who will monitor the patient at home, a work-excuse letter for the patient so he does not lose his job, and short-term disability paperwork to ensure income for the patient during the manic period.
These interventions can be challenging for the entire family system because they place family members in a paternalistic role and reduce the patient’s autonomy within the family. This is problematic when these role changes occur between spouses or between a patient-parent and his (her) children. Such changes typically need to be reversed over time and may require the help of a family or couple’s therapist. To support the psychological health of the patient’s family, refer them to the National Alliance on Mental Illness for family support groups or to individual psychotherapists.
Outpatient management can be rewarding
For “non-dangerous” manic patients who cannot be hospitalized involuntarily and refuse full or partial hospitalization, a psychiatrist must choose between beginning treatment in the clinic and referring the patient to another provider. The latter option is consistent with the APA’s ethical guidelines,31 but must be done appropriately to avoid legal liability.32 This decision may disappoint a family desperate to see their loved one recover quickly and may leave them feeling betrayed by the mental health system. On the other hand, choosing to treat mania in the outpatient setting can be rewarding when resolution of mania restores the family’s homeostasis.
To achieve this outcome, the outpatient psychiatrist must engage the patient’s family to ensure that the patient adheres to the treatment plan and monitor for potentially dangerous behavior. The psychiatrist also must use his knowledge of mood symptoms, cognitive impairments, and the psychological experience of manic patients to create a safe and effective treatment strategy that the patient and family can implement.
Because of mania’s unpredictability and destructive potential, psychiatrists who agree to treat manic patients as outpatients should be familiar with their state’s statutes and case law that pertain to the refusal to accept a new patient, patient abandonment, involuntary hospitalization, confidentiality, and mandatory reporting. They also should seek clinical or legal consultation if they feel overwhelmed or uncertain about the safest and most legally sound approach.
Bottom Line
Treating mania in the outpatient setting is risky but can be accomplished in select patients with the help of the patient’s family and a strategy that integrates evidence-based pharmacotherapeutic and psychotherapeutic strategies. Because manic patients could display dangerous behavior, be familiar with your state’s laws regarding involuntary commitment, patient abandonment, and mandatory reporting.
Related Resources
• National Alliance on Mental Illness. www.NAMI.org.
• Depression and Bipolar Support Alliance. www.DBSAlliance.org.
Drug Brand Names
Aripiprazole • Abilify Mirtazapine • Remeron
Asenapine • Saphris Olanzapine • Zyprexa
Carbamazepine • Equetro, Tegretol Pregabalin • Lyrica
Chlorpromazine • Thorazine Quetiapine • Seroquel
Clonazepam • Klonopin Risperidone • Risperdal
Diphrenhydramine • Benadryl Trazodone • Desyrel
Eszopiclone • Lunesta Valproate • Divalproex
Gabapentin • Neurontin Zaleplon • Sonata
Lamotrigine • Lamictal Ziprasidone • Geodon
Lithium • Eskalith, Lithobid Zolpidem • Ambien
Lorazepam • Ativan
Acknowledgement
The authors thank Peter Ash, MD, for carefully reviewing this manuscript and providing feedback.
Disclosures
Dr. Rakofsky receives research or grant support from Takeda. Dr. Dunlop receives research or grant support from Forest, GlaxoSmithKline, and Otsuka.
1. Diagnostic and statistical manual of mental disorders. 5th ed. Washington, DC: American Psychiatric Association; 2013.
2. Cassidy F, Murry E, Forest K, et al. Signs and symptoms of mania in pure and mixed episodes. J Affect Disord. 1998;50(2-3):187-201.
3. Yen CF, Chen CS, Ko CH, et al. Changes in insight among patients with bipolar I disorder: a 2-year prospective study. Bipolar Disord. 2007;9(3):238-242.
4. Ng F, Mammen OK, Wilting I, et al. The International Society for Bipolar Disorders (ISBD) consensus guidelines for the safety monitoring of bipolar disorder treatments. Bipolar Disord. 2009;11(6):559-595.
5. American Psychiatric Association. Practice guideline for the treatment of patients with bipolar disorder (revision). Am J Psychiatry. 2002;159(suppl 4):1-50.
6. Allison JB, Wilson WP. Sexual behavior of manic patients: a preliminary report. South Med J. 1960;53:870-874.
7. Cipriani A, Barbui C, Salanti G, et al. Comparative efficacy and acceptability of antimanic drugs in acute mania: a multiple-treatments meta-analysis. Lancet. 2011; 378(9799):1306-1315.
8. Yildiz A, Vieta E, Leucht S, et al. Efficacy of antimanic treatments: meta-analysis of randomized, controlled trials. Neuropsychopharmacology. 2011;36(2):375-389.
9. Nivoli AM, Murru A, Goikolea JM, et al. New treatment guidelines for acute bipolar mania: a critical review. J Affect Disord. 2012;140(2):125-141.
10. Grunze H, Vieta E, Goodwin GM, et al. The World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the biological treatment of bipolar disorders: update 2009 on the treatment of acute mania. World J Biol Psychiatry. 2009;10(2):85-116.
11. Tohen M, Chengappa KN, Suppes T, et al. Efficacy of olanzapine in combination with valproate or lithium in the treatment of mania in patients partially nonresponsive to valproate or lithium monotherapy. Arch Gen Psychiatry. 2002;59(1):62-69.
12. Tohen M, Jacobs TG, Feldman PD. Onset of action of antipsychotics in the treatment of mania. Bipolar Disord. 2000;2(3 pt 2):261-268.
13. Goikolea JM, Colom F, Capapey J, et al. Faster onset of antimanic action with haloperidol compared to second-generation antipsychotics. A meta-analysis of randomized clinical trials in acute mania. Eur Neuropsychopharmacol. 2013;23(4):305-316.
14. Zajecka JM, Weisler R, Sachs G, et al. A comparison of the efficacy, safety, and tolerability of divalproex sodium and olanzapine in the treatment of bipolar disorder. J Clin Psychiatry. 2002;63(12):1148-1155.
15. Tohen M, Baker RW, Altshuler LL, et al. Olanzapine versus divalproex in the treatment of acute mania. Am J Psychiatry. 2002;159(6):1011-1017.
16. Tohen M, Vieta E, Goodwin GM, et al. Olanzapine versus divalproex versus placebo in the treatment of mild to moderate mania: a randomized, 12-week, double-blind study. J Clin Psychiatry. 2008;69(11):1776-1789.
17. Martínez-Arán A, Vieta E, Reinares M, et al. Cognitive function across manic or hypomanic, depressed, and euthymic states in bipolar disorder. Am J Psychiatry. 2004; 161(2):262-270.
18. Edwards R, Stephenson U, Flewett T. Clonazepam in acute mania: a double blind trial. Aust N Z J Psychiatry. 1991;25(2):238-242.
19. Bradwejn J, Shriqui C, Koszycki D, et al. Double-blind comparison of the effects of clonazepam and lorazepam in acute mania. J Clin Psychopharmacol. 1990;10(6):403-408.
20. Clark HM, Berk M, Brook S. A randomized controlled single blind study of the efficacy of clonazepam and lithium in the treatment of acute mania. Human Psychopharmacology: Clinical and Experimental. 1997;12(4):325-328.
21. Lenox RH, Newhouse PA, Creelman WL, et al. Adjunctive treatment of manic agitation with lorazepam versus haloperidol: a double-blind study. J Clin Psychiatry. 1992;53(2):47-52.
22. Meehan K, Zhang F, David S, et al. A double-blind, randomized comparison of the efficacy and safety of intramuscular injections of olanzapine, lorazepam, or placebo in treating acutely agitated patients diagnosed with bipolar mania. J Clin Psychopharmacol. 2001;21(4):389-397.
23. Huedo-Medina TB, Kirsch I, Middlemass J, et al. Effectiveness of non-benzodiazepine hypnotics in treatment of adult insomnia: meta-analysis of data submitted to the Food and Drug Administration. BMJ. 2012;345:e8343. doi: 10.1136/bmj.e8343.
24. Fava M, Asnis GM, Shrivastava RK, et al. Improved insomnia symptoms and sleep-related next-day functioning in patients with comorbid major depressive disorder and insomnia following concomitant zolpidem extended-release 12.5 mg and escitalopram treatment: a randomized controlled trial. J Clin Psychiatry. 2011;72(7):914-928.
25. Plante DT, Winkelman JW. Sleep disturbance in bipolar disorder: therapeutic implications. Am J Psychiatry. 2008;165(7):830-843.
26. Linkowski P, Kerkhofs M, Rielaert C, et al. Sleep during mania in manic-depressive males. Eur Arch Psychiatry Neurol Sci. 1986;235(6):339-341.
27. Poceta JS. Zolpidem ingestion, automatisms, and sleep driving: a clinical and legal case series. J Clin Sleep Med. 2011;7(6):632-638.
28. Sattar SP, Ramaswamy S, Bhatia SC, et al. Somnambulism due to probable interaction of valproic acid and zolpidem. Ann Pharmacother. 2003;37(10):1429-1433.
29. Rybakowski JK, Koszewska I, Puzynski S. Anticholinergic mechanisms: a forgotten cause of the switch process in bipolar disorder [Comment on: The neurolobiology of the switch process in bipolar disorder: a review. J Clin Psychiatry. 2010]. J Clin Psychiatry. 2010;71(12):1698-1699; author reply 1699-1700.
30. Miklowitz DJ. Adjunctive psychotherapy for bipolar disorder: state of the evidence. Am J Psychiatry. 2008;165(11):1408-1419.
31. American Psychiatric Association. The principles of medical ethics with annotations especially applicable to psychiatry. Arlington, VA: American Psychiatric Association; 2013.
32. Simon RI, Shuman DW. Clinical manual of psychiatry and law. Arlington, VA: American Psychiatric Publishing; 2007:17-36.
1. Diagnostic and statistical manual of mental disorders. 5th ed. Washington, DC: American Psychiatric Association; 2013.
2. Cassidy F, Murry E, Forest K, et al. Signs and symptoms of mania in pure and mixed episodes. J Affect Disord. 1998;50(2-3):187-201.
3. Yen CF, Chen CS, Ko CH, et al. Changes in insight among patients with bipolar I disorder: a 2-year prospective study. Bipolar Disord. 2007;9(3):238-242.
4. Ng F, Mammen OK, Wilting I, et al. The International Society for Bipolar Disorders (ISBD) consensus guidelines for the safety monitoring of bipolar disorder treatments. Bipolar Disord. 2009;11(6):559-595.
5. American Psychiatric Association. Practice guideline for the treatment of patients with bipolar disorder (revision). Am J Psychiatry. 2002;159(suppl 4):1-50.
6. Allison JB, Wilson WP. Sexual behavior of manic patients: a preliminary report. South Med J. 1960;53:870-874.
7. Cipriani A, Barbui C, Salanti G, et al. Comparative efficacy and acceptability of antimanic drugs in acute mania: a multiple-treatments meta-analysis. Lancet. 2011; 378(9799):1306-1315.
8. Yildiz A, Vieta E, Leucht S, et al. Efficacy of antimanic treatments: meta-analysis of randomized, controlled trials. Neuropsychopharmacology. 2011;36(2):375-389.
9. Nivoli AM, Murru A, Goikolea JM, et al. New treatment guidelines for acute bipolar mania: a critical review. J Affect Disord. 2012;140(2):125-141.
10. Grunze H, Vieta E, Goodwin GM, et al. The World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the biological treatment of bipolar disorders: update 2009 on the treatment of acute mania. World J Biol Psychiatry. 2009;10(2):85-116.
11. Tohen M, Chengappa KN, Suppes T, et al. Efficacy of olanzapine in combination with valproate or lithium in the treatment of mania in patients partially nonresponsive to valproate or lithium monotherapy. Arch Gen Psychiatry. 2002;59(1):62-69.
12. Tohen M, Jacobs TG, Feldman PD. Onset of action of antipsychotics in the treatment of mania. Bipolar Disord. 2000;2(3 pt 2):261-268.
13. Goikolea JM, Colom F, Capapey J, et al. Faster onset of antimanic action with haloperidol compared to second-generation antipsychotics. A meta-analysis of randomized clinical trials in acute mania. Eur Neuropsychopharmacol. 2013;23(4):305-316.
14. Zajecka JM, Weisler R, Sachs G, et al. A comparison of the efficacy, safety, and tolerability of divalproex sodium and olanzapine in the treatment of bipolar disorder. J Clin Psychiatry. 2002;63(12):1148-1155.
15. Tohen M, Baker RW, Altshuler LL, et al. Olanzapine versus divalproex in the treatment of acute mania. Am J Psychiatry. 2002;159(6):1011-1017.
16. Tohen M, Vieta E, Goodwin GM, et al. Olanzapine versus divalproex versus placebo in the treatment of mild to moderate mania: a randomized, 12-week, double-blind study. J Clin Psychiatry. 2008;69(11):1776-1789.
17. Martínez-Arán A, Vieta E, Reinares M, et al. Cognitive function across manic or hypomanic, depressed, and euthymic states in bipolar disorder. Am J Psychiatry. 2004; 161(2):262-270.
18. Edwards R, Stephenson U, Flewett T. Clonazepam in acute mania: a double blind trial. Aust N Z J Psychiatry. 1991;25(2):238-242.
19. Bradwejn J, Shriqui C, Koszycki D, et al. Double-blind comparison of the effects of clonazepam and lorazepam in acute mania. J Clin Psychopharmacol. 1990;10(6):403-408.
20. Clark HM, Berk M, Brook S. A randomized controlled single blind study of the efficacy of clonazepam and lithium in the treatment of acute mania. Human Psychopharmacology: Clinical and Experimental. 1997;12(4):325-328.
21. Lenox RH, Newhouse PA, Creelman WL, et al. Adjunctive treatment of manic agitation with lorazepam versus haloperidol: a double-blind study. J Clin Psychiatry. 1992;53(2):47-52.
22. Meehan K, Zhang F, David S, et al. A double-blind, randomized comparison of the efficacy and safety of intramuscular injections of olanzapine, lorazepam, or placebo in treating acutely agitated patients diagnosed with bipolar mania. J Clin Psychopharmacol. 2001;21(4):389-397.
23. Huedo-Medina TB, Kirsch I, Middlemass J, et al. Effectiveness of non-benzodiazepine hypnotics in treatment of adult insomnia: meta-analysis of data submitted to the Food and Drug Administration. BMJ. 2012;345:e8343. doi: 10.1136/bmj.e8343.
24. Fava M, Asnis GM, Shrivastava RK, et al. Improved insomnia symptoms and sleep-related next-day functioning in patients with comorbid major depressive disorder and insomnia following concomitant zolpidem extended-release 12.5 mg and escitalopram treatment: a randomized controlled trial. J Clin Psychiatry. 2011;72(7):914-928.
25. Plante DT, Winkelman JW. Sleep disturbance in bipolar disorder: therapeutic implications. Am J Psychiatry. 2008;165(7):830-843.
26. Linkowski P, Kerkhofs M, Rielaert C, et al. Sleep during mania in manic-depressive males. Eur Arch Psychiatry Neurol Sci. 1986;235(6):339-341.
27. Poceta JS. Zolpidem ingestion, automatisms, and sleep driving: a clinical and legal case series. J Clin Sleep Med. 2011;7(6):632-638.
28. Sattar SP, Ramaswamy S, Bhatia SC, et al. Somnambulism due to probable interaction of valproic acid and zolpidem. Ann Pharmacother. 2003;37(10):1429-1433.
29. Rybakowski JK, Koszewska I, Puzynski S. Anticholinergic mechanisms: a forgotten cause of the switch process in bipolar disorder [Comment on: The neurolobiology of the switch process in bipolar disorder: a review. J Clin Psychiatry. 2010]. J Clin Psychiatry. 2010;71(12):1698-1699; author reply 1699-1700.
30. Miklowitz DJ. Adjunctive psychotherapy for bipolar disorder: state of the evidence. Am J Psychiatry. 2008;165(11):1408-1419.
31. American Psychiatric Association. The principles of medical ethics with annotations especially applicable to psychiatry. Arlington, VA: American Psychiatric Association; 2013.
32. Simon RI, Shuman DW. Clinical manual of psychiatry and law. Arlington, VA: American Psychiatric Publishing; 2007:17-36.
Depressed, suicidal, and brittle in her bones
CASE Broken down
Ms. E, age 20, is a college student who has had major depressive disorder for several years and a genetic bone disease (osteogenesis imperfecta, mixed type III and IV). She presents with depression, anxiety, and suicidal ideation. She reports recent worsening of her depressive symptoms, including anhedonia, excessive sleep, difficulty concentrating, and feeling overwhelmed, hopeless, and worthless. She also describes frequent thoughts of suicide with the plan of putting herself in oncoming traffic, although she has no history of suicide attempts.
Previously, her primary care physician prescribed lorazepam, 0.5 mg, as needed for anxiety, and sertraline, 100 mg/d, for depression and anxiety. She experienced only partial improvement in symptoms, however.
In addition to depressive symptoms, Ms. E describes manic symptoms lasting for as long as 3 to 5 days, including decreased need for sleep, increased energy, pressured speech, racing thoughts, distractibility, spending excessive money on cosmetics, and risking her safety—given her skeletal disorder— by participating in high-impact stage-combat classes. She denies auditory and visual hallucinations, homicidal ideation, and delusions.
The medical history is significant for osteogenesis imperfecta, which has caused 62 fractures and required 16 surgeries. Ms. E is a theater major who, despite her short stature and wheelchair use, reports enjoying her acting career and says she does not feel demoralized by her medical condition. She describes overcoming her physical disabilities with pride and confidence. However, her recent worsening mood symptoms have left her unable to concentrate and feeling overwhelmed with school.
Ms. E is voluntarily admitted to an inpatient psychiatric unit with a diagnosis of bipolar I disorder with rapid cycling, most recent episode mixed. Because of her bone fragility, the treatment team considers what would be an appropriate course of drug treatment to control bipolar symptoms while minimizing risk of bone loss.
Which medications are associated with decreased bone mineral density?
a) citalopram
b) haloperidol
c) carbamazepine
d) paliperidone
e) all of the above
The authors’ observations
Osteogenesis imperfecta is a genetic condition caused by mutations in genes implicated in collagen production. As a result, bones are brittle and prone to fracture. Different classes of psychotropics have been shown to increase risk of bone fractures through a variety of mechanisms. Clinicians often must choose appropriate pharmacotherapy for patients at high risk of fracture, including postmenopausal women, older patients, malnourished persons, and those with hormonal deficiencies leading to osteoporosis.
To assist our clinical decision-making, we reviewed the literature to establish appropriate management of a patient with increased bone fragility and new-onset bipolar disorder. We considered all classes of medications used to treat bipolar disorder, including antipsychotics, antidepressants, lithium, and anticonvulsants.
Antipsychotics
In population-based studies, prolactin-elevating antipsychotics have been associated with decreased bone mineral density and increased risk of fracture.1 Additional studies on geriatric and non-geriatric populations have supported these findings.2,3
The mechanism through which fracture risk is increased likely is related to antipsychotics’ effect on serum prolactin and cortisol levels. Antipsychotics act as antagonists on D2 receptors in the hypothalamic tubero-infundibular pathway, therefore preventing inhibition of prolactin. Long-term elevation in serum prolactin can cause loss of bone mineral density through secondary hypogonadism and direct effects on target tissues. Additional modifying factors include smoking and estrogen use.
The degree to which antipsychotics increase fracture risk might be related to the degree of serum prolactin elevation.4 Antipsychotics previously have been grouped by the degree of prolactin elevation, categorizing them as high, medium, and low or no potential to elevate serum prolactin.4 Based on this classification, typical antipsychotics, risperidone, and paliperidone have the highest potential to elevate prolactin. Accordingly, antipsychotics with the lowest fracture risk are those that have the lowest risk of serum prolactin elevation: ziprasidone, asenapine, quetiapine, and clozapine. Aripiprazole may lower prolactin in some patients. This is supported by studies noting reduced bone mineral density5,6 and increased risk of fracture1 with high-potential vs low- or no-potential antipsychotics. Because of these findings, it is crucial to consider the potential risk of prolactin elevation when treating patients at increased risk of fracture. Providers should consider low/no potential antipsychotic medications before considering those with medium or high potential (Table).
Antidepressants
In a meta-analysis, antidepressants were shown to increase fracture risk by 70% to 90%.2 However, the relative risk varies by antidepressant class. Several studies have shown that selective serotonin reuptake inhibitors (SSRIs) are associated with a higher risk of fracture compared with tricyclic antidepressants (TCAs).7 In addition, antidepressants with a high affinity for the serotonin transporter, including citalopram, fluoxetine, fluvoxamine, paroxetine, sertraline, and imipramine, have been associated with greater risk of osteoporotic fracture compared with those with low affinity.8
The mechanisms by which antidepressants increase fracture risk are complex, although the strongest evidence implicates a direct effect on bone metabolism via the 5-HTT receptor. This receptor, found on osteoblasts and osteoclasts, plays an important role in bone metabolism; it is through this receptor that SSRIs might inhibit osteoblasts and promote osteoclast activity, thereby disrupting bone microarchitecture. Additional studies are needed to further describe the mechanism of the association among antidepressants, bone mineral density, and fracture risk.
Fracture risk is associated with duration of use rather than dosage. Population-based studies show a higher fracture risk for new users of TCAs compared with continuous users, and the risk of fracture with SSRIs seems to increase slightly over time.9 No association has been identified between fracture risk and antidepressant dosage. According to the literature, drugs with low affinity for the serotonin transporter, such as maprotiline and mirtazapine, likely are the safest antidepressants for patients at increased risk of fracture. Options also include other TCAs and any antidepressant with low affinity for the serotonin receptor.7,8
Lithium
Studies on lithium and bone mineral density have shown mixed results. Older studies found that lithium had a negative or no effect on bone mineral density or the parathyroid hormone level.10 More recent investigations, however, suggest that the drug has a protective effect on bone mineral density, although this has not been replicated in all studies.
In a mouse model, lithium has been shown to enhance bone formation and improve bone mass, at least in part by activation of the Wnt signaling pathway through an inhibitory effect on glycogen synthase kinase-3β.11 In humans, lithium-treated adults had lower serum alkaline phosphate, osteocalcin, and C-telopeptide levels compared with controls, suggesting a state of decreased bone remodeling and increased turnover.12 There is a paucity of clinical data on the effect of lithium on fracture risk. Additional studies are necessary to elucidate lithium’s mechanism on bone mineral density and determine the magnitude of the clinical effect.
Anticonvulsants
The association among anticonvulsants, decreased bone mineral density, and increased risk of fracture is well-established in the literature.13 However, causality is difficult to determine, because many studies were of patients with a seizure disorder, who often have additional risk factors for fracture, including seizure-related trauma, drowsiness, and slowed reflexes.
Mechanisms through which anticonvulsants increase fracture risk include increased bone resorption, secondary hypoparathyroidism, and pseudohypoparathyroidism. Markers of bone resorption were elevated in patients receiving an antiepileptic.14 This effect might be enhanced by co-administration of cytochrome P450 (CYP450) enzyme-inducing anticonvulsants and CYP450 enzyme-inhibiting medications, such as valproate. Long-term treatment with valproate may produce reduction of bone mass and increased risk of fractures; however, other studies disagree with this finding.15
In addition to CYP450-inducing effects, phenytoin, carbamezapine, and phenobarbital can increase catabolism of vitamin D, which is associated with osteomalacia.14 This results in decreased intestinal absorption of calcium, hypocalcemia, and secondary hyperparathyroidism, which also increases fracture risk. Anticonvulsants also might increase resistance to pseudohypoparathyroidism and inhibit calcitonin secretion.
Lamotrigine has not been shown to interfere with bone accrual16 and may be a safer mood stabilizer for patients at high risk of fracture. For patients at increased risk of fracture, it is important to select an anticonvulsant wisely to minimize fracture risk.
How would you treat Ms. E during her hospitalization for bipolar disorder?
a) carbamazepine
b) lithium
c) risperidone
d) mirtazapine
TREATMENT Minimizing polypharmacy
Because many pharmacotherapeutic options for managing bipolar disorder can increase the risk of fracture, clinicians must be aware of the relative risk of each class of medication and each individual drug. We initiated lithium, 300 mg, 3 times a day, to stabilize Ms. E’s mood. Although clinical data are inconclusive regarding lithium’s effect on fracture risk, we felt that the benefit of acute mood stabilization outweighed the risk of decreased bone mineral index.
We selected aripiprazole, 10 mg/d, as an adjunctive treatment because of its minimal effect on serum prolactin levels.4 We considered prescribing an antidepressant but decided against it because we were concerned about manic switching.
Polypharmacy is another important consideration for Ms. E. Several studies have identified polypharmacy, particularly with antipsychotics, as an independent risk factor for fracture.3 Therefore, we sought to minimize the number of medications Ms. E receives. Although lithium monotherapy is an option, we thought that her mood symptoms were severe enough that the risk of inadequately treating her bipolar symptoms outweighed the additional risk of fracture from dual therapy with lithium and aripiprazole. Untreated or inadequately treated depression is associated with a higher fracture risk. Therefore, we avoided prescribing >2 medications to mitigate any excessive risk of fracture from polypharmacy.
Bottom Line
Different classes of medications—antipsychotics, anticonvulsants, antidepressants, and lithium—used for treating bipolar disorder have been shown to increase risk of bone fracture through a variety of mechanisms. Anticonvulsants and prolactin-elevating antipsychotics are associated with increased fracture risk; evidence on lithium is mixed. Fracture risk with antidepressants is associated with duration of use, rather than dosage.
Disclosures
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Howard L, Kirkwood G, Leese M. Risk of hip fracture in patients with a history of schizophrenia. Br J Psychiatry. 2007;190:129-134.
2. Takkouche B, Montes-Martínez A, Gill SS, et al. Psychotropic medications and the risk of fracture: a meta-analysis. Drug Saf. 2007;30(2):171-184.
3. Sørensen HJ, Jensen SO, Nielsen J. Schizophrenia, antipsychotics and risk of hip fracture: a population-based analysis. Eur Neuropsychopharmacol. 2013;23(8):872-878.
4. Rahman T, Clevenger CV, Kaklamani V, et al. Antipsychotic treatment in breast cancer patients. Am J Psychiatry. 2014;171(6):616-621.
5. Bilici M, Cakirbay H, Guler M, et al. Classical and atypical neuroleptics, and bone mineral density, in patients with schizophrenia. Int J Neurosci. 2002;112(7):817-828.
6. Becker D, Liver O, Mester R, et al. Risperidone, but not olanzapine, decreases bone mineral density in female premenopausal schizophrenia patients. J Clin Psychiatry. 2003;64(7):761-766.
7. Bolton JM, Metge C, Lix L, et al. Fracture risk from psychotropic medications: a population-based analysis. J Clin Psychopharmacol. 2008;28(4):384-391.
8. Verdel BM, Souverein PC, Egberts TC, et al. Use of antidepressant drugs and risk of osteoporotic and non-osteoporotic fractures. Bone. 2010;47(3):604-609.
9. Diem SJ, Ruppert K, Cauley JA. Rates of bone loss among women initiating antidepressant medication use in midlife. J Clin Endocrinol Metab. 2013;(11):4355-4363.
10. Plenge P, Rafaelsen OJ. Lithium effects on calcium, magnesium and phosphate in man: effects on balance, bone mineral content, faecal and urinary excretion. Acta Psychiatr Scand. 1982;66(5):361-373.
11. Clément-Lacroix P, Ai M, Morvan F, et al. Lrp5-independent activation of Wnt signaling by lithium chloride increases bone formation and bone mass in mice. Proc Natl Acad Sci U S A. 2005;102(48):17406-17411.
12. Zamani A, Omrani GR, Nasab MM. Lithium’s effect on bone mineral density. Bone. 2009;44(2):331-334.
13. Swanton J, Simister R, Altmann D, et al. Bone mineral density in institutionalised patients with refractory epilepsy. Seizure. 2007;16(6):538-541.
14. Pack AM, Morrell MJ. Epilepsy and bone health in adults. Epilepsy Behav. 2004;5(suppl 2):S24-S29.
15. Pack AM. Bone disease in epilepsy. Curr Neurol Neurosci Rep. 2004;4(4):329-334.
16. Sheth RD, Hermann BP. Bone mineral density with lamotrigine monotherapy for epilepsy. Pediatr Neurol. 2007;37(4):250-254.
CASE Broken down
Ms. E, age 20, is a college student who has had major depressive disorder for several years and a genetic bone disease (osteogenesis imperfecta, mixed type III and IV). She presents with depression, anxiety, and suicidal ideation. She reports recent worsening of her depressive symptoms, including anhedonia, excessive sleep, difficulty concentrating, and feeling overwhelmed, hopeless, and worthless. She also describes frequent thoughts of suicide with the plan of putting herself in oncoming traffic, although she has no history of suicide attempts.
Previously, her primary care physician prescribed lorazepam, 0.5 mg, as needed for anxiety, and sertraline, 100 mg/d, for depression and anxiety. She experienced only partial improvement in symptoms, however.
In addition to depressive symptoms, Ms. E describes manic symptoms lasting for as long as 3 to 5 days, including decreased need for sleep, increased energy, pressured speech, racing thoughts, distractibility, spending excessive money on cosmetics, and risking her safety—given her skeletal disorder— by participating in high-impact stage-combat classes. She denies auditory and visual hallucinations, homicidal ideation, and delusions.
The medical history is significant for osteogenesis imperfecta, which has caused 62 fractures and required 16 surgeries. Ms. E is a theater major who, despite her short stature and wheelchair use, reports enjoying her acting career and says she does not feel demoralized by her medical condition. She describes overcoming her physical disabilities with pride and confidence. However, her recent worsening mood symptoms have left her unable to concentrate and feeling overwhelmed with school.
Ms. E is voluntarily admitted to an inpatient psychiatric unit with a diagnosis of bipolar I disorder with rapid cycling, most recent episode mixed. Because of her bone fragility, the treatment team considers what would be an appropriate course of drug treatment to control bipolar symptoms while minimizing risk of bone loss.
Which medications are associated with decreased bone mineral density?
a) citalopram
b) haloperidol
c) carbamazepine
d) paliperidone
e) all of the above
The authors’ observations
Osteogenesis imperfecta is a genetic condition caused by mutations in genes implicated in collagen production. As a result, bones are brittle and prone to fracture. Different classes of psychotropics have been shown to increase risk of bone fractures through a variety of mechanisms. Clinicians often must choose appropriate pharmacotherapy for patients at high risk of fracture, including postmenopausal women, older patients, malnourished persons, and those with hormonal deficiencies leading to osteoporosis.
To assist our clinical decision-making, we reviewed the literature to establish appropriate management of a patient with increased bone fragility and new-onset bipolar disorder. We considered all classes of medications used to treat bipolar disorder, including antipsychotics, antidepressants, lithium, and anticonvulsants.
Antipsychotics
In population-based studies, prolactin-elevating antipsychotics have been associated with decreased bone mineral density and increased risk of fracture.1 Additional studies on geriatric and non-geriatric populations have supported these findings.2,3
The mechanism through which fracture risk is increased likely is related to antipsychotics’ effect on serum prolactin and cortisol levels. Antipsychotics act as antagonists on D2 receptors in the hypothalamic tubero-infundibular pathway, therefore preventing inhibition of prolactin. Long-term elevation in serum prolactin can cause loss of bone mineral density through secondary hypogonadism and direct effects on target tissues. Additional modifying factors include smoking and estrogen use.
The degree to which antipsychotics increase fracture risk might be related to the degree of serum prolactin elevation.4 Antipsychotics previously have been grouped by the degree of prolactin elevation, categorizing them as high, medium, and low or no potential to elevate serum prolactin.4 Based on this classification, typical antipsychotics, risperidone, and paliperidone have the highest potential to elevate prolactin. Accordingly, antipsychotics with the lowest fracture risk are those that have the lowest risk of serum prolactin elevation: ziprasidone, asenapine, quetiapine, and clozapine. Aripiprazole may lower prolactin in some patients. This is supported by studies noting reduced bone mineral density5,6 and increased risk of fracture1 with high-potential vs low- or no-potential antipsychotics. Because of these findings, it is crucial to consider the potential risk of prolactin elevation when treating patients at increased risk of fracture. Providers should consider low/no potential antipsychotic medications before considering those with medium or high potential (Table).
Antidepressants
In a meta-analysis, antidepressants were shown to increase fracture risk by 70% to 90%.2 However, the relative risk varies by antidepressant class. Several studies have shown that selective serotonin reuptake inhibitors (SSRIs) are associated with a higher risk of fracture compared with tricyclic antidepressants (TCAs).7 In addition, antidepressants with a high affinity for the serotonin transporter, including citalopram, fluoxetine, fluvoxamine, paroxetine, sertraline, and imipramine, have been associated with greater risk of osteoporotic fracture compared with those with low affinity.8
The mechanisms by which antidepressants increase fracture risk are complex, although the strongest evidence implicates a direct effect on bone metabolism via the 5-HTT receptor. This receptor, found on osteoblasts and osteoclasts, plays an important role in bone metabolism; it is through this receptor that SSRIs might inhibit osteoblasts and promote osteoclast activity, thereby disrupting bone microarchitecture. Additional studies are needed to further describe the mechanism of the association among antidepressants, bone mineral density, and fracture risk.
Fracture risk is associated with duration of use rather than dosage. Population-based studies show a higher fracture risk for new users of TCAs compared with continuous users, and the risk of fracture with SSRIs seems to increase slightly over time.9 No association has been identified between fracture risk and antidepressant dosage. According to the literature, drugs with low affinity for the serotonin transporter, such as maprotiline and mirtazapine, likely are the safest antidepressants for patients at increased risk of fracture. Options also include other TCAs and any antidepressant with low affinity for the serotonin receptor.7,8
Lithium
Studies on lithium and bone mineral density have shown mixed results. Older studies found that lithium had a negative or no effect on bone mineral density or the parathyroid hormone level.10 More recent investigations, however, suggest that the drug has a protective effect on bone mineral density, although this has not been replicated in all studies.
In a mouse model, lithium has been shown to enhance bone formation and improve bone mass, at least in part by activation of the Wnt signaling pathway through an inhibitory effect on glycogen synthase kinase-3β.11 In humans, lithium-treated adults had lower serum alkaline phosphate, osteocalcin, and C-telopeptide levels compared with controls, suggesting a state of decreased bone remodeling and increased turnover.12 There is a paucity of clinical data on the effect of lithium on fracture risk. Additional studies are necessary to elucidate lithium’s mechanism on bone mineral density and determine the magnitude of the clinical effect.
Anticonvulsants
The association among anticonvulsants, decreased bone mineral density, and increased risk of fracture is well-established in the literature.13 However, causality is difficult to determine, because many studies were of patients with a seizure disorder, who often have additional risk factors for fracture, including seizure-related trauma, drowsiness, and slowed reflexes.
Mechanisms through which anticonvulsants increase fracture risk include increased bone resorption, secondary hypoparathyroidism, and pseudohypoparathyroidism. Markers of bone resorption were elevated in patients receiving an antiepileptic.14 This effect might be enhanced by co-administration of cytochrome P450 (CYP450) enzyme-inducing anticonvulsants and CYP450 enzyme-inhibiting medications, such as valproate. Long-term treatment with valproate may produce reduction of bone mass and increased risk of fractures; however, other studies disagree with this finding.15
In addition to CYP450-inducing effects, phenytoin, carbamezapine, and phenobarbital can increase catabolism of vitamin D, which is associated with osteomalacia.14 This results in decreased intestinal absorption of calcium, hypocalcemia, and secondary hyperparathyroidism, which also increases fracture risk. Anticonvulsants also might increase resistance to pseudohypoparathyroidism and inhibit calcitonin secretion.
Lamotrigine has not been shown to interfere with bone accrual16 and may be a safer mood stabilizer for patients at high risk of fracture. For patients at increased risk of fracture, it is important to select an anticonvulsant wisely to minimize fracture risk.
How would you treat Ms. E during her hospitalization for bipolar disorder?
a) carbamazepine
b) lithium
c) risperidone
d) mirtazapine
TREATMENT Minimizing polypharmacy
Because many pharmacotherapeutic options for managing bipolar disorder can increase the risk of fracture, clinicians must be aware of the relative risk of each class of medication and each individual drug. We initiated lithium, 300 mg, 3 times a day, to stabilize Ms. E’s mood. Although clinical data are inconclusive regarding lithium’s effect on fracture risk, we felt that the benefit of acute mood stabilization outweighed the risk of decreased bone mineral index.
We selected aripiprazole, 10 mg/d, as an adjunctive treatment because of its minimal effect on serum prolactin levels.4 We considered prescribing an antidepressant but decided against it because we were concerned about manic switching.
Polypharmacy is another important consideration for Ms. E. Several studies have identified polypharmacy, particularly with antipsychotics, as an independent risk factor for fracture.3 Therefore, we sought to minimize the number of medications Ms. E receives. Although lithium monotherapy is an option, we thought that her mood symptoms were severe enough that the risk of inadequately treating her bipolar symptoms outweighed the additional risk of fracture from dual therapy with lithium and aripiprazole. Untreated or inadequately treated depression is associated with a higher fracture risk. Therefore, we avoided prescribing >2 medications to mitigate any excessive risk of fracture from polypharmacy.
Bottom Line
Different classes of medications—antipsychotics, anticonvulsants, antidepressants, and lithium—used for treating bipolar disorder have been shown to increase risk of bone fracture through a variety of mechanisms. Anticonvulsants and prolactin-elevating antipsychotics are associated with increased fracture risk; evidence on lithium is mixed. Fracture risk with antidepressants is associated with duration of use, rather than dosage.
Disclosures
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
CASE Broken down
Ms. E, age 20, is a college student who has had major depressive disorder for several years and a genetic bone disease (osteogenesis imperfecta, mixed type III and IV). She presents with depression, anxiety, and suicidal ideation. She reports recent worsening of her depressive symptoms, including anhedonia, excessive sleep, difficulty concentrating, and feeling overwhelmed, hopeless, and worthless. She also describes frequent thoughts of suicide with the plan of putting herself in oncoming traffic, although she has no history of suicide attempts.
Previously, her primary care physician prescribed lorazepam, 0.5 mg, as needed for anxiety, and sertraline, 100 mg/d, for depression and anxiety. She experienced only partial improvement in symptoms, however.
In addition to depressive symptoms, Ms. E describes manic symptoms lasting for as long as 3 to 5 days, including decreased need for sleep, increased energy, pressured speech, racing thoughts, distractibility, spending excessive money on cosmetics, and risking her safety—given her skeletal disorder— by participating in high-impact stage-combat classes. She denies auditory and visual hallucinations, homicidal ideation, and delusions.
The medical history is significant for osteogenesis imperfecta, which has caused 62 fractures and required 16 surgeries. Ms. E is a theater major who, despite her short stature and wheelchair use, reports enjoying her acting career and says she does not feel demoralized by her medical condition. She describes overcoming her physical disabilities with pride and confidence. However, her recent worsening mood symptoms have left her unable to concentrate and feeling overwhelmed with school.
Ms. E is voluntarily admitted to an inpatient psychiatric unit with a diagnosis of bipolar I disorder with rapid cycling, most recent episode mixed. Because of her bone fragility, the treatment team considers what would be an appropriate course of drug treatment to control bipolar symptoms while minimizing risk of bone loss.
Which medications are associated with decreased bone mineral density?
a) citalopram
b) haloperidol
c) carbamazepine
d) paliperidone
e) all of the above
The authors’ observations
Osteogenesis imperfecta is a genetic condition caused by mutations in genes implicated in collagen production. As a result, bones are brittle and prone to fracture. Different classes of psychotropics have been shown to increase risk of bone fractures through a variety of mechanisms. Clinicians often must choose appropriate pharmacotherapy for patients at high risk of fracture, including postmenopausal women, older patients, malnourished persons, and those with hormonal deficiencies leading to osteoporosis.
To assist our clinical decision-making, we reviewed the literature to establish appropriate management of a patient with increased bone fragility and new-onset bipolar disorder. We considered all classes of medications used to treat bipolar disorder, including antipsychotics, antidepressants, lithium, and anticonvulsants.
Antipsychotics
In population-based studies, prolactin-elevating antipsychotics have been associated with decreased bone mineral density and increased risk of fracture.1 Additional studies on geriatric and non-geriatric populations have supported these findings.2,3
The mechanism through which fracture risk is increased likely is related to antipsychotics’ effect on serum prolactin and cortisol levels. Antipsychotics act as antagonists on D2 receptors in the hypothalamic tubero-infundibular pathway, therefore preventing inhibition of prolactin. Long-term elevation in serum prolactin can cause loss of bone mineral density through secondary hypogonadism and direct effects on target tissues. Additional modifying factors include smoking and estrogen use.
The degree to which antipsychotics increase fracture risk might be related to the degree of serum prolactin elevation.4 Antipsychotics previously have been grouped by the degree of prolactin elevation, categorizing them as high, medium, and low or no potential to elevate serum prolactin.4 Based on this classification, typical antipsychotics, risperidone, and paliperidone have the highest potential to elevate prolactin. Accordingly, antipsychotics with the lowest fracture risk are those that have the lowest risk of serum prolactin elevation: ziprasidone, asenapine, quetiapine, and clozapine. Aripiprazole may lower prolactin in some patients. This is supported by studies noting reduced bone mineral density5,6 and increased risk of fracture1 with high-potential vs low- or no-potential antipsychotics. Because of these findings, it is crucial to consider the potential risk of prolactin elevation when treating patients at increased risk of fracture. Providers should consider low/no potential antipsychotic medications before considering those with medium or high potential (Table).
Antidepressants
In a meta-analysis, antidepressants were shown to increase fracture risk by 70% to 90%.2 However, the relative risk varies by antidepressant class. Several studies have shown that selective serotonin reuptake inhibitors (SSRIs) are associated with a higher risk of fracture compared with tricyclic antidepressants (TCAs).7 In addition, antidepressants with a high affinity for the serotonin transporter, including citalopram, fluoxetine, fluvoxamine, paroxetine, sertraline, and imipramine, have been associated with greater risk of osteoporotic fracture compared with those with low affinity.8
The mechanisms by which antidepressants increase fracture risk are complex, although the strongest evidence implicates a direct effect on bone metabolism via the 5-HTT receptor. This receptor, found on osteoblasts and osteoclasts, plays an important role in bone metabolism; it is through this receptor that SSRIs might inhibit osteoblasts and promote osteoclast activity, thereby disrupting bone microarchitecture. Additional studies are needed to further describe the mechanism of the association among antidepressants, bone mineral density, and fracture risk.
Fracture risk is associated with duration of use rather than dosage. Population-based studies show a higher fracture risk for new users of TCAs compared with continuous users, and the risk of fracture with SSRIs seems to increase slightly over time.9 No association has been identified between fracture risk and antidepressant dosage. According to the literature, drugs with low affinity for the serotonin transporter, such as maprotiline and mirtazapine, likely are the safest antidepressants for patients at increased risk of fracture. Options also include other TCAs and any antidepressant with low affinity for the serotonin receptor.7,8
Lithium
Studies on lithium and bone mineral density have shown mixed results. Older studies found that lithium had a negative or no effect on bone mineral density or the parathyroid hormone level.10 More recent investigations, however, suggest that the drug has a protective effect on bone mineral density, although this has not been replicated in all studies.
In a mouse model, lithium has been shown to enhance bone formation and improve bone mass, at least in part by activation of the Wnt signaling pathway through an inhibitory effect on glycogen synthase kinase-3β.11 In humans, lithium-treated adults had lower serum alkaline phosphate, osteocalcin, and C-telopeptide levels compared with controls, suggesting a state of decreased bone remodeling and increased turnover.12 There is a paucity of clinical data on the effect of lithium on fracture risk. Additional studies are necessary to elucidate lithium’s mechanism on bone mineral density and determine the magnitude of the clinical effect.
Anticonvulsants
The association among anticonvulsants, decreased bone mineral density, and increased risk of fracture is well-established in the literature.13 However, causality is difficult to determine, because many studies were of patients with a seizure disorder, who often have additional risk factors for fracture, including seizure-related trauma, drowsiness, and slowed reflexes.
Mechanisms through which anticonvulsants increase fracture risk include increased bone resorption, secondary hypoparathyroidism, and pseudohypoparathyroidism. Markers of bone resorption were elevated in patients receiving an antiepileptic.14 This effect might be enhanced by co-administration of cytochrome P450 (CYP450) enzyme-inducing anticonvulsants and CYP450 enzyme-inhibiting medications, such as valproate. Long-term treatment with valproate may produce reduction of bone mass and increased risk of fractures; however, other studies disagree with this finding.15
In addition to CYP450-inducing effects, phenytoin, carbamezapine, and phenobarbital can increase catabolism of vitamin D, which is associated with osteomalacia.14 This results in decreased intestinal absorption of calcium, hypocalcemia, and secondary hyperparathyroidism, which also increases fracture risk. Anticonvulsants also might increase resistance to pseudohypoparathyroidism and inhibit calcitonin secretion.
Lamotrigine has not been shown to interfere with bone accrual16 and may be a safer mood stabilizer for patients at high risk of fracture. For patients at increased risk of fracture, it is important to select an anticonvulsant wisely to minimize fracture risk.
How would you treat Ms. E during her hospitalization for bipolar disorder?
a) carbamazepine
b) lithium
c) risperidone
d) mirtazapine
TREATMENT Minimizing polypharmacy
Because many pharmacotherapeutic options for managing bipolar disorder can increase the risk of fracture, clinicians must be aware of the relative risk of each class of medication and each individual drug. We initiated lithium, 300 mg, 3 times a day, to stabilize Ms. E’s mood. Although clinical data are inconclusive regarding lithium’s effect on fracture risk, we felt that the benefit of acute mood stabilization outweighed the risk of decreased bone mineral index.
We selected aripiprazole, 10 mg/d, as an adjunctive treatment because of its minimal effect on serum prolactin levels.4 We considered prescribing an antidepressant but decided against it because we were concerned about manic switching.
Polypharmacy is another important consideration for Ms. E. Several studies have identified polypharmacy, particularly with antipsychotics, as an independent risk factor for fracture.3 Therefore, we sought to minimize the number of medications Ms. E receives. Although lithium monotherapy is an option, we thought that her mood symptoms were severe enough that the risk of inadequately treating her bipolar symptoms outweighed the additional risk of fracture from dual therapy with lithium and aripiprazole. Untreated or inadequately treated depression is associated with a higher fracture risk. Therefore, we avoided prescribing >2 medications to mitigate any excessive risk of fracture from polypharmacy.
Bottom Line
Different classes of medications—antipsychotics, anticonvulsants, antidepressants, and lithium—used for treating bipolar disorder have been shown to increase risk of bone fracture through a variety of mechanisms. Anticonvulsants and prolactin-elevating antipsychotics are associated with increased fracture risk; evidence on lithium is mixed. Fracture risk with antidepressants is associated with duration of use, rather than dosage.
Disclosures
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Howard L, Kirkwood G, Leese M. Risk of hip fracture in patients with a history of schizophrenia. Br J Psychiatry. 2007;190:129-134.
2. Takkouche B, Montes-Martínez A, Gill SS, et al. Psychotropic medications and the risk of fracture: a meta-analysis. Drug Saf. 2007;30(2):171-184.
3. Sørensen HJ, Jensen SO, Nielsen J. Schizophrenia, antipsychotics and risk of hip fracture: a population-based analysis. Eur Neuropsychopharmacol. 2013;23(8):872-878.
4. Rahman T, Clevenger CV, Kaklamani V, et al. Antipsychotic treatment in breast cancer patients. Am J Psychiatry. 2014;171(6):616-621.
5. Bilici M, Cakirbay H, Guler M, et al. Classical and atypical neuroleptics, and bone mineral density, in patients with schizophrenia. Int J Neurosci. 2002;112(7):817-828.
6. Becker D, Liver O, Mester R, et al. Risperidone, but not olanzapine, decreases bone mineral density in female premenopausal schizophrenia patients. J Clin Psychiatry. 2003;64(7):761-766.
7. Bolton JM, Metge C, Lix L, et al. Fracture risk from psychotropic medications: a population-based analysis. J Clin Psychopharmacol. 2008;28(4):384-391.
8. Verdel BM, Souverein PC, Egberts TC, et al. Use of antidepressant drugs and risk of osteoporotic and non-osteoporotic fractures. Bone. 2010;47(3):604-609.
9. Diem SJ, Ruppert K, Cauley JA. Rates of bone loss among women initiating antidepressant medication use in midlife. J Clin Endocrinol Metab. 2013;(11):4355-4363.
10. Plenge P, Rafaelsen OJ. Lithium effects on calcium, magnesium and phosphate in man: effects on balance, bone mineral content, faecal and urinary excretion. Acta Psychiatr Scand. 1982;66(5):361-373.
11. Clément-Lacroix P, Ai M, Morvan F, et al. Lrp5-independent activation of Wnt signaling by lithium chloride increases bone formation and bone mass in mice. Proc Natl Acad Sci U S A. 2005;102(48):17406-17411.
12. Zamani A, Omrani GR, Nasab MM. Lithium’s effect on bone mineral density. Bone. 2009;44(2):331-334.
13. Swanton J, Simister R, Altmann D, et al. Bone mineral density in institutionalised patients with refractory epilepsy. Seizure. 2007;16(6):538-541.
14. Pack AM, Morrell MJ. Epilepsy and bone health in adults. Epilepsy Behav. 2004;5(suppl 2):S24-S29.
15. Pack AM. Bone disease in epilepsy. Curr Neurol Neurosci Rep. 2004;4(4):329-334.
16. Sheth RD, Hermann BP. Bone mineral density with lamotrigine monotherapy for epilepsy. Pediatr Neurol. 2007;37(4):250-254.
1. Howard L, Kirkwood G, Leese M. Risk of hip fracture in patients with a history of schizophrenia. Br J Psychiatry. 2007;190:129-134.
2. Takkouche B, Montes-Martínez A, Gill SS, et al. Psychotropic medications and the risk of fracture: a meta-analysis. Drug Saf. 2007;30(2):171-184.
3. Sørensen HJ, Jensen SO, Nielsen J. Schizophrenia, antipsychotics and risk of hip fracture: a population-based analysis. Eur Neuropsychopharmacol. 2013;23(8):872-878.
4. Rahman T, Clevenger CV, Kaklamani V, et al. Antipsychotic treatment in breast cancer patients. Am J Psychiatry. 2014;171(6):616-621.
5. Bilici M, Cakirbay H, Guler M, et al. Classical and atypical neuroleptics, and bone mineral density, in patients with schizophrenia. Int J Neurosci. 2002;112(7):817-828.
6. Becker D, Liver O, Mester R, et al. Risperidone, but not olanzapine, decreases bone mineral density in female premenopausal schizophrenia patients. J Clin Psychiatry. 2003;64(7):761-766.
7. Bolton JM, Metge C, Lix L, et al. Fracture risk from psychotropic medications: a population-based analysis. J Clin Psychopharmacol. 2008;28(4):384-391.
8. Verdel BM, Souverein PC, Egberts TC, et al. Use of antidepressant drugs and risk of osteoporotic and non-osteoporotic fractures. Bone. 2010;47(3):604-609.
9. Diem SJ, Ruppert K, Cauley JA. Rates of bone loss among women initiating antidepressant medication use in midlife. J Clin Endocrinol Metab. 2013;(11):4355-4363.
10. Plenge P, Rafaelsen OJ. Lithium effects on calcium, magnesium and phosphate in man: effects on balance, bone mineral content, faecal and urinary excretion. Acta Psychiatr Scand. 1982;66(5):361-373.
11. Clément-Lacroix P, Ai M, Morvan F, et al. Lrp5-independent activation of Wnt signaling by lithium chloride increases bone formation and bone mass in mice. Proc Natl Acad Sci U S A. 2005;102(48):17406-17411.
12. Zamani A, Omrani GR, Nasab MM. Lithium’s effect on bone mineral density. Bone. 2009;44(2):331-334.
13. Swanton J, Simister R, Altmann D, et al. Bone mineral density in institutionalised patients with refractory epilepsy. Seizure. 2007;16(6):538-541.
14. Pack AM, Morrell MJ. Epilepsy and bone health in adults. Epilepsy Behav. 2004;5(suppl 2):S24-S29.
15. Pack AM. Bone disease in epilepsy. Curr Neurol Neurosci Rep. 2004;4(4):329-334.
16. Sheth RD, Hermann BP. Bone mineral density with lamotrigine monotherapy for epilepsy. Pediatr Neurol. 2007;37(4):250-254.
Monitoring calcium with lithium treatment
I appreciate Dr. McInnis’s article and his recommendation to monitor the comprehensive metabolic profile, including the calcium level, before and during lithium treatment. There is an association among lithium treatment, hypercalcemia, and hyperparathyroidism.1,2 This can occur by lithium reducing parathyroid hormone suppression or stimulating parathyroid glands.3
Surprisingly, many guidelines do not include a recommendation to monitor the calcium level; however, the International Society for Bipolar Disorders and other experts do recommend obtaining a calcium level before initiating lithium therapy and at least annually thereafter.1,4 If hypercalcemia is present, assessing lithium and the parathyroid hormone level is recommended.3
Clinicians can continue lithium and monitor calcium if treatment is beneficial, hypercalcemia is mild, and the patient is asymptomatic.2 For a symptomatic patient or one who has significant hypercalcemia, clinicians should consider discontinuing lithium and monitoring for a normalizing calcium level.2 For patients with significant hypercalcemia who need lithium therapy, consultation with an endocrinologist is advised.3
Jonathan R. Scarff, MD
VA Outpatient Clinic
Spartanburg, South Carolina
Dr. McInnis responds
Generally, calcium is included in the comprehensive biochemistry panel (Table 1). Typically, magnesium or phosphorus is overlooked, and therefore was specifically included in the table of recommendations. There is a complex relationship between lithium and calcium; Dr. Scarff’s points highlight this. It is noteworthy that lithium normalizes the calcium amplitude during action potentials in neurons derived from induced pluripotent stem cells from persons with BD1; this suggests that there might be a direct mode of action in BD involving lithium and calcium. This finding further emphasizes the importance of monitoring calcium, and the wise clinician will verify that it is included in the comprehensive biochemistry panel.
1. McKnight RF, Adida M, Budge K, et al. Lithium toxicity profile: a systematic review and meta-analysis. Lancet. 2012;379(9817):721-728.
2. Lehmann SW, Lee J. Lithium-associated hyper-calcemia and hyperparathyroidism in the elderly: what do we know? J Affect Disord. 2013;146(2): 151-157.
3. Broome JT, Solorzano CC. Lithium use and primary hyperparathyroidism. Endocr Pract. 2011; 17(suppl 1):31-35.
4. Ng F, Mammen OK, Wilting I, et al. The International Society for Bipolar Disorders (ISBD) consensus guidelines for the safety monitoring of bipolar disorder treatments. Bipolar Disord. 2009; 11(6):559-595.
Reference
1. Chen HM, DeLong CJ, Bame M, et al. Transcripts involved in calcium signaling and telencephalic neuronal fate are altered in induced pluripotent stem cells from bipolar disorder patients. Transl Psychiatry. 2014;4:e375. doi:10.1038/tp.2014.12.
I appreciate Dr. McInnis’s article and his recommendation to monitor the comprehensive metabolic profile, including the calcium level, before and during lithium treatment. There is an association among lithium treatment, hypercalcemia, and hyperparathyroidism.1,2 This can occur by lithium reducing parathyroid hormone suppression or stimulating parathyroid glands.3
Surprisingly, many guidelines do not include a recommendation to monitor the calcium level; however, the International Society for Bipolar Disorders and other experts do recommend obtaining a calcium level before initiating lithium therapy and at least annually thereafter.1,4 If hypercalcemia is present, assessing lithium and the parathyroid hormone level is recommended.3
Clinicians can continue lithium and monitor calcium if treatment is beneficial, hypercalcemia is mild, and the patient is asymptomatic.2 For a symptomatic patient or one who has significant hypercalcemia, clinicians should consider discontinuing lithium and monitoring for a normalizing calcium level.2 For patients with significant hypercalcemia who need lithium therapy, consultation with an endocrinologist is advised.3
Jonathan R. Scarff, MD
VA Outpatient Clinic
Spartanburg, South Carolina
Dr. McInnis responds
Generally, calcium is included in the comprehensive biochemistry panel (Table 1). Typically, magnesium or phosphorus is overlooked, and therefore was specifically included in the table of recommendations. There is a complex relationship between lithium and calcium; Dr. Scarff’s points highlight this. It is noteworthy that lithium normalizes the calcium amplitude during action potentials in neurons derived from induced pluripotent stem cells from persons with BD1; this suggests that there might be a direct mode of action in BD involving lithium and calcium. This finding further emphasizes the importance of monitoring calcium, and the wise clinician will verify that it is included in the comprehensive biochemistry panel.
I appreciate Dr. McInnis’s article and his recommendation to monitor the comprehensive metabolic profile, including the calcium level, before and during lithium treatment. There is an association among lithium treatment, hypercalcemia, and hyperparathyroidism.1,2 This can occur by lithium reducing parathyroid hormone suppression or stimulating parathyroid glands.3
Surprisingly, many guidelines do not include a recommendation to monitor the calcium level; however, the International Society for Bipolar Disorders and other experts do recommend obtaining a calcium level before initiating lithium therapy and at least annually thereafter.1,4 If hypercalcemia is present, assessing lithium and the parathyroid hormone level is recommended.3
Clinicians can continue lithium and monitor calcium if treatment is beneficial, hypercalcemia is mild, and the patient is asymptomatic.2 For a symptomatic patient or one who has significant hypercalcemia, clinicians should consider discontinuing lithium and monitoring for a normalizing calcium level.2 For patients with significant hypercalcemia who need lithium therapy, consultation with an endocrinologist is advised.3
Jonathan R. Scarff, MD
VA Outpatient Clinic
Spartanburg, South Carolina
Dr. McInnis responds
Generally, calcium is included in the comprehensive biochemistry panel (Table 1). Typically, magnesium or phosphorus is overlooked, and therefore was specifically included in the table of recommendations. There is a complex relationship between lithium and calcium; Dr. Scarff’s points highlight this. It is noteworthy that lithium normalizes the calcium amplitude during action potentials in neurons derived from induced pluripotent stem cells from persons with BD1; this suggests that there might be a direct mode of action in BD involving lithium and calcium. This finding further emphasizes the importance of monitoring calcium, and the wise clinician will verify that it is included in the comprehensive biochemistry panel.
1. McKnight RF, Adida M, Budge K, et al. Lithium toxicity profile: a systematic review and meta-analysis. Lancet. 2012;379(9817):721-728.
2. Lehmann SW, Lee J. Lithium-associated hyper-calcemia and hyperparathyroidism in the elderly: what do we know? J Affect Disord. 2013;146(2): 151-157.
3. Broome JT, Solorzano CC. Lithium use and primary hyperparathyroidism. Endocr Pract. 2011; 17(suppl 1):31-35.
4. Ng F, Mammen OK, Wilting I, et al. The International Society for Bipolar Disorders (ISBD) consensus guidelines for the safety monitoring of bipolar disorder treatments. Bipolar Disord. 2009; 11(6):559-595.
Reference
1. Chen HM, DeLong CJ, Bame M, et al. Transcripts involved in calcium signaling and telencephalic neuronal fate are altered in induced pluripotent stem cells from bipolar disorder patients. Transl Psychiatry. 2014;4:e375. doi:10.1038/tp.2014.12.
1. McKnight RF, Adida M, Budge K, et al. Lithium toxicity profile: a systematic review and meta-analysis. Lancet. 2012;379(9817):721-728.
2. Lehmann SW, Lee J. Lithium-associated hyper-calcemia and hyperparathyroidism in the elderly: what do we know? J Affect Disord. 2013;146(2): 151-157.
3. Broome JT, Solorzano CC. Lithium use and primary hyperparathyroidism. Endocr Pract. 2011; 17(suppl 1):31-35.
4. Ng F, Mammen OK, Wilting I, et al. The International Society for Bipolar Disorders (ISBD) consensus guidelines for the safety monitoring of bipolar disorder treatments. Bipolar Disord. 2009; 11(6):559-595.
Reference
1. Chen HM, DeLong CJ, Bame M, et al. Transcripts involved in calcium signaling and telencephalic neuronal fate are altered in induced pluripotent stem cells from bipolar disorder patients. Transl Psychiatry. 2014;4:e375. doi:10.1038/tp.2014.12.
Cautions when prescribing lithium
I was astonished that Dr. Melvin G. McInnis’ article on using lithium to treat bipolar disorder (BD) (Current Psychiatry, June 2014, p. 38-44; [http://bit.ly/1sszAUr]) did not address all the potential hazards of the medication. He discussed side effects, but only how to manage them so that patients will adhere to treatment.
I have used lithium for patients with BD, and often it is efficacious, although hazardous in overdose. Lithium toxicity can cause cardiac arrhythmias, and must be monitored closely. In addition, the effects of hydration and exercise on the lithium level, especially during summer, often are ignored.
Two of my patients, an adolescent and an adult, were well-maintained on lithium, adhered to treatment, and had no concurrent medical problems, but developed significant toxicity for no reason that I could determine. The adult had a lithium level of 2.0 mEq/L in the emergency room; the adolescent had a lithium level of 1.8 mEq/L. Levels this high are considered potentially lethal, and because it happened without warning and without a cause that I could determine, I consider lithium to be one of the riskier mood stabilizers. I still prescribe it, but with great caution.
Dr. McInnis also did not mention the possibility of lithium-induced diabetes insipidus, a condition in which the kidneys are no longer able to concentrate urine and that is marked by excessive urination, concomitant water intake, and low urine specific gravity. It is uncommon, but I have seen it 3 times in 30 years, in a practice that specializes in psychotherapy and does not see a high percentage of patients with BD. I consider it a condition that must be kept in mind as we follow our patients in long-term treatment.
Mary Davis, MD
Lancaster, Pennsylvania
Dr. McInnis responds
Dr. Davis raises the issue of lithium toxicity and provides examples of 2 patients who developed levels of 2.0 mEq/L and 1.8 mEq/L. These levels clearly are well beyond the toxicity threshold of 1.3 mEq/L, and the patients wisely sought urgent care. These scenarios exemplify the need for regular monitoring of the lithium level—in particular, when there is any change in physical or mental health status. Development of significant toxicity generally has some lead-time with emerging short-term side effects (outlined in Table 2 of my article), which underscores the importance of discussing the nature of emerging side effects with your patient.
Dr. Davis is correct in noting that the practitioner must be aware of long-term side effects of lithium. I find it helpful to discuss these effects with the patient in the context of short-term (days or weeks), intermediate (weeks or months), and long-term (months or years) time frames (Table 2). Diabetes insipidus is listed as an intermediate side effect.
I am grateful to Dr. Davis for raising the issue of hydration and summer heat, a concern among parents and coaches when student athletes practice strenuously for extended hours.1 Miller et al2 found that the concentration of lithium was between 1.2- and 4.6-fold in forearm sweat compared with serum levels, with the implication that heat-induced sweating may lower lithium levels. Jefferson et al3 studied 4 athletes after a 20-km race and found that all had become dehydrated but had a decrease in the serum lithium level. This is contrary to the widely held belief that excessive sweating predisposes to lithium toxicity.
BD is among the more lethal psychiatric disorders, and lithium is among the few medications shown to mitigate suicidal behavior.4 As with any medication, lithium is not without risk, and there is a clear need for informed medical management. Any notable change in health status or physical activity in a patient taking lithium is worthy of review, with recommendations based on knowledge of the patient and medical science.
1. Reardon CL, Factor RM. Sport psychiatry: a systematic review of diagnosis and medical treatment of mental illness in athletes. Sports Med. 2010;40:961-980.
2. Miller EB, Pain RW, Skripal PJ. Sweat lithium in manic-depression. Br J Psychiatry. 1978;133:477-478.
3. Jefferson JW, Greist JH, Clagnaz PJ, et al. Effect of strenuous exercise on serum lithium level in man. Am J Psychiatry. 1982;139(12):1593-1595.
4. Goodwin FK, Fireman B, Simon GE, et al. Suicide risk in bipolar disorder during treatment with lithium and divalproex. JAMA. 2003;290(11):1467-1473.
I was astonished that Dr. Melvin G. McInnis’ article on using lithium to treat bipolar disorder (BD) (Current Psychiatry, June 2014, p. 38-44; [http://bit.ly/1sszAUr]) did not address all the potential hazards of the medication. He discussed side effects, but only how to manage them so that patients will adhere to treatment.
I have used lithium for patients with BD, and often it is efficacious, although hazardous in overdose. Lithium toxicity can cause cardiac arrhythmias, and must be monitored closely. In addition, the effects of hydration and exercise on the lithium level, especially during summer, often are ignored.
Two of my patients, an adolescent and an adult, were well-maintained on lithium, adhered to treatment, and had no concurrent medical problems, but developed significant toxicity for no reason that I could determine. The adult had a lithium level of 2.0 mEq/L in the emergency room; the adolescent had a lithium level of 1.8 mEq/L. Levels this high are considered potentially lethal, and because it happened without warning and without a cause that I could determine, I consider lithium to be one of the riskier mood stabilizers. I still prescribe it, but with great caution.
Dr. McInnis also did not mention the possibility of lithium-induced diabetes insipidus, a condition in which the kidneys are no longer able to concentrate urine and that is marked by excessive urination, concomitant water intake, and low urine specific gravity. It is uncommon, but I have seen it 3 times in 30 years, in a practice that specializes in psychotherapy and does not see a high percentage of patients with BD. I consider it a condition that must be kept in mind as we follow our patients in long-term treatment.
Mary Davis, MD
Lancaster, Pennsylvania
Dr. McInnis responds
Dr. Davis raises the issue of lithium toxicity and provides examples of 2 patients who developed levels of 2.0 mEq/L and 1.8 mEq/L. These levels clearly are well beyond the toxicity threshold of 1.3 mEq/L, and the patients wisely sought urgent care. These scenarios exemplify the need for regular monitoring of the lithium level—in particular, when there is any change in physical or mental health status. Development of significant toxicity generally has some lead-time with emerging short-term side effects (outlined in Table 2 of my article), which underscores the importance of discussing the nature of emerging side effects with your patient.
Dr. Davis is correct in noting that the practitioner must be aware of long-term side effects of lithium. I find it helpful to discuss these effects with the patient in the context of short-term (days or weeks), intermediate (weeks or months), and long-term (months or years) time frames (Table 2). Diabetes insipidus is listed as an intermediate side effect.
I am grateful to Dr. Davis for raising the issue of hydration and summer heat, a concern among parents and coaches when student athletes practice strenuously for extended hours.1 Miller et al2 found that the concentration of lithium was between 1.2- and 4.6-fold in forearm sweat compared with serum levels, with the implication that heat-induced sweating may lower lithium levels. Jefferson et al3 studied 4 athletes after a 20-km race and found that all had become dehydrated but had a decrease in the serum lithium level. This is contrary to the widely held belief that excessive sweating predisposes to lithium toxicity.
BD is among the more lethal psychiatric disorders, and lithium is among the few medications shown to mitigate suicidal behavior.4 As with any medication, lithium is not without risk, and there is a clear need for informed medical management. Any notable change in health status or physical activity in a patient taking lithium is worthy of review, with recommendations based on knowledge of the patient and medical science.
I was astonished that Dr. Melvin G. McInnis’ article on using lithium to treat bipolar disorder (BD) (Current Psychiatry, June 2014, p. 38-44; [http://bit.ly/1sszAUr]) did not address all the potential hazards of the medication. He discussed side effects, but only how to manage them so that patients will adhere to treatment.
I have used lithium for patients with BD, and often it is efficacious, although hazardous in overdose. Lithium toxicity can cause cardiac arrhythmias, and must be monitored closely. In addition, the effects of hydration and exercise on the lithium level, especially during summer, often are ignored.
Two of my patients, an adolescent and an adult, were well-maintained on lithium, adhered to treatment, and had no concurrent medical problems, but developed significant toxicity for no reason that I could determine. The adult had a lithium level of 2.0 mEq/L in the emergency room; the adolescent had a lithium level of 1.8 mEq/L. Levels this high are considered potentially lethal, and because it happened without warning and without a cause that I could determine, I consider lithium to be one of the riskier mood stabilizers. I still prescribe it, but with great caution.
Dr. McInnis also did not mention the possibility of lithium-induced diabetes insipidus, a condition in which the kidneys are no longer able to concentrate urine and that is marked by excessive urination, concomitant water intake, and low urine specific gravity. It is uncommon, but I have seen it 3 times in 30 years, in a practice that specializes in psychotherapy and does not see a high percentage of patients with BD. I consider it a condition that must be kept in mind as we follow our patients in long-term treatment.
Mary Davis, MD
Lancaster, Pennsylvania
Dr. McInnis responds
Dr. Davis raises the issue of lithium toxicity and provides examples of 2 patients who developed levels of 2.0 mEq/L and 1.8 mEq/L. These levels clearly are well beyond the toxicity threshold of 1.3 mEq/L, and the patients wisely sought urgent care. These scenarios exemplify the need for regular monitoring of the lithium level—in particular, when there is any change in physical or mental health status. Development of significant toxicity generally has some lead-time with emerging short-term side effects (outlined in Table 2 of my article), which underscores the importance of discussing the nature of emerging side effects with your patient.
Dr. Davis is correct in noting that the practitioner must be aware of long-term side effects of lithium. I find it helpful to discuss these effects with the patient in the context of short-term (days or weeks), intermediate (weeks or months), and long-term (months or years) time frames (Table 2). Diabetes insipidus is listed as an intermediate side effect.
I am grateful to Dr. Davis for raising the issue of hydration and summer heat, a concern among parents and coaches when student athletes practice strenuously for extended hours.1 Miller et al2 found that the concentration of lithium was between 1.2- and 4.6-fold in forearm sweat compared with serum levels, with the implication that heat-induced sweating may lower lithium levels. Jefferson et al3 studied 4 athletes after a 20-km race and found that all had become dehydrated but had a decrease in the serum lithium level. This is contrary to the widely held belief that excessive sweating predisposes to lithium toxicity.
BD is among the more lethal psychiatric disorders, and lithium is among the few medications shown to mitigate suicidal behavior.4 As with any medication, lithium is not without risk, and there is a clear need for informed medical management. Any notable change in health status or physical activity in a patient taking lithium is worthy of review, with recommendations based on knowledge of the patient and medical science.
1. Reardon CL, Factor RM. Sport psychiatry: a systematic review of diagnosis and medical treatment of mental illness in athletes. Sports Med. 2010;40:961-980.
2. Miller EB, Pain RW, Skripal PJ. Sweat lithium in manic-depression. Br J Psychiatry. 1978;133:477-478.
3. Jefferson JW, Greist JH, Clagnaz PJ, et al. Effect of strenuous exercise on serum lithium level in man. Am J Psychiatry. 1982;139(12):1593-1595.
4. Goodwin FK, Fireman B, Simon GE, et al. Suicide risk in bipolar disorder during treatment with lithium and divalproex. JAMA. 2003;290(11):1467-1473.
1. Reardon CL, Factor RM. Sport psychiatry: a systematic review of diagnosis and medical treatment of mental illness in athletes. Sports Med. 2010;40:961-980.
2. Miller EB, Pain RW, Skripal PJ. Sweat lithium in manic-depression. Br J Psychiatry. 1978;133:477-478.
3. Jefferson JW, Greist JH, Clagnaz PJ, et al. Effect of strenuous exercise on serum lithium level in man. Am J Psychiatry. 1982;139(12):1593-1595.
4. Goodwin FK, Fireman B, Simon GE, et al. Suicide risk in bipolar disorder during treatment with lithium and divalproex. JAMA. 2003;290(11):1467-1473.
Severity of side effects key to choosing between lithium and quetiapine in bipolar I and II
HOLLYWOOD, FLA. – Because there is no clinically significant difference in bipolar spectrum symptom relief offered by either lithium or quetiapine, which mood stabilizer clinicians choose to prescribe should largely depend on what side effects patients can tolerate.
That’s the conclusion of a panel of experts who presented data from the Bipolar CHOICE (Clinical Health Outcomes Initiative in Comparative Effectiveness for Bipolar Disorder) clinical trial at a meeting of the American Society of Clinical Psychopharmacology, formerly known as the New Clinical Drug Evaluation Unit meeting.
By randomly assigning the study’s 482 participating outpatient adults to either lithium or quetiapine, while allowing clinicians to prescribe adjunctive treatment as necessary; and by using a broader-than-usual set of inclusion criteria, the investigators hoped to provide clinicians with "real world" data on how the respective mood stabilizers perform.
"To my knowledge, this is the first randomized comparative effectiveness study of regular, expert care that looks at lithium versus quetiapine strategies of treatment, both with adjunctive personalized treatment when necessary," study coauthor Dr. Andrew A. Nierenberg told the audience in a well-attended session.
"We tortured the data to get a P value to pop out," said Dr. Nierenberg, who is the director of the Bipolar Clinic and Research Program at Massachusetts General Hospital in Boston. "The bottom line was that the treatment strategies, based on either lithium or quetiapine, were very similar. I think the clinical implication is that the choice of treatment really depends on someone’s tolerability. This may resurrect lithium as something [clinicians] should at least consider, because its use has gone down dramatically in the last 20 years or so."
Side effects such as metabolic or sleep disturbances were slightly less frequent in the lithium group, although the difference was not statistically significant; rates of suicidality and discontinuation of medication due to side effects were also similar across the two groups, according to Dr. David E. Kemp of the psychiatry department at Case Western Reserve University, Cleveland, and a study coauthor who also presented data during the session. Changes from baseline in mood stabilization were also virtually the same across the groups.
‘Rules of engagement’
The study enrolled adults aged 18-68 years who were at least mildly ill with either bipolar disorder I (68%) or II, with a Clinical Global Impression (CGI) score of at least 3, although the average CGI score was 4.5 (standard deviation of plus or minus 0.9). Of the 482 enrolled, 364 completed the study.
The so-called rules of engagement for clinicians from the 11 participating centers were that "they do everything they could to get their patients well," with the caveat that the lithium patient group not receive any antipsychotic, and the quetiapine patient group not receive lithium or any other antipsychotic.
"But they could get anything else that they needed," Dr. Nierenberg said.
At the time of enrollment, subjects were, in the estimation of their treating physician, experiencing symptoms that warranted a change in treatment, and that either lithium or quetiapine would be viable therapeutic options. Patients did not need to be lithium- or quetiapine-naive, but they could not have been treated with either drug in the previous 30 days. If they were already taking a second-generation antipsychotic, participants had to be willing to discontinue that medication and to be randomly assigned to either of the study drugs.
This strict adherence to one mood stabilizer, combined with adjunctive personalized treatments such as antidepressants or benzodiazepines, prescribed at the discretion of the treating clinician, ensured that the study tested the strategy of using either lithium or quetiapine as the base of treatment, Dr. Nierenberg said.
‘Similar’ results
Clinicians also were instructed to use the maximum tolerated dose of the respective mood stabilizer. For the 240 patients in the lithium group, the mean maximum tolerated dose was 1,007.5 mg, with the median dose being a "perfectly reasonable 900 mg," Dr. Nierenberg said. Blood lithium levels were taken at weeks 2, 16, and 24 and also were all found to be "reasonable," with most patients getting to at least 0.6 mEq/L by week 24.
For the 242 members of the quetiapine group, the maximum tolerated dose was 344.9 mg, with a median tolerated dose of 300 mg, which Dr. Nierenberg said was "what you would expect."
The overall result was that regardless of which mood stabilizer was prescribed along with adjunctive personalized treatment, the coprimary outcomes of the benefits and harms ratio from treatment effects over time (the CGI efficacy index), and the number of necessary clinical adjustments (changes in medication excluding titrations according to scale), were virtually parallel: For each primary outcome, the P value for the estimated change in baseline was less than .0001 for both lithium plus adjunctive treatment, and quetiapine plus adjunctive treatment.
Further, Dr. Nierenberg reported the "unexpected" result that at 6 months, about a quarter of each group was observed to be doing well without any adjunctive personalized treatments (23.8% of the lithium group; 27.3% of the quetiapine group; P = .14). "We thought that everybody would be on multiple things," Dr. Nierenberg said.
There was also no real difference in the time to discontinuation. "We were actually surprised that at 6 months, about three-quarters of the patients were still on the study drug, which was pretty good considering that this was a very ill population," Dr. Nierenberg said.
Using a CGI bipolar severity scale rating of up to 2, maintained for at least 8 weeks, the investigators found that overall, 20% were considered doing "really well"; it was also about 20% for the lithium plus adjunctive therapy group and the quetiapine and adjunctive therapy group (P = .14 for all three).
Because the Agency for Healthcare Research and Quality–funded study mandated that researchers end their investigation at precisely 36 months, Dr. Nierenberg said the study sites "randomized the 482 patients to 6 months of treatment within 36 months from start to finish," but he added that the investigators would have preferred to conduct a full 2-year study of participants.
"The good news was that most of the patients actually did improve substantially; the bad news is that while maybe a quarter did really well by the end of 6 months, we don’t know whether if we were to extend the study out further, we would see a difference between the two groups, whether they would continue to improve, or whether they would relapse," he said.
Adverse effects, predictors of response
Because study participants were followed for only 6 months, the long-term adverse effects of many years of exposure to either mood stabilizer, such as renal impairment with lithium or type II diabetes with quetiapine, were not considered, Dr. Kemp said. The adverse-effect profile for lithium generally includes a narrow therapeutic index; nausea, vomiting, and diarrhea; renal impairment; and hypothyroidism. Although Dr. Kemp said that the long-term adverse effects of quetiapine are understudied, known short-term side effects include the potential for sedation or somnolence, and weight gain.
Changes in baseline on the Frequency and Intensity of Side Effects Ratings for the quetiapine plus adjunctive personalized treatment arm were slightly more adverse than for the lithium and additional treatment arm. For the lithium plus adjunctive treatment group, the mean frequency of side effects was –1.41 (–1.78, –1.04 SD); the mean intensity of side effects was –1.48 (–1.80, –1.15 SD); and the mean level of impairment was –1.13 (–1.43, –0.82 SD). For the quetiapine and adjunctive treatment group, the mean frequency measure was –1.08 (–1.45, –0.72 SD); the intensity was –1.12 (–1.44, –0.79 SD); and the impairment was –0.77 (–1.07, –0.47 SD). For all scores in both groups, P was less than .001.
The changes from baseline on the Bipolar Index Severity Scale (BISS) overall for the quetiapine plus adjunctive treatment group was –28.56 (–10.91, –26.21 SD; P less than .0001). For the lithium plus adjunctive treatment group, it was –27.61 (–29.99, –25.24 SD; P less than .0001). The overall difference between the two groups was 0.94 (–2.10, 3.99 SD; P = .54).
Scores on the Longitudinal Interval Follow-up Evaluation Range of Impaired Functioning Tool also slightly favored lithium: –3.74 (–4.29, –3.19 SD) vs. –3.6 (–4.15, –3.07 SD) for quetiapine (P less than .0001 for all).
To the investigators’ surprise, among those with comorbid anxiety, the lithium plus adjunctive treatment group had fewer necessary clinical adjustments per month than did the quetiapine plus adjunctive treatment group, according to Dr. Nierenberg: –0.83 vs. 1.11 (P = .02).
"That seemed counterintuitive, and this difference was only with anxiety, not with any other comorbid psychiatric conditions," Dr. Nierenberg said. He hypothesized that it was possible benzodiazepines were used more frequently and easily with lithium than with quetiapine, but said future analyses would give a clearer answer. "We have a detailed database of every other medication used: when it was started, when it was stopped, and the reason why everything was done."
Not having current anxiety disorder was predictive of a better outcome (odds ratio, 1.81; P = .02), as was employment (OR, 1.67; P = .04). Those with bipolar II disorder responded better to treatment than those with bipolar disorder I, having an OR of response to treatment of nearly 1.8 P = .03). "That was a surprise to us, too," Dr. Nierenberg said.
Just over a quarter (27%) of the study population had metabolic syndrome, according to Dr. Kemp, who said this was not found to influence treatment outcomes.
Nearly half of the overall population was obese (48%), and slightly less than half (44%) had adipose. Prospective analysis indicated that patients with either obesity or adiposity tended to show less improvement in their CGI scores in a statistically significant way, regardless of which mood stabilizer they were on.
"Part of that might be due to overlapping pathophysiology between bipolar disorder and obesity," Dr. Kemp said. "Much of that is centered around inflammation in the central nervous system, as well as alterations in adipokines receptor levels and mitochondrial dysfunction."
"A key issue is to take care of obesity," said panel discussant Dr. Mauricio Tohen, chairman of psychiatry at the University of New Mexico in Albuquerque. "Regardless of the treatment, when there is obesity, the outcome will not be as good."
Efficacy vs. ‘generalizability’
Adjunctive personalized treatment was measured to help minimize the lack of assay sensitivity and internal validity typically inherent in comparative effectiveness trials, while also achieving more broadly applicable results.
"The right study depends on the right question," Dr. Tohen said. "If the question is whether a particular treatment has efficacy and is safe, then of course, we need an efficacy study. The problem with efficacy studies is that they limit the generalizability."
Instead, Dr. Tohen, who disclosed he spent more than a decade evaluating efficacy studies for both Eli Lily and AstraZeneca (makers of quetiapine), praised the metric of the adjunctive personalized treatments, and said a study should answer the question, "Of all the patients in my clinic, who will do better on which treatment? With efficacy studies you cannot answer that question because of the exclusion criteria that need to be taken into account."
In addition, Dr. Tohen said the metric likely would increase in importance under the Affordable Care Act, where efficacy is not the only consideration. "For example, if a patient relapses earlier, it might not be reimbursed, so asking what other outcomes we need to measure in [these kinds of] comparative effectiveness studies is very important."
In the CHOICE study, which had no placebo group, participating physicians were asked to track each patient’s dosage changes, missed doses, new medications added, discontinued medications, and the specific reasons for any of these changes. Changes that were made because of lack of effectiveness or intolerance were measured; however, planned dosage titrations according to normal scales were not considered necessary clinical adjustments but simply the regular course of treatment.
This heterogeneity of adjunctive treatment was seen by the investigators as a way to reflect "real world" practice, although they also noted it could be seen as a limitation to the study.
Another way the study was designed to reflect real-world practice was the inclusion of a broader-than-usual group of participants, and asking them for their feedback at the end of the study period.
"Before we put together these data, we held a stakeholder summit," Dr. Nierenberg said. "We invited patients and their advocates to help us interpret the study. They felt strongly that this was a reasonably positive study, because they thought that people really did get better. You could report these data toward the negative or the positive, but we reported toward the positive because that is what the stakeholders asked us to do." Dr. Tohen said that this kind of feedback in a comparative effectiveness study was another way these kinds of studies could help guide practice in the era of the Affordable Care Act.
As for who was actually admitted to the study, "exclusions were minimal," said Dr. Edward S. Friedman of the psychiatry department at the University of Pittsburgh. Those with a history of drug or alcohol dependence in the previous 30 days, a demonstrated intolerance to either study drug, or severe cardiovascular or renal disease were excluded, as were those with unstable thyroid disease, and pregnant or breastfeeding women.
Overall, the group was multiethnic, although nearly three-quarters were white and 20% were black. Roughly a third of the entire study population was employed, a third unemployed, and the rest were students, retirees, or those on disability.
The overall BISS score at enrollment averaged 56.1 (SD plus or minus 18.8). The average BISS depression score was 37.6 (SD plus or minus 14.0), and the average BISS mania score was 18.5 (SD plus or minus 12.1).
There were many comorbid psychiatric conditions, ranging from panic disorder to agoraphobia, although substance use was highest (61.4%). Current anxiety disorder also was prevalent at 58%.
"The CHOICE sample was similar in age and gender to previous efficacy studies," Dr. Friedman said. "It was more representative of the U.S. population than efficacy studies such as STEP-BD or LITMUS."
The 283 women (59% of the study) were more likely to report having spent a greater percentage of time depressed in the previous year, although the percentage of time spent in manic or hypomanic states was equal across both genders.
"This study was composed of sick individuals who had been sick for a considerable period of time," Dr. Friedman said.
For example, in the CHOICE study, 47% had been hospitalized for their bipolar disorder previously, vs. 43% in the LITMUS study. CHOICE participants had an attempted suicide rate of 36%, which fell between the 41% in the LITMUS trial and the 33.3% in an efficacy study conducted by Dr. Joseph R. Calabrese and his colleagues (J. Clin. Psychiatry 1999;60:79-88).
The average age for the first depressive episode for CHOICE participants was 16.4 years; the first manic episode tended to be at 20 years; and the first mood episode tended to be around 15.5 years. These data were similar to those in the LITMUS study, but differed from data in several efficacy studies such as the one by Dr. Calabrese, where the average age was 31.3 years.
"You might anticipate that in these specialty clinics we would have very few medication-naive patients, and yet we did," Dr. Friedman said during a postpanel audience participation session. "Looking for patients with a low threshold, we brought in patients who were very sick. They were also older, with lots of previous episodes. Maybe we missed younger patients who didn’t have as many previous episodes."
"These are the patients you would see in your practice," Dr. Nierenberg said before asking the audience what they think should be studied next in a comparative effectiveness trial. "We could look at the use of different antipsychotics, the combinations we use, with or without lamotrigine," he said.
"Most of the decisions that are made in medicine are made without evidence. Most of the things that are done are done without evidence. You have combinations that were never studied before or never even used before, and it’s across all medicine." Because medicine needs to be learning more, he said the audience needed to participate. "What are the questions that need to be asked?"
On Twitter @whitneymcknight
HOLLYWOOD, FLA. – Because there is no clinically significant difference in bipolar spectrum symptom relief offered by either lithium or quetiapine, which mood stabilizer clinicians choose to prescribe should largely depend on what side effects patients can tolerate.
That’s the conclusion of a panel of experts who presented data from the Bipolar CHOICE (Clinical Health Outcomes Initiative in Comparative Effectiveness for Bipolar Disorder) clinical trial at a meeting of the American Society of Clinical Psychopharmacology, formerly known as the New Clinical Drug Evaluation Unit meeting.
By randomly assigning the study’s 482 participating outpatient adults to either lithium or quetiapine, while allowing clinicians to prescribe adjunctive treatment as necessary; and by using a broader-than-usual set of inclusion criteria, the investigators hoped to provide clinicians with "real world" data on how the respective mood stabilizers perform.
"To my knowledge, this is the first randomized comparative effectiveness study of regular, expert care that looks at lithium versus quetiapine strategies of treatment, both with adjunctive personalized treatment when necessary," study coauthor Dr. Andrew A. Nierenberg told the audience in a well-attended session.
"We tortured the data to get a P value to pop out," said Dr. Nierenberg, who is the director of the Bipolar Clinic and Research Program at Massachusetts General Hospital in Boston. "The bottom line was that the treatment strategies, based on either lithium or quetiapine, were very similar. I think the clinical implication is that the choice of treatment really depends on someone’s tolerability. This may resurrect lithium as something [clinicians] should at least consider, because its use has gone down dramatically in the last 20 years or so."
Side effects such as metabolic or sleep disturbances were slightly less frequent in the lithium group, although the difference was not statistically significant; rates of suicidality and discontinuation of medication due to side effects were also similar across the two groups, according to Dr. David E. Kemp of the psychiatry department at Case Western Reserve University, Cleveland, and a study coauthor who also presented data during the session. Changes from baseline in mood stabilization were also virtually the same across the groups.
‘Rules of engagement’
The study enrolled adults aged 18-68 years who were at least mildly ill with either bipolar disorder I (68%) or II, with a Clinical Global Impression (CGI) score of at least 3, although the average CGI score was 4.5 (standard deviation of plus or minus 0.9). Of the 482 enrolled, 364 completed the study.
The so-called rules of engagement for clinicians from the 11 participating centers were that "they do everything they could to get their patients well," with the caveat that the lithium patient group not receive any antipsychotic, and the quetiapine patient group not receive lithium or any other antipsychotic.
"But they could get anything else that they needed," Dr. Nierenberg said.
At the time of enrollment, subjects were, in the estimation of their treating physician, experiencing symptoms that warranted a change in treatment, and that either lithium or quetiapine would be viable therapeutic options. Patients did not need to be lithium- or quetiapine-naive, but they could not have been treated with either drug in the previous 30 days. If they were already taking a second-generation antipsychotic, participants had to be willing to discontinue that medication and to be randomly assigned to either of the study drugs.
This strict adherence to one mood stabilizer, combined with adjunctive personalized treatments such as antidepressants or benzodiazepines, prescribed at the discretion of the treating clinician, ensured that the study tested the strategy of using either lithium or quetiapine as the base of treatment, Dr. Nierenberg said.
‘Similar’ results
Clinicians also were instructed to use the maximum tolerated dose of the respective mood stabilizer. For the 240 patients in the lithium group, the mean maximum tolerated dose was 1,007.5 mg, with the median dose being a "perfectly reasonable 900 mg," Dr. Nierenberg said. Blood lithium levels were taken at weeks 2, 16, and 24 and also were all found to be "reasonable," with most patients getting to at least 0.6 mEq/L by week 24.
For the 242 members of the quetiapine group, the maximum tolerated dose was 344.9 mg, with a median tolerated dose of 300 mg, which Dr. Nierenberg said was "what you would expect."
The overall result was that regardless of which mood stabilizer was prescribed along with adjunctive personalized treatment, the coprimary outcomes of the benefits and harms ratio from treatment effects over time (the CGI efficacy index), and the number of necessary clinical adjustments (changes in medication excluding titrations according to scale), were virtually parallel: For each primary outcome, the P value for the estimated change in baseline was less than .0001 for both lithium plus adjunctive treatment, and quetiapine plus adjunctive treatment.
Further, Dr. Nierenberg reported the "unexpected" result that at 6 months, about a quarter of each group was observed to be doing well without any adjunctive personalized treatments (23.8% of the lithium group; 27.3% of the quetiapine group; P = .14). "We thought that everybody would be on multiple things," Dr. Nierenberg said.
There was also no real difference in the time to discontinuation. "We were actually surprised that at 6 months, about three-quarters of the patients were still on the study drug, which was pretty good considering that this was a very ill population," Dr. Nierenberg said.
Using a CGI bipolar severity scale rating of up to 2, maintained for at least 8 weeks, the investigators found that overall, 20% were considered doing "really well"; it was also about 20% for the lithium plus adjunctive therapy group and the quetiapine and adjunctive therapy group (P = .14 for all three).
Because the Agency for Healthcare Research and Quality–funded study mandated that researchers end their investigation at precisely 36 months, Dr. Nierenberg said the study sites "randomized the 482 patients to 6 months of treatment within 36 months from start to finish," but he added that the investigators would have preferred to conduct a full 2-year study of participants.
"The good news was that most of the patients actually did improve substantially; the bad news is that while maybe a quarter did really well by the end of 6 months, we don’t know whether if we were to extend the study out further, we would see a difference between the two groups, whether they would continue to improve, or whether they would relapse," he said.
Adverse effects, predictors of response
Because study participants were followed for only 6 months, the long-term adverse effects of many years of exposure to either mood stabilizer, such as renal impairment with lithium or type II diabetes with quetiapine, were not considered, Dr. Kemp said. The adverse-effect profile for lithium generally includes a narrow therapeutic index; nausea, vomiting, and diarrhea; renal impairment; and hypothyroidism. Although Dr. Kemp said that the long-term adverse effects of quetiapine are understudied, known short-term side effects include the potential for sedation or somnolence, and weight gain.
Changes in baseline on the Frequency and Intensity of Side Effects Ratings for the quetiapine plus adjunctive personalized treatment arm were slightly more adverse than for the lithium and additional treatment arm. For the lithium plus adjunctive treatment group, the mean frequency of side effects was –1.41 (–1.78, –1.04 SD); the mean intensity of side effects was –1.48 (–1.80, –1.15 SD); and the mean level of impairment was –1.13 (–1.43, –0.82 SD). For the quetiapine and adjunctive treatment group, the mean frequency measure was –1.08 (–1.45, –0.72 SD); the intensity was –1.12 (–1.44, –0.79 SD); and the impairment was –0.77 (–1.07, –0.47 SD). For all scores in both groups, P was less than .001.
The changes from baseline on the Bipolar Index Severity Scale (BISS) overall for the quetiapine plus adjunctive treatment group was –28.56 (–10.91, –26.21 SD; P less than .0001). For the lithium plus adjunctive treatment group, it was –27.61 (–29.99, –25.24 SD; P less than .0001). The overall difference between the two groups was 0.94 (–2.10, 3.99 SD; P = .54).
Scores on the Longitudinal Interval Follow-up Evaluation Range of Impaired Functioning Tool also slightly favored lithium: –3.74 (–4.29, –3.19 SD) vs. –3.6 (–4.15, –3.07 SD) for quetiapine (P less than .0001 for all).
To the investigators’ surprise, among those with comorbid anxiety, the lithium plus adjunctive treatment group had fewer necessary clinical adjustments per month than did the quetiapine plus adjunctive treatment group, according to Dr. Nierenberg: –0.83 vs. 1.11 (P = .02).
"That seemed counterintuitive, and this difference was only with anxiety, not with any other comorbid psychiatric conditions," Dr. Nierenberg said. He hypothesized that it was possible benzodiazepines were used more frequently and easily with lithium than with quetiapine, but said future analyses would give a clearer answer. "We have a detailed database of every other medication used: when it was started, when it was stopped, and the reason why everything was done."
Not having current anxiety disorder was predictive of a better outcome (odds ratio, 1.81; P = .02), as was employment (OR, 1.67; P = .04). Those with bipolar II disorder responded better to treatment than those with bipolar disorder I, having an OR of response to treatment of nearly 1.8 P = .03). "That was a surprise to us, too," Dr. Nierenberg said.
Just over a quarter (27%) of the study population had metabolic syndrome, according to Dr. Kemp, who said this was not found to influence treatment outcomes.
Nearly half of the overall population was obese (48%), and slightly less than half (44%) had adipose. Prospective analysis indicated that patients with either obesity or adiposity tended to show less improvement in their CGI scores in a statistically significant way, regardless of which mood stabilizer they were on.
"Part of that might be due to overlapping pathophysiology between bipolar disorder and obesity," Dr. Kemp said. "Much of that is centered around inflammation in the central nervous system, as well as alterations in adipokines receptor levels and mitochondrial dysfunction."
"A key issue is to take care of obesity," said panel discussant Dr. Mauricio Tohen, chairman of psychiatry at the University of New Mexico in Albuquerque. "Regardless of the treatment, when there is obesity, the outcome will not be as good."
Efficacy vs. ‘generalizability’
Adjunctive personalized treatment was measured to help minimize the lack of assay sensitivity and internal validity typically inherent in comparative effectiveness trials, while also achieving more broadly applicable results.
"The right study depends on the right question," Dr. Tohen said. "If the question is whether a particular treatment has efficacy and is safe, then of course, we need an efficacy study. The problem with efficacy studies is that they limit the generalizability."
Instead, Dr. Tohen, who disclosed he spent more than a decade evaluating efficacy studies for both Eli Lily and AstraZeneca (makers of quetiapine), praised the metric of the adjunctive personalized treatments, and said a study should answer the question, "Of all the patients in my clinic, who will do better on which treatment? With efficacy studies you cannot answer that question because of the exclusion criteria that need to be taken into account."
In addition, Dr. Tohen said the metric likely would increase in importance under the Affordable Care Act, where efficacy is not the only consideration. "For example, if a patient relapses earlier, it might not be reimbursed, so asking what other outcomes we need to measure in [these kinds of] comparative effectiveness studies is very important."
In the CHOICE study, which had no placebo group, participating physicians were asked to track each patient’s dosage changes, missed doses, new medications added, discontinued medications, and the specific reasons for any of these changes. Changes that were made because of lack of effectiveness or intolerance were measured; however, planned dosage titrations according to normal scales were not considered necessary clinical adjustments but simply the regular course of treatment.
This heterogeneity of adjunctive treatment was seen by the investigators as a way to reflect "real world" practice, although they also noted it could be seen as a limitation to the study.
Another way the study was designed to reflect real-world practice was the inclusion of a broader-than-usual group of participants, and asking them for their feedback at the end of the study period.
"Before we put together these data, we held a stakeholder summit," Dr. Nierenberg said. "We invited patients and their advocates to help us interpret the study. They felt strongly that this was a reasonably positive study, because they thought that people really did get better. You could report these data toward the negative or the positive, but we reported toward the positive because that is what the stakeholders asked us to do." Dr. Tohen said that this kind of feedback in a comparative effectiveness study was another way these kinds of studies could help guide practice in the era of the Affordable Care Act.
As for who was actually admitted to the study, "exclusions were minimal," said Dr. Edward S. Friedman of the psychiatry department at the University of Pittsburgh. Those with a history of drug or alcohol dependence in the previous 30 days, a demonstrated intolerance to either study drug, or severe cardiovascular or renal disease were excluded, as were those with unstable thyroid disease, and pregnant or breastfeeding women.
Overall, the group was multiethnic, although nearly three-quarters were white and 20% were black. Roughly a third of the entire study population was employed, a third unemployed, and the rest were students, retirees, or those on disability.
The overall BISS score at enrollment averaged 56.1 (SD plus or minus 18.8). The average BISS depression score was 37.6 (SD plus or minus 14.0), and the average BISS mania score was 18.5 (SD plus or minus 12.1).
There were many comorbid psychiatric conditions, ranging from panic disorder to agoraphobia, although substance use was highest (61.4%). Current anxiety disorder also was prevalent at 58%.
"The CHOICE sample was similar in age and gender to previous efficacy studies," Dr. Friedman said. "It was more representative of the U.S. population than efficacy studies such as STEP-BD or LITMUS."
The 283 women (59% of the study) were more likely to report having spent a greater percentage of time depressed in the previous year, although the percentage of time spent in manic or hypomanic states was equal across both genders.
"This study was composed of sick individuals who had been sick for a considerable period of time," Dr. Friedman said.
For example, in the CHOICE study, 47% had been hospitalized for their bipolar disorder previously, vs. 43% in the LITMUS study. CHOICE participants had an attempted suicide rate of 36%, which fell between the 41% in the LITMUS trial and the 33.3% in an efficacy study conducted by Dr. Joseph R. Calabrese and his colleagues (J. Clin. Psychiatry 1999;60:79-88).
The average age for the first depressive episode for CHOICE participants was 16.4 years; the first manic episode tended to be at 20 years; and the first mood episode tended to be around 15.5 years. These data were similar to those in the LITMUS study, but differed from data in several efficacy studies such as the one by Dr. Calabrese, where the average age was 31.3 years.
"You might anticipate that in these specialty clinics we would have very few medication-naive patients, and yet we did," Dr. Friedman said during a postpanel audience participation session. "Looking for patients with a low threshold, we brought in patients who were very sick. They were also older, with lots of previous episodes. Maybe we missed younger patients who didn’t have as many previous episodes."
"These are the patients you would see in your practice," Dr. Nierenberg said before asking the audience what they think should be studied next in a comparative effectiveness trial. "We could look at the use of different antipsychotics, the combinations we use, with or without lamotrigine," he said.
"Most of the decisions that are made in medicine are made without evidence. Most of the things that are done are done without evidence. You have combinations that were never studied before or never even used before, and it’s across all medicine." Because medicine needs to be learning more, he said the audience needed to participate. "What are the questions that need to be asked?"
On Twitter @whitneymcknight
HOLLYWOOD, FLA. – Because there is no clinically significant difference in bipolar spectrum symptom relief offered by either lithium or quetiapine, which mood stabilizer clinicians choose to prescribe should largely depend on what side effects patients can tolerate.
That’s the conclusion of a panel of experts who presented data from the Bipolar CHOICE (Clinical Health Outcomes Initiative in Comparative Effectiveness for Bipolar Disorder) clinical trial at a meeting of the American Society of Clinical Psychopharmacology, formerly known as the New Clinical Drug Evaluation Unit meeting.
By randomly assigning the study’s 482 participating outpatient adults to either lithium or quetiapine, while allowing clinicians to prescribe adjunctive treatment as necessary; and by using a broader-than-usual set of inclusion criteria, the investigators hoped to provide clinicians with "real world" data on how the respective mood stabilizers perform.
"To my knowledge, this is the first randomized comparative effectiveness study of regular, expert care that looks at lithium versus quetiapine strategies of treatment, both with adjunctive personalized treatment when necessary," study coauthor Dr. Andrew A. Nierenberg told the audience in a well-attended session.
"We tortured the data to get a P value to pop out," said Dr. Nierenberg, who is the director of the Bipolar Clinic and Research Program at Massachusetts General Hospital in Boston. "The bottom line was that the treatment strategies, based on either lithium or quetiapine, were very similar. I think the clinical implication is that the choice of treatment really depends on someone’s tolerability. This may resurrect lithium as something [clinicians] should at least consider, because its use has gone down dramatically in the last 20 years or so."
Side effects such as metabolic or sleep disturbances were slightly less frequent in the lithium group, although the difference was not statistically significant; rates of suicidality and discontinuation of medication due to side effects were also similar across the two groups, according to Dr. David E. Kemp of the psychiatry department at Case Western Reserve University, Cleveland, and a study coauthor who also presented data during the session. Changes from baseline in mood stabilization were also virtually the same across the groups.
‘Rules of engagement’
The study enrolled adults aged 18-68 years who were at least mildly ill with either bipolar disorder I (68%) or II, with a Clinical Global Impression (CGI) score of at least 3, although the average CGI score was 4.5 (standard deviation of plus or minus 0.9). Of the 482 enrolled, 364 completed the study.
The so-called rules of engagement for clinicians from the 11 participating centers were that "they do everything they could to get their patients well," with the caveat that the lithium patient group not receive any antipsychotic, and the quetiapine patient group not receive lithium or any other antipsychotic.
"But they could get anything else that they needed," Dr. Nierenberg said.
At the time of enrollment, subjects were, in the estimation of their treating physician, experiencing symptoms that warranted a change in treatment, and that either lithium or quetiapine would be viable therapeutic options. Patients did not need to be lithium- or quetiapine-naive, but they could not have been treated with either drug in the previous 30 days. If they were already taking a second-generation antipsychotic, participants had to be willing to discontinue that medication and to be randomly assigned to either of the study drugs.
This strict adherence to one mood stabilizer, combined with adjunctive personalized treatments such as antidepressants or benzodiazepines, prescribed at the discretion of the treating clinician, ensured that the study tested the strategy of using either lithium or quetiapine as the base of treatment, Dr. Nierenberg said.
‘Similar’ results
Clinicians also were instructed to use the maximum tolerated dose of the respective mood stabilizer. For the 240 patients in the lithium group, the mean maximum tolerated dose was 1,007.5 mg, with the median dose being a "perfectly reasonable 900 mg," Dr. Nierenberg said. Blood lithium levels were taken at weeks 2, 16, and 24 and also were all found to be "reasonable," with most patients getting to at least 0.6 mEq/L by week 24.
For the 242 members of the quetiapine group, the maximum tolerated dose was 344.9 mg, with a median tolerated dose of 300 mg, which Dr. Nierenberg said was "what you would expect."
The overall result was that regardless of which mood stabilizer was prescribed along with adjunctive personalized treatment, the coprimary outcomes of the benefits and harms ratio from treatment effects over time (the CGI efficacy index), and the number of necessary clinical adjustments (changes in medication excluding titrations according to scale), were virtually parallel: For each primary outcome, the P value for the estimated change in baseline was less than .0001 for both lithium plus adjunctive treatment, and quetiapine plus adjunctive treatment.
Further, Dr. Nierenberg reported the "unexpected" result that at 6 months, about a quarter of each group was observed to be doing well without any adjunctive personalized treatments (23.8% of the lithium group; 27.3% of the quetiapine group; P = .14). "We thought that everybody would be on multiple things," Dr. Nierenberg said.
There was also no real difference in the time to discontinuation. "We were actually surprised that at 6 months, about three-quarters of the patients were still on the study drug, which was pretty good considering that this was a very ill population," Dr. Nierenberg said.
Using a CGI bipolar severity scale rating of up to 2, maintained for at least 8 weeks, the investigators found that overall, 20% were considered doing "really well"; it was also about 20% for the lithium plus adjunctive therapy group and the quetiapine and adjunctive therapy group (P = .14 for all three).
Because the Agency for Healthcare Research and Quality–funded study mandated that researchers end their investigation at precisely 36 months, Dr. Nierenberg said the study sites "randomized the 482 patients to 6 months of treatment within 36 months from start to finish," but he added that the investigators would have preferred to conduct a full 2-year study of participants.
"The good news was that most of the patients actually did improve substantially; the bad news is that while maybe a quarter did really well by the end of 6 months, we don’t know whether if we were to extend the study out further, we would see a difference between the two groups, whether they would continue to improve, or whether they would relapse," he said.
Adverse effects, predictors of response
Because study participants were followed for only 6 months, the long-term adverse effects of many years of exposure to either mood stabilizer, such as renal impairment with lithium or type II diabetes with quetiapine, were not considered, Dr. Kemp said. The adverse-effect profile for lithium generally includes a narrow therapeutic index; nausea, vomiting, and diarrhea; renal impairment; and hypothyroidism. Although Dr. Kemp said that the long-term adverse effects of quetiapine are understudied, known short-term side effects include the potential for sedation or somnolence, and weight gain.
Changes in baseline on the Frequency and Intensity of Side Effects Ratings for the quetiapine plus adjunctive personalized treatment arm were slightly more adverse than for the lithium and additional treatment arm. For the lithium plus adjunctive treatment group, the mean frequency of side effects was –1.41 (–1.78, –1.04 SD); the mean intensity of side effects was –1.48 (–1.80, –1.15 SD); and the mean level of impairment was –1.13 (–1.43, –0.82 SD). For the quetiapine and adjunctive treatment group, the mean frequency measure was –1.08 (–1.45, –0.72 SD); the intensity was –1.12 (–1.44, –0.79 SD); and the impairment was –0.77 (–1.07, –0.47 SD). For all scores in both groups, P was less than .001.
The changes from baseline on the Bipolar Index Severity Scale (BISS) overall for the quetiapine plus adjunctive treatment group was –28.56 (–10.91, –26.21 SD; P less than .0001). For the lithium plus adjunctive treatment group, it was –27.61 (–29.99, –25.24 SD; P less than .0001). The overall difference between the two groups was 0.94 (–2.10, 3.99 SD; P = .54).
Scores on the Longitudinal Interval Follow-up Evaluation Range of Impaired Functioning Tool also slightly favored lithium: –3.74 (–4.29, –3.19 SD) vs. –3.6 (–4.15, –3.07 SD) for quetiapine (P less than .0001 for all).
To the investigators’ surprise, among those with comorbid anxiety, the lithium plus adjunctive treatment group had fewer necessary clinical adjustments per month than did the quetiapine plus adjunctive treatment group, according to Dr. Nierenberg: –0.83 vs. 1.11 (P = .02).
"That seemed counterintuitive, and this difference was only with anxiety, not with any other comorbid psychiatric conditions," Dr. Nierenberg said. He hypothesized that it was possible benzodiazepines were used more frequently and easily with lithium than with quetiapine, but said future analyses would give a clearer answer. "We have a detailed database of every other medication used: when it was started, when it was stopped, and the reason why everything was done."
Not having current anxiety disorder was predictive of a better outcome (odds ratio, 1.81; P = .02), as was employment (OR, 1.67; P = .04). Those with bipolar II disorder responded better to treatment than those with bipolar disorder I, having an OR of response to treatment of nearly 1.8 P = .03). "That was a surprise to us, too," Dr. Nierenberg said.
Just over a quarter (27%) of the study population had metabolic syndrome, according to Dr. Kemp, who said this was not found to influence treatment outcomes.
Nearly half of the overall population was obese (48%), and slightly less than half (44%) had adipose. Prospective analysis indicated that patients with either obesity or adiposity tended to show less improvement in their CGI scores in a statistically significant way, regardless of which mood stabilizer they were on.
"Part of that might be due to overlapping pathophysiology between bipolar disorder and obesity," Dr. Kemp said. "Much of that is centered around inflammation in the central nervous system, as well as alterations in adipokines receptor levels and mitochondrial dysfunction."
"A key issue is to take care of obesity," said panel discussant Dr. Mauricio Tohen, chairman of psychiatry at the University of New Mexico in Albuquerque. "Regardless of the treatment, when there is obesity, the outcome will not be as good."
Efficacy vs. ‘generalizability’
Adjunctive personalized treatment was measured to help minimize the lack of assay sensitivity and internal validity typically inherent in comparative effectiveness trials, while also achieving more broadly applicable results.
"The right study depends on the right question," Dr. Tohen said. "If the question is whether a particular treatment has efficacy and is safe, then of course, we need an efficacy study. The problem with efficacy studies is that they limit the generalizability."
Instead, Dr. Tohen, who disclosed he spent more than a decade evaluating efficacy studies for both Eli Lily and AstraZeneca (makers of quetiapine), praised the metric of the adjunctive personalized treatments, and said a study should answer the question, "Of all the patients in my clinic, who will do better on which treatment? With efficacy studies you cannot answer that question because of the exclusion criteria that need to be taken into account."
In addition, Dr. Tohen said the metric likely would increase in importance under the Affordable Care Act, where efficacy is not the only consideration. "For example, if a patient relapses earlier, it might not be reimbursed, so asking what other outcomes we need to measure in [these kinds of] comparative effectiveness studies is very important."
In the CHOICE study, which had no placebo group, participating physicians were asked to track each patient’s dosage changes, missed doses, new medications added, discontinued medications, and the specific reasons for any of these changes. Changes that were made because of lack of effectiveness or intolerance were measured; however, planned dosage titrations according to normal scales were not considered necessary clinical adjustments but simply the regular course of treatment.
This heterogeneity of adjunctive treatment was seen by the investigators as a way to reflect "real world" practice, although they also noted it could be seen as a limitation to the study.
Another way the study was designed to reflect real-world practice was the inclusion of a broader-than-usual group of participants, and asking them for their feedback at the end of the study period.
"Before we put together these data, we held a stakeholder summit," Dr. Nierenberg said. "We invited patients and their advocates to help us interpret the study. They felt strongly that this was a reasonably positive study, because they thought that people really did get better. You could report these data toward the negative or the positive, but we reported toward the positive because that is what the stakeholders asked us to do." Dr. Tohen said that this kind of feedback in a comparative effectiveness study was another way these kinds of studies could help guide practice in the era of the Affordable Care Act.
As for who was actually admitted to the study, "exclusions were minimal," said Dr. Edward S. Friedman of the psychiatry department at the University of Pittsburgh. Those with a history of drug or alcohol dependence in the previous 30 days, a demonstrated intolerance to either study drug, or severe cardiovascular or renal disease were excluded, as were those with unstable thyroid disease, and pregnant or breastfeeding women.
Overall, the group was multiethnic, although nearly three-quarters were white and 20% were black. Roughly a third of the entire study population was employed, a third unemployed, and the rest were students, retirees, or those on disability.
The overall BISS score at enrollment averaged 56.1 (SD plus or minus 18.8). The average BISS depression score was 37.6 (SD plus or minus 14.0), and the average BISS mania score was 18.5 (SD plus or minus 12.1).
There were many comorbid psychiatric conditions, ranging from panic disorder to agoraphobia, although substance use was highest (61.4%). Current anxiety disorder also was prevalent at 58%.
"The CHOICE sample was similar in age and gender to previous efficacy studies," Dr. Friedman said. "It was more representative of the U.S. population than efficacy studies such as STEP-BD or LITMUS."
The 283 women (59% of the study) were more likely to report having spent a greater percentage of time depressed in the previous year, although the percentage of time spent in manic or hypomanic states was equal across both genders.
"This study was composed of sick individuals who had been sick for a considerable period of time," Dr. Friedman said.
For example, in the CHOICE study, 47% had been hospitalized for their bipolar disorder previously, vs. 43% in the LITMUS study. CHOICE participants had an attempted suicide rate of 36%, which fell between the 41% in the LITMUS trial and the 33.3% in an efficacy study conducted by Dr. Joseph R. Calabrese and his colleagues (J. Clin. Psychiatry 1999;60:79-88).
The average age for the first depressive episode for CHOICE participants was 16.4 years; the first manic episode tended to be at 20 years; and the first mood episode tended to be around 15.5 years. These data were similar to those in the LITMUS study, but differed from data in several efficacy studies such as the one by Dr. Calabrese, where the average age was 31.3 years.
"You might anticipate that in these specialty clinics we would have very few medication-naive patients, and yet we did," Dr. Friedman said during a postpanel audience participation session. "Looking for patients with a low threshold, we brought in patients who were very sick. They were also older, with lots of previous episodes. Maybe we missed younger patients who didn’t have as many previous episodes."
"These are the patients you would see in your practice," Dr. Nierenberg said before asking the audience what they think should be studied next in a comparative effectiveness trial. "We could look at the use of different antipsychotics, the combinations we use, with or without lamotrigine," he said.
"Most of the decisions that are made in medicine are made without evidence. Most of the things that are done are done without evidence. You have combinations that were never studied before or never even used before, and it’s across all medicine." Because medicine needs to be learning more, he said the audience needed to participate. "What are the questions that need to be asked?"
On Twitter @whitneymcknight
AT THE ASCP ANNUAL MEETING
Key clinical point: Determining whether to use lithium or quetiapine in bipolar I and II comes down to side effects.
Major finding: No clinically significant difference was found for bipolar I and bipolar II symptom improvements in patients given lithium or quetiapine (P less than .0001).
Data source: Multicenter, randomized comparative effectiveness study of 482 outpatients on the bipolar spectrum, followed for 6 months as they were given either lithium plus adjunctive therapy, quetiapine plus adjunctive therapy, or monotherapy of either mood stabilizer.
Disclosures: The Agency for Healthcare Research and Quality funded this study in its entirety.
Telepsychiatry is a tool that we must exploit
As psychiatrists, we are particularly attuned to the value of face-to-face contact with patients. After all, so much is communicated nonverbally.
Fortunately, telepsychiatry has the capacity to give us the information we need to provide effective interventions for patients with mental illness. Even patients with serious mental illness can benefit from these interventions.
Take, for example, a literature review of 390 studies using terms that included "schizophrenia" and/or "telepsychiatry," "telemedicine," or "telepsychology" (Clin. Schizophr. Relat. Psychoses 2014 [doi:10.3371/CSRP.KAFE.021513]). The review, conducted by Dr. John Kasckow of the Veterans Affairs Pittsburgh Health Care System, found that modalities involving the telephone, the Internet, and videoconferencing "appear to be feasible in patients with schizophrenia." Furthermore, they found that those modalities appear to improve patient outcomes, although they acknowledge that more research is needed.
A subset of patients that can benefit from telepsychiatry is those in correctional facilities. Another literature review that looked at the implementation of telepsychiatry in correctional facilities in seven states, including my own state of California, and found that the modality "may improve living conditions and safety inside correctional facilities" (Perm. J. 2013 Summer;17:80-6). This review, conducted by Stacie Anne Deslich of the Marshall University in South Charleston, W.Va., and her colleagues, also found that using telepsychiatry improved access and saved those facilities $12,000 to more than $1 million.
These researchers also called for more study, particularly a case-control examination of the cost of providing psychiatric care through telemedicine vs. face-to-face psychiatric treatment. Using telepsychiatry for this population of patients is particularly important in light of depth and breadth of untreated mental illness in correctional facilities, such as depression, anxiety, bipolar disorders, and schizophrenia. "In addition, costs for providers traveling to distant facilities have been a deterrent to providing adequate care to inmates," Ms. Deslich wrote.
Yet another population of patients that can benefit from telepsychiatry is those with mental illness who come to emergency departments. A program implemented in Elizabeth City, N.C., connected patients in the ED with psychiatric providers who were at remote locations using telemedicine carts that were equipped with wireless technology (ED Manag. 2013;25:121-4). The program’s administrators reported that almost 30% of the patients who had involuntary commitment orders were stabilized to the extent that those orders could be rescinded and they were discharged to outpatient care. Furthermore, the researchers reported, the average length of stay for ED patients who were discharged to inpatient treatment facilities dropped by more than half, from 48 hours to 22.5 hours.
Not so surprisingly, telepsychiatry also is establishing a solid track record among young patients. A study of the perspectives of psychiatrists who provide consultation services to schools found "students were more likely to disclose clinical information via video, compared with face-to-face contact" (Telemed. J.E. Health 2013; 19;794-9). However, the psychiatrists did express concerns about technological difficulties, logistics, and information sharing.
Telepsychiatry also is gaining a foothold in other areas, such as in geriatric and consultation psychiatry. In other settings, telepsychiatry is being introduced, and evidence is still accumulating.
The primary driver of telepsychiatry is the psychiatrist shortage. In 2009, a total of 77% of U.S. counties reported a shortage. Additionally, recent increases in coverage for mental health care create a demand for more psychiatrist time. These factors, coupled with an aging psychiatry workforce, led to a growing imbalance between supply and demand that telepsychiatry can help to alleviate. Telepsychiatry can increase the efficiency of psychiatric care by allowing one psychiatrist to serve patients in multiple settings without burdensome travel. Although telepsychiatry was first used more than 30 years ago, only recently have demographic, economic, and cultural trends led to its rapid expansion.
Opportunities for telepsychiatry implementation exist across the spectrum of psychiatric care. So far, research has shown little or no difference between the outcomes yielded by traditional care and telepsychiatry. Work remains to be done, but clearly telepsychiatry is here to stay.
Dr. Kornbluh is assistant medical director for program improvement and telepsychiatry at the California Department of State Hospitals.
As psychiatrists, we are particularly attuned to the value of face-to-face contact with patients. After all, so much is communicated nonverbally.
Fortunately, telepsychiatry has the capacity to give us the information we need to provide effective interventions for patients with mental illness. Even patients with serious mental illness can benefit from these interventions.
Take, for example, a literature review of 390 studies using terms that included "schizophrenia" and/or "telepsychiatry," "telemedicine," or "telepsychology" (Clin. Schizophr. Relat. Psychoses 2014 [doi:10.3371/CSRP.KAFE.021513]). The review, conducted by Dr. John Kasckow of the Veterans Affairs Pittsburgh Health Care System, found that modalities involving the telephone, the Internet, and videoconferencing "appear to be feasible in patients with schizophrenia." Furthermore, they found that those modalities appear to improve patient outcomes, although they acknowledge that more research is needed.
A subset of patients that can benefit from telepsychiatry is those in correctional facilities. Another literature review that looked at the implementation of telepsychiatry in correctional facilities in seven states, including my own state of California, and found that the modality "may improve living conditions and safety inside correctional facilities" (Perm. J. 2013 Summer;17:80-6). This review, conducted by Stacie Anne Deslich of the Marshall University in South Charleston, W.Va., and her colleagues, also found that using telepsychiatry improved access and saved those facilities $12,000 to more than $1 million.
These researchers also called for more study, particularly a case-control examination of the cost of providing psychiatric care through telemedicine vs. face-to-face psychiatric treatment. Using telepsychiatry for this population of patients is particularly important in light of depth and breadth of untreated mental illness in correctional facilities, such as depression, anxiety, bipolar disorders, and schizophrenia. "In addition, costs for providers traveling to distant facilities have been a deterrent to providing adequate care to inmates," Ms. Deslich wrote.
Yet another population of patients that can benefit from telepsychiatry is those with mental illness who come to emergency departments. A program implemented in Elizabeth City, N.C., connected patients in the ED with psychiatric providers who were at remote locations using telemedicine carts that were equipped with wireless technology (ED Manag. 2013;25:121-4). The program’s administrators reported that almost 30% of the patients who had involuntary commitment orders were stabilized to the extent that those orders could be rescinded and they were discharged to outpatient care. Furthermore, the researchers reported, the average length of stay for ED patients who were discharged to inpatient treatment facilities dropped by more than half, from 48 hours to 22.5 hours.
Not so surprisingly, telepsychiatry also is establishing a solid track record among young patients. A study of the perspectives of psychiatrists who provide consultation services to schools found "students were more likely to disclose clinical information via video, compared with face-to-face contact" (Telemed. J.E. Health 2013; 19;794-9). However, the psychiatrists did express concerns about technological difficulties, logistics, and information sharing.
Telepsychiatry also is gaining a foothold in other areas, such as in geriatric and consultation psychiatry. In other settings, telepsychiatry is being introduced, and evidence is still accumulating.
The primary driver of telepsychiatry is the psychiatrist shortage. In 2009, a total of 77% of U.S. counties reported a shortage. Additionally, recent increases in coverage for mental health care create a demand for more psychiatrist time. These factors, coupled with an aging psychiatry workforce, led to a growing imbalance between supply and demand that telepsychiatry can help to alleviate. Telepsychiatry can increase the efficiency of psychiatric care by allowing one psychiatrist to serve patients in multiple settings without burdensome travel. Although telepsychiatry was first used more than 30 years ago, only recently have demographic, economic, and cultural trends led to its rapid expansion.
Opportunities for telepsychiatry implementation exist across the spectrum of psychiatric care. So far, research has shown little or no difference between the outcomes yielded by traditional care and telepsychiatry. Work remains to be done, but clearly telepsychiatry is here to stay.
Dr. Kornbluh is assistant medical director for program improvement and telepsychiatry at the California Department of State Hospitals.
As psychiatrists, we are particularly attuned to the value of face-to-face contact with patients. After all, so much is communicated nonverbally.
Fortunately, telepsychiatry has the capacity to give us the information we need to provide effective interventions for patients with mental illness. Even patients with serious mental illness can benefit from these interventions.
Take, for example, a literature review of 390 studies using terms that included "schizophrenia" and/or "telepsychiatry," "telemedicine," or "telepsychology" (Clin. Schizophr. Relat. Psychoses 2014 [doi:10.3371/CSRP.KAFE.021513]). The review, conducted by Dr. John Kasckow of the Veterans Affairs Pittsburgh Health Care System, found that modalities involving the telephone, the Internet, and videoconferencing "appear to be feasible in patients with schizophrenia." Furthermore, they found that those modalities appear to improve patient outcomes, although they acknowledge that more research is needed.
A subset of patients that can benefit from telepsychiatry is those in correctional facilities. Another literature review that looked at the implementation of telepsychiatry in correctional facilities in seven states, including my own state of California, and found that the modality "may improve living conditions and safety inside correctional facilities" (Perm. J. 2013 Summer;17:80-6). This review, conducted by Stacie Anne Deslich of the Marshall University in South Charleston, W.Va., and her colleagues, also found that using telepsychiatry improved access and saved those facilities $12,000 to more than $1 million.
These researchers also called for more study, particularly a case-control examination of the cost of providing psychiatric care through telemedicine vs. face-to-face psychiatric treatment. Using telepsychiatry for this population of patients is particularly important in light of depth and breadth of untreated mental illness in correctional facilities, such as depression, anxiety, bipolar disorders, and schizophrenia. "In addition, costs for providers traveling to distant facilities have been a deterrent to providing adequate care to inmates," Ms. Deslich wrote.
Yet another population of patients that can benefit from telepsychiatry is those with mental illness who come to emergency departments. A program implemented in Elizabeth City, N.C., connected patients in the ED with psychiatric providers who were at remote locations using telemedicine carts that were equipped with wireless technology (ED Manag. 2013;25:121-4). The program’s administrators reported that almost 30% of the patients who had involuntary commitment orders were stabilized to the extent that those orders could be rescinded and they were discharged to outpatient care. Furthermore, the researchers reported, the average length of stay for ED patients who were discharged to inpatient treatment facilities dropped by more than half, from 48 hours to 22.5 hours.
Not so surprisingly, telepsychiatry also is establishing a solid track record among young patients. A study of the perspectives of psychiatrists who provide consultation services to schools found "students were more likely to disclose clinical information via video, compared with face-to-face contact" (Telemed. J.E. Health 2013; 19;794-9). However, the psychiatrists did express concerns about technological difficulties, logistics, and information sharing.
Telepsychiatry also is gaining a foothold in other areas, such as in geriatric and consultation psychiatry. In other settings, telepsychiatry is being introduced, and evidence is still accumulating.
The primary driver of telepsychiatry is the psychiatrist shortage. In 2009, a total of 77% of U.S. counties reported a shortage. Additionally, recent increases in coverage for mental health care create a demand for more psychiatrist time. These factors, coupled with an aging psychiatry workforce, led to a growing imbalance between supply and demand that telepsychiatry can help to alleviate. Telepsychiatry can increase the efficiency of psychiatric care by allowing one psychiatrist to serve patients in multiple settings without burdensome travel. Although telepsychiatry was first used more than 30 years ago, only recently have demographic, economic, and cultural trends led to its rapid expansion.
Opportunities for telepsychiatry implementation exist across the spectrum of psychiatric care. So far, research has shown little or no difference between the outcomes yielded by traditional care and telepsychiatry. Work remains to be done, but clearly telepsychiatry is here to stay.
Dr. Kornbluh is assistant medical director for program improvement and telepsychiatry at the California Department of State Hospitals.
Should lithium and ECT be used concurrently in geriatric patients?
Delirium has been described as a potential complication of concurrent lithium and electroconvulsive therapy (ECT) for depression, in association with a range of serum lithium levels. Although debate persists about the safety of continuing previously established lithium therapy during a course of ECT for mood symptoms, withholding lithium for 24 hours before administering ECT and measuring the serum lithium level before ECT were found to decrease the risk of post-ECT neurocognitive effects.1
We have found that the conventional practice of holding lithium for 24 hours before ECT might need to be re-evaluated in geriatric patients, as the following case demonstrates. Only 24 hours of holding lithium therapy might result in a lithium level sufficient to contribute to delirium after ECT.
CASE REPORT
An older woman with recurrent unipolar psychotic depression
Mrs. A, age 81, was admitted to the hospital with a 1-week history of depressed mood, anhedonia, insomnia, anergia, anorexia, and nihilistic somatic delusions that her organs were “rotting and shutting down.” Treatment included nortriptyline, 40 mg/d; lithium, 150 mg/d; and haloperidol, 0.5 mg/d. Her serum lithium level was 0.3 mEq/L (reference range, 0.6 to 1.2 mEq/L); the serum nortriptyline level was 68 ng/mL (reference range, 50 to 150 ng/mL). CT of the head and an electrocardiogram were unremarkable.
A twice-weekly course of ECT was initiated.
The day before Treatment 1 of ECT, the serum lithium level (drawn 12 hours after the last dose) was 0.4 mEq/L. Lithium was withheld 24 hours before ECT; nortriptyline and haloperidol were continued at prescribed dosages.
Right unilateral stimulation was used at 50%/mC energy (Thymatron DG, with methohexital anesthesia, and succinylcholine for muscle relaxation). Seizure duration, measured by EEG, was 57 seconds.
Mrs. A developed postictal delirium after the first 2 ECT sessions. The serum lithium level was unchanged. Subsequently, lithium treatment was discontinued and ECT was continued; once lithium was stopped, delirium resolved. ECT sessions 3 and 4 were uneventful, with no post-treatment delirium. Seizure duration for Treatment 4 was 58 seconds. She started breathing easily after all ECT sessions.
After Treatment 4, Mrs. A experienced full remission of depressive and psychotic symptoms. Repeat CT of head, after Treatment 4, was unchanged from baseline.
What is the role of lithium?
Mrs. A did not exhibit typical signs of lithium intoxication (diarrhea, vomiting, tremor). Notably, lithium has an intrinsic anticholinergic activity2; concurrent nortriptyline, a secondary amine tricyclic antidepressant with fewer anticholinergic side effects than other tricyclics,2 could precipitate delirium in a vulnerable patient secondary to excessive cumulative anticholinergic exposure.
No prolonged time-to-respiration or time-to-awakening occurred during treatments in which concurrent lithium and ECT were used; seizure duration with and without concurrent lithium was relatively similar.
There are potential complications of concurrent use of lithium and ECT:
• prolongation of the duration of muscle paralysis and apnea induced by commonly used neuromuscular-blocking agents (eg, succinylcholine)
• post-ECT cognitive disturbance.1,3,4
There is debate about the safety of continuing lithium during, or in close proximity to, ECT. In a case series of 12 patients who underwent combined lithium therapy and ECT, the authors concluded that this combination can be safe, regardless of age, as long as appropriate clinical monitoring is provided.4 In Mrs. A’s case, once post-ECT delirium was noted, lithium was discontinued for subsequent ECT sessions.
Because further ECT was uneventful without lithium, and no other clear acute cause of delirium could be identified, we concluded that lithium likely played a role in Mrs. A’s delirium. Notably, nortriptyline had been continued, suggesting that the degree of anticholinergic blockade provided by nortriptyline was insufficient to provoke delirium post-ECT in the absence of potentiation of this effect, as it had been when lithium also was used initially.
Guidelines for dosing and serum lithium concentrations in geriatric patients are not well-established; the current traditional range of 0.6 to 1.2 mEq/L, is too high for geriatric patients and can result in episodes of lithium toxicity, including delirium.5 Although our patient’s lithium level was below the reference range for all patients, a level of 0.3 mEq/L can be considered at the low end of the reference range for geriatric patients.5 Inasmuch as the lithium-assisted post-ECT delirium could represent a clinical sign of lithium toxicity, perhaps even a subtherapeutic level in a certain patient could be paradoxically “toxic.”
Although the serum lithium level in our patient remained below the toxic level for the general population (>1.5 mEq/L), delirium in a geriatric patient could result from:
• age-related changes in the pharmacokinetics of lithium, a water-soluble drug; these changes reduce renal clearance of the drug and extend plasma elimination half-life of a single dose to 36 hours, with the result that lithium remains in the body longer and necessitating a lower dosage (ie, a dosage that yields a serum level of approximately 0.5 mEq/L)
• the CNS tissue concentration of lithium, which can be high even though the serum level is not toxic
• an age-related increase in blood-brain barrier permeability, making the barrier more porous for drugs
• changes in blood-brain barrier permeability by post-ECT biochemical induction, with subsequent increased drug availability in the CNS.5,6
What we recommend
Possible interactions between lithium and ECT that lead to ECT-associated delirium need further elucidation, but discontinuing lithium during the course of ECT in a geriatric patient warrants your consideration. Following a safe interval after the last ECT session, lithium likely can be safely re-introduced 1) if there is clinical need and 2) as long as clinical surveillance for cognitive side effects is provided— especially if ECT will need to be reconsidered in the future.
Two additional considerations:
• Actively reassess lithium dosing in all geriatric psychiatric patients, especially those with renal insufficiency and other systemic metabolic considerations.
• Actively examine the use of all other anticholinergic agents in the course of evaluating a patient’s candidacy for ECT.
Disclosures
The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
1. American Psychiatric Association. The practice of electroconvulsive therapy: recommendations for treatment, training, and privileging. A task force report of the American Psychiatric Association. 2nd ed. Washington, DC: American Psychiatric Publishing; 2001.
2. Chew ML, Mulsant BH, Pollock BG, et al. Anticholinergic activity of 107 medications commonly used by older adults. J Am Geriatr Soc. 2008;56(7):1333-1341.
3. Hill GE, Wong KC, Hodges MR. Potentiation of succinylcholine neuromuscular blockade by lithium carbonate. Anesthesiology. 1976;44(5):439-442.
4. Dolenc TJ, Rasmussen KG. The safety of electroconvulsive therapy and lithium in combination: a case series and review of the literature. J ECT. 2005;21(3):165-170.
5. Shulman KI. Lithium for older adults with bipolar disorder: should it still be considered a first line agent? Drugs Aging. 2010;27(8):607-615.
6. Grandjean EM, Aubry JM. Lithium: updated human knowledge using an evidence-based approach. Part II: clinical pharmacology and therapeutic monitoring. CNS Drugs. 2009;23(4):331-349.
Delirium has been described as a potential complication of concurrent lithium and electroconvulsive therapy (ECT) for depression, in association with a range of serum lithium levels. Although debate persists about the safety of continuing previously established lithium therapy during a course of ECT for mood symptoms, withholding lithium for 24 hours before administering ECT and measuring the serum lithium level before ECT were found to decrease the risk of post-ECT neurocognitive effects.1
We have found that the conventional practice of holding lithium for 24 hours before ECT might need to be re-evaluated in geriatric patients, as the following case demonstrates. Only 24 hours of holding lithium therapy might result in a lithium level sufficient to contribute to delirium after ECT.
CASE REPORT
An older woman with recurrent unipolar psychotic depression
Mrs. A, age 81, was admitted to the hospital with a 1-week history of depressed mood, anhedonia, insomnia, anergia, anorexia, and nihilistic somatic delusions that her organs were “rotting and shutting down.” Treatment included nortriptyline, 40 mg/d; lithium, 150 mg/d; and haloperidol, 0.5 mg/d. Her serum lithium level was 0.3 mEq/L (reference range, 0.6 to 1.2 mEq/L); the serum nortriptyline level was 68 ng/mL (reference range, 50 to 150 ng/mL). CT of the head and an electrocardiogram were unremarkable.
A twice-weekly course of ECT was initiated.
The day before Treatment 1 of ECT, the serum lithium level (drawn 12 hours after the last dose) was 0.4 mEq/L. Lithium was withheld 24 hours before ECT; nortriptyline and haloperidol were continued at prescribed dosages.
Right unilateral stimulation was used at 50%/mC energy (Thymatron DG, with methohexital anesthesia, and succinylcholine for muscle relaxation). Seizure duration, measured by EEG, was 57 seconds.
Mrs. A developed postictal delirium after the first 2 ECT sessions. The serum lithium level was unchanged. Subsequently, lithium treatment was discontinued and ECT was continued; once lithium was stopped, delirium resolved. ECT sessions 3 and 4 were uneventful, with no post-treatment delirium. Seizure duration for Treatment 4 was 58 seconds. She started breathing easily after all ECT sessions.
After Treatment 4, Mrs. A experienced full remission of depressive and psychotic symptoms. Repeat CT of head, after Treatment 4, was unchanged from baseline.
What is the role of lithium?
Mrs. A did not exhibit typical signs of lithium intoxication (diarrhea, vomiting, tremor). Notably, lithium has an intrinsic anticholinergic activity2; concurrent nortriptyline, a secondary amine tricyclic antidepressant with fewer anticholinergic side effects than other tricyclics,2 could precipitate delirium in a vulnerable patient secondary to excessive cumulative anticholinergic exposure.
No prolonged time-to-respiration or time-to-awakening occurred during treatments in which concurrent lithium and ECT were used; seizure duration with and without concurrent lithium was relatively similar.
There are potential complications of concurrent use of lithium and ECT:
• prolongation of the duration of muscle paralysis and apnea induced by commonly used neuromuscular-blocking agents (eg, succinylcholine)
• post-ECT cognitive disturbance.1,3,4
There is debate about the safety of continuing lithium during, or in close proximity to, ECT. In a case series of 12 patients who underwent combined lithium therapy and ECT, the authors concluded that this combination can be safe, regardless of age, as long as appropriate clinical monitoring is provided.4 In Mrs. A’s case, once post-ECT delirium was noted, lithium was discontinued for subsequent ECT sessions.
Because further ECT was uneventful without lithium, and no other clear acute cause of delirium could be identified, we concluded that lithium likely played a role in Mrs. A’s delirium. Notably, nortriptyline had been continued, suggesting that the degree of anticholinergic blockade provided by nortriptyline was insufficient to provoke delirium post-ECT in the absence of potentiation of this effect, as it had been when lithium also was used initially.
Guidelines for dosing and serum lithium concentrations in geriatric patients are not well-established; the current traditional range of 0.6 to 1.2 mEq/L, is too high for geriatric patients and can result in episodes of lithium toxicity, including delirium.5 Although our patient’s lithium level was below the reference range for all patients, a level of 0.3 mEq/L can be considered at the low end of the reference range for geriatric patients.5 Inasmuch as the lithium-assisted post-ECT delirium could represent a clinical sign of lithium toxicity, perhaps even a subtherapeutic level in a certain patient could be paradoxically “toxic.”
Although the serum lithium level in our patient remained below the toxic level for the general population (>1.5 mEq/L), delirium in a geriatric patient could result from:
• age-related changes in the pharmacokinetics of lithium, a water-soluble drug; these changes reduce renal clearance of the drug and extend plasma elimination half-life of a single dose to 36 hours, with the result that lithium remains in the body longer and necessitating a lower dosage (ie, a dosage that yields a serum level of approximately 0.5 mEq/L)
• the CNS tissue concentration of lithium, which can be high even though the serum level is not toxic
• an age-related increase in blood-brain barrier permeability, making the barrier more porous for drugs
• changes in blood-brain barrier permeability by post-ECT biochemical induction, with subsequent increased drug availability in the CNS.5,6
What we recommend
Possible interactions between lithium and ECT that lead to ECT-associated delirium need further elucidation, but discontinuing lithium during the course of ECT in a geriatric patient warrants your consideration. Following a safe interval after the last ECT session, lithium likely can be safely re-introduced 1) if there is clinical need and 2) as long as clinical surveillance for cognitive side effects is provided— especially if ECT will need to be reconsidered in the future.
Two additional considerations:
• Actively reassess lithium dosing in all geriatric psychiatric patients, especially those with renal insufficiency and other systemic metabolic considerations.
• Actively examine the use of all other anticholinergic agents in the course of evaluating a patient’s candidacy for ECT.
Disclosures
The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
Delirium has been described as a potential complication of concurrent lithium and electroconvulsive therapy (ECT) for depression, in association with a range of serum lithium levels. Although debate persists about the safety of continuing previously established lithium therapy during a course of ECT for mood symptoms, withholding lithium for 24 hours before administering ECT and measuring the serum lithium level before ECT were found to decrease the risk of post-ECT neurocognitive effects.1
We have found that the conventional practice of holding lithium for 24 hours before ECT might need to be re-evaluated in geriatric patients, as the following case demonstrates. Only 24 hours of holding lithium therapy might result in a lithium level sufficient to contribute to delirium after ECT.
CASE REPORT
An older woman with recurrent unipolar psychotic depression
Mrs. A, age 81, was admitted to the hospital with a 1-week history of depressed mood, anhedonia, insomnia, anergia, anorexia, and nihilistic somatic delusions that her organs were “rotting and shutting down.” Treatment included nortriptyline, 40 mg/d; lithium, 150 mg/d; and haloperidol, 0.5 mg/d. Her serum lithium level was 0.3 mEq/L (reference range, 0.6 to 1.2 mEq/L); the serum nortriptyline level was 68 ng/mL (reference range, 50 to 150 ng/mL). CT of the head and an electrocardiogram were unremarkable.
A twice-weekly course of ECT was initiated.
The day before Treatment 1 of ECT, the serum lithium level (drawn 12 hours after the last dose) was 0.4 mEq/L. Lithium was withheld 24 hours before ECT; nortriptyline and haloperidol were continued at prescribed dosages.
Right unilateral stimulation was used at 50%/mC energy (Thymatron DG, with methohexital anesthesia, and succinylcholine for muscle relaxation). Seizure duration, measured by EEG, was 57 seconds.
Mrs. A developed postictal delirium after the first 2 ECT sessions. The serum lithium level was unchanged. Subsequently, lithium treatment was discontinued and ECT was continued; once lithium was stopped, delirium resolved. ECT sessions 3 and 4 were uneventful, with no post-treatment delirium. Seizure duration for Treatment 4 was 58 seconds. She started breathing easily after all ECT sessions.
After Treatment 4, Mrs. A experienced full remission of depressive and psychotic symptoms. Repeat CT of head, after Treatment 4, was unchanged from baseline.
What is the role of lithium?
Mrs. A did not exhibit typical signs of lithium intoxication (diarrhea, vomiting, tremor). Notably, lithium has an intrinsic anticholinergic activity2; concurrent nortriptyline, a secondary amine tricyclic antidepressant with fewer anticholinergic side effects than other tricyclics,2 could precipitate delirium in a vulnerable patient secondary to excessive cumulative anticholinergic exposure.
No prolonged time-to-respiration or time-to-awakening occurred during treatments in which concurrent lithium and ECT were used; seizure duration with and without concurrent lithium was relatively similar.
There are potential complications of concurrent use of lithium and ECT:
• prolongation of the duration of muscle paralysis and apnea induced by commonly used neuromuscular-blocking agents (eg, succinylcholine)
• post-ECT cognitive disturbance.1,3,4
There is debate about the safety of continuing lithium during, or in close proximity to, ECT. In a case series of 12 patients who underwent combined lithium therapy and ECT, the authors concluded that this combination can be safe, regardless of age, as long as appropriate clinical monitoring is provided.4 In Mrs. A’s case, once post-ECT delirium was noted, lithium was discontinued for subsequent ECT sessions.
Because further ECT was uneventful without lithium, and no other clear acute cause of delirium could be identified, we concluded that lithium likely played a role in Mrs. A’s delirium. Notably, nortriptyline had been continued, suggesting that the degree of anticholinergic blockade provided by nortriptyline was insufficient to provoke delirium post-ECT in the absence of potentiation of this effect, as it had been when lithium also was used initially.
Guidelines for dosing and serum lithium concentrations in geriatric patients are not well-established; the current traditional range of 0.6 to 1.2 mEq/L, is too high for geriatric patients and can result in episodes of lithium toxicity, including delirium.5 Although our patient’s lithium level was below the reference range for all patients, a level of 0.3 mEq/L can be considered at the low end of the reference range for geriatric patients.5 Inasmuch as the lithium-assisted post-ECT delirium could represent a clinical sign of lithium toxicity, perhaps even a subtherapeutic level in a certain patient could be paradoxically “toxic.”
Although the serum lithium level in our patient remained below the toxic level for the general population (>1.5 mEq/L), delirium in a geriatric patient could result from:
• age-related changes in the pharmacokinetics of lithium, a water-soluble drug; these changes reduce renal clearance of the drug and extend plasma elimination half-life of a single dose to 36 hours, with the result that lithium remains in the body longer and necessitating a lower dosage (ie, a dosage that yields a serum level of approximately 0.5 mEq/L)
• the CNS tissue concentration of lithium, which can be high even though the serum level is not toxic
• an age-related increase in blood-brain barrier permeability, making the barrier more porous for drugs
• changes in blood-brain barrier permeability by post-ECT biochemical induction, with subsequent increased drug availability in the CNS.5,6
What we recommend
Possible interactions between lithium and ECT that lead to ECT-associated delirium need further elucidation, but discontinuing lithium during the course of ECT in a geriatric patient warrants your consideration. Following a safe interval after the last ECT session, lithium likely can be safely re-introduced 1) if there is clinical need and 2) as long as clinical surveillance for cognitive side effects is provided— especially if ECT will need to be reconsidered in the future.
Two additional considerations:
• Actively reassess lithium dosing in all geriatric psychiatric patients, especially those with renal insufficiency and other systemic metabolic considerations.
• Actively examine the use of all other anticholinergic agents in the course of evaluating a patient’s candidacy for ECT.
Disclosures
The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
1. American Psychiatric Association. The practice of electroconvulsive therapy: recommendations for treatment, training, and privileging. A task force report of the American Psychiatric Association. 2nd ed. Washington, DC: American Psychiatric Publishing; 2001.
2. Chew ML, Mulsant BH, Pollock BG, et al. Anticholinergic activity of 107 medications commonly used by older adults. J Am Geriatr Soc. 2008;56(7):1333-1341.
3. Hill GE, Wong KC, Hodges MR. Potentiation of succinylcholine neuromuscular blockade by lithium carbonate. Anesthesiology. 1976;44(5):439-442.
4. Dolenc TJ, Rasmussen KG. The safety of electroconvulsive therapy and lithium in combination: a case series and review of the literature. J ECT. 2005;21(3):165-170.
5. Shulman KI. Lithium for older adults with bipolar disorder: should it still be considered a first line agent? Drugs Aging. 2010;27(8):607-615.
6. Grandjean EM, Aubry JM. Lithium: updated human knowledge using an evidence-based approach. Part II: clinical pharmacology and therapeutic monitoring. CNS Drugs. 2009;23(4):331-349.
1. American Psychiatric Association. The practice of electroconvulsive therapy: recommendations for treatment, training, and privileging. A task force report of the American Psychiatric Association. 2nd ed. Washington, DC: American Psychiatric Publishing; 2001.
2. Chew ML, Mulsant BH, Pollock BG, et al. Anticholinergic activity of 107 medications commonly used by older adults. J Am Geriatr Soc. 2008;56(7):1333-1341.
3. Hill GE, Wong KC, Hodges MR. Potentiation of succinylcholine neuromuscular blockade by lithium carbonate. Anesthesiology. 1976;44(5):439-442.
4. Dolenc TJ, Rasmussen KG. The safety of electroconvulsive therapy and lithium in combination: a case series and review of the literature. J ECT. 2005;21(3):165-170.
5. Shulman KI. Lithium for older adults with bipolar disorder: should it still be considered a first line agent? Drugs Aging. 2010;27(8):607-615.
6. Grandjean EM, Aubry JM. Lithium: updated human knowledge using an evidence-based approach. Part II: clinical pharmacology and therapeutic monitoring. CNS Drugs. 2009;23(4):331-349.