LayerRx Mapping ID
157
Slot System
Featured Buckets
Featured Buckets Admin
Medscape Lead Concept
877

For couples seeking to conceive, offer advice on reducing the risk of schizophrenia in their child

Article Type
Changed
Mon, 04/16/2018 - 14:33
Display Headline
For couples seeking to conceive, offer advice on reducing the risk of schizophrenia in their child

I explained to them that schizophrenia is a neurodevelopmental syndrome that comprises hundreds of different disorders of genetic or non-genetic etiology, all of which share a similar psychotic phenotype. Although the various genetic causes of schizophre­nia are difficult to prevent—but may be prevented in the future with epi­genetic techniques—the many non-genetic (environmental) pathways to schizophrenia can be avoided to significantly reduce the incidence of schizophrenia by 40% to 50%, accord­ing to some estimates.

I will share what I told this couple, because even couples without any family history of psychosis may have a child who develops schizophrenia because of a variety of environmental risk factors.

Genetic risk factors
One-half of the 20,000 genes in the 23 chromosomes of the human genome participate in constructing and sculpt­ing the extremely intricate and com­plex human brain. There are many ways that genetic factors can increase the risk of schizophrenia,1 and only some are transmitted by parents:

Risk genes. More than 30 risk genes have been identified as heritable in schizophrenia. They are spread over many chromosomes and more are likely to be discovered. Most of those risk genes regulate glutamate— not dopamine—pathways, and each increases the risk by 2% to 4%.

Copy number variations (CNVs) are pro­duced via meiosis mishaps, where 1 or 3 alleles of certain genes are formed instead of the usual 2. A high frequency of CNVs have been found in schizophrenia com­pared with the general population—but also are found in autism and bipolar disorders—and are believed to disrupt brain development in various ways.

De novo mutations. Recent studies on large samples of people with schizo­phrenia (50,000 to 100,000) uncovered a much higher rate of mutations (some code for proteins while others are non­sense mutations that code for noth­ing). Obviously, these mutations led to anomalous neurodevelopment.

There are hundreds, maybe thou­sands, of genetic subtypes within the schizophrenia syndrome. Advances in epigenetics, which allow silencing of culprit genes or overexpression of protective genes, one day may enable psychiatric geneticists to prevent schizo­phrenia in fetuses at risk.

Non-genetic risk factors
Just as with the genetic patho-genic heterogeneity, the schizo­phrenia syndrome can be caused by numerous environmental adverse events,2 many of which can be avoided, including:

Older paternal age (>45) at time of conception doubles or triples the risk of schizophrenia3 as well as autism and bipolar disorder. Aging sperm are associated with a higher rate of DNA fragmentation and genetic mutations.

Prenatal complications, especially during the second trimester when CNS development takes place. These adverse prenatal events skew fetal brain development to produce psy­chosis in adulthood and can be mini­mized with optimal prenatal care, which sadly is lacking among the poor. These include:
   • Vaginal infections before preg­nancy,4 such as herpes simplex virus, can cause fetal brain inflammation and increased risk of schizophrenia.
   • Infections during pregnancy— whether bacterial, viral, or proto­zoan (Toxoplasma gondii)—have been shown to significantly increase the risk of schizophrenia in offspring.5 An increase in serum C-reactive protein during pregnancy also is a biomarker of increased risk.
   • Poor diet, especially starva­tion, can double or triple the risk of schizophrenia.
   • Vitamin deficiency, especially folate and vitamin D, are critical for normal brain development.6 Vitamin D is vital to mitigate neuroinflammation.
   Smoking before and during pregnancy.4
   • Medical illness during preg­nancy, especially gestational diabetes, increases the risk of schizophrenia in the fetus by 800%.7
   • Severe stress during pregnancy, such as the death of the spouse, dou­bles the risk of schizophrenia.2
   • Schizophrenia risk is 400% to 500% higher among those born and raised in an urban area, compared with a rural area.8
   • Babies born in northern latitudes, such as in Sweden, Norway, or Canada, have a 10-fold risk of schizophrenia in adulthood compared with babies born near the equator.6 This has been attrib­uted to lack of sunshine and the risk of severe vitamin D deficiency in north­ern latitudes.
   • High maternal body mass index during the first trimester7 increases the child’s risk of schizophrenia.
   Low number of prenatal vis­its is associated with higher risk of schizophrenia.
   • Obstetric complications that cause hypoxia and a low Apgar score after birth increase the risk of schizophrenia. This includes long labor, cord around the neck, meco­nium spillage into the amniotic fluid, and mechanical injury with forceps delivery.
   • Infection in the newborn shortly after birth.

Severe physical or sexual abuse before age 5 is associated with increased risk of schizophrenia in adulthood.2 This may be because of stress-induced epigenetic mechanisms (silencing or overexpressing certain genes).

Migration has been shown to increase the risk of schizophrenia by 3 to 5 fold. The exact reason is unclear, but it could be a combination of social stress, exposure to new types of germs, less sunshine, and even a different diet.

 

 

My advice to the couple? Get a good obstetrician well before conception; get the mother immunized against infections; eat a lot of fish (omega-3 fatty acids); take ade­quate doses of folate and vitamin D, per­haps even choline9; avoid smoking before and during pregnancy; adopt a healthy, balanced diet; avoid excessive weight gain and/or gestational diabetes; avoid contact with people with infections; avoid expo­sure to cat feces (toxoplasmosis); sched­ule frequent prenatal visits; and hope for a smooth and uneventful delivery and a newborn with an Apgar score of 9 or 10. All this will greatly reduce the non-genetic risks of schizophrenia, but is unlikely to modify the genetic risks. However, it has been shown that a combination of both genetic and non-genetic risk factors is associated with a more severe form of schizophrenia.10

Optimal prenatal and postnatal care can be helpful for couples with a family history of schizophrenia (without mov­ing to deliver their baby in a rural village near the equator). However, if their child starts using marijuana during adolescence, all bets are off. The risk of schizophrenia and serious cortical tissue loss increases dramatically when a carrier of risk genes use Cannabis. But that’s another editorial, to be read by clinicians in states where marijuana has been (foolishly, I believe) legalized.

References


1. Rodriguez-Murillo L, Gogos JA, Karayiorgou M. The genetic architecture of schizophrenia: new mutations and emerging paradigms. Annu Rev Med. 2012;63:63-80.
2. van Os J, Kenis G, Rutten BP. The environment and schizophrenia. Nature. 2010;468(7321):203-212.
3. Brown AS, Schefer CA, Wyatt RJ, et al. Paternal age and risk of schizophrenia in adult offspring. Am J Psychiatry. 2002;159(9):1528-1533.
4. Betts KS, Williams GM, Najman JM, et al. Maternal prenatal infection, early susceptibility to illness and adult psychotic experiences: a birth cohort study. Schizophr Res. 2014;156(2- 3):161-167.
5. Brown AS, Derkits EJ. Prental infection and schizophrenia: a review of epidemiologic and translational studies. Am J Psychiatry. 2010;167(3):261-280.
6. Kinney DK, Teixeira P, Hsu D, et al. Relation of schizophrenia prevalence to latitude, climate, fish consumption, infant mortality, and skin color: a role for prenatal vitamin d deficiency and infections? Schizophr Bull. 2009;35(3): 582-595.
7. Kawai M, Minabe Y, Takagai S, et al. Poor maternal care and high maternal body mass index in pregnancy as a risk factor for schizophrenia in offspring. Acta Psychiatry Scand. 2004;110(4):257-263.
8. Kelly BD, O’Callaghan E, Waddington JL, et al. Schizophrenia and the city: a review of literature and prospective study of psychosis and urbanicity in Ireland. Schizophr Res. 2010;116(1):75-89.
9. Ross RG, Hunter SK, McCarthy L, et al. Perinatal choline effects on neonatal pathophysiology related to later schizophrenia risk. Am J Psychiatry. 2013; 170(3):290-298.
10. Maynard TM, Sikich L, Lieberman JA, et al. Neural development, cell-cell signaling, and the “two-hit” hypothesis of schizophrenia. Schizophr Bull. 2001;27(3): 457-476.

Article PDF
Author and Disclosure Information

 

Henry A. Nasrallah, MD
Editor-in-Chief

Issue
Current Psychiatry - 13(8)
Publications
Topics
Page Number
11-12, 44
Legacy Keywords
schizophrenia, family history of schizophrenia, mitigating risk of schizophrenia, child
Sections
Author and Disclosure Information

 

Henry A. Nasrallah, MD
Editor-in-Chief

Author and Disclosure Information

 

Henry A. Nasrallah, MD
Editor-in-Chief

Article PDF
Article PDF
Related Articles

I explained to them that schizophrenia is a neurodevelopmental syndrome that comprises hundreds of different disorders of genetic or non-genetic etiology, all of which share a similar psychotic phenotype. Although the various genetic causes of schizophre­nia are difficult to prevent—but may be prevented in the future with epi­genetic techniques—the many non-genetic (environmental) pathways to schizophrenia can be avoided to significantly reduce the incidence of schizophrenia by 40% to 50%, accord­ing to some estimates.

I will share what I told this couple, because even couples without any family history of psychosis may have a child who develops schizophrenia because of a variety of environmental risk factors.

Genetic risk factors
One-half of the 20,000 genes in the 23 chromosomes of the human genome participate in constructing and sculpt­ing the extremely intricate and com­plex human brain. There are many ways that genetic factors can increase the risk of schizophrenia,1 and only some are transmitted by parents:

Risk genes. More than 30 risk genes have been identified as heritable in schizophrenia. They are spread over many chromosomes and more are likely to be discovered. Most of those risk genes regulate glutamate— not dopamine—pathways, and each increases the risk by 2% to 4%.

Copy number variations (CNVs) are pro­duced via meiosis mishaps, where 1 or 3 alleles of certain genes are formed instead of the usual 2. A high frequency of CNVs have been found in schizophrenia com­pared with the general population—but also are found in autism and bipolar disorders—and are believed to disrupt brain development in various ways.

De novo mutations. Recent studies on large samples of people with schizo­phrenia (50,000 to 100,000) uncovered a much higher rate of mutations (some code for proteins while others are non­sense mutations that code for noth­ing). Obviously, these mutations led to anomalous neurodevelopment.

There are hundreds, maybe thou­sands, of genetic subtypes within the schizophrenia syndrome. Advances in epigenetics, which allow silencing of culprit genes or overexpression of protective genes, one day may enable psychiatric geneticists to prevent schizo­phrenia in fetuses at risk.

Non-genetic risk factors
Just as with the genetic patho-genic heterogeneity, the schizo­phrenia syndrome can be caused by numerous environmental adverse events,2 many of which can be avoided, including:

Older paternal age (>45) at time of conception doubles or triples the risk of schizophrenia3 as well as autism and bipolar disorder. Aging sperm are associated with a higher rate of DNA fragmentation and genetic mutations.

Prenatal complications, especially during the second trimester when CNS development takes place. These adverse prenatal events skew fetal brain development to produce psy­chosis in adulthood and can be mini­mized with optimal prenatal care, which sadly is lacking among the poor. These include:
   • Vaginal infections before preg­nancy,4 such as herpes simplex virus, can cause fetal brain inflammation and increased risk of schizophrenia.
   • Infections during pregnancy— whether bacterial, viral, or proto­zoan (Toxoplasma gondii)—have been shown to significantly increase the risk of schizophrenia in offspring.5 An increase in serum C-reactive protein during pregnancy also is a biomarker of increased risk.
   • Poor diet, especially starva­tion, can double or triple the risk of schizophrenia.
   • Vitamin deficiency, especially folate and vitamin D, are critical for normal brain development.6 Vitamin D is vital to mitigate neuroinflammation.
   Smoking before and during pregnancy.4
   • Medical illness during preg­nancy, especially gestational diabetes, increases the risk of schizophrenia in the fetus by 800%.7
   • Severe stress during pregnancy, such as the death of the spouse, dou­bles the risk of schizophrenia.2
   • Schizophrenia risk is 400% to 500% higher among those born and raised in an urban area, compared with a rural area.8
   • Babies born in northern latitudes, such as in Sweden, Norway, or Canada, have a 10-fold risk of schizophrenia in adulthood compared with babies born near the equator.6 This has been attrib­uted to lack of sunshine and the risk of severe vitamin D deficiency in north­ern latitudes.
   • High maternal body mass index during the first trimester7 increases the child’s risk of schizophrenia.
   Low number of prenatal vis­its is associated with higher risk of schizophrenia.
   • Obstetric complications that cause hypoxia and a low Apgar score after birth increase the risk of schizophrenia. This includes long labor, cord around the neck, meco­nium spillage into the amniotic fluid, and mechanical injury with forceps delivery.
   • Infection in the newborn shortly after birth.

Severe physical or sexual abuse before age 5 is associated with increased risk of schizophrenia in adulthood.2 This may be because of stress-induced epigenetic mechanisms (silencing or overexpressing certain genes).

Migration has been shown to increase the risk of schizophrenia by 3 to 5 fold. The exact reason is unclear, but it could be a combination of social stress, exposure to new types of germs, less sunshine, and even a different diet.

 

 

My advice to the couple? Get a good obstetrician well before conception; get the mother immunized against infections; eat a lot of fish (omega-3 fatty acids); take ade­quate doses of folate and vitamin D, per­haps even choline9; avoid smoking before and during pregnancy; adopt a healthy, balanced diet; avoid excessive weight gain and/or gestational diabetes; avoid contact with people with infections; avoid expo­sure to cat feces (toxoplasmosis); sched­ule frequent prenatal visits; and hope for a smooth and uneventful delivery and a newborn with an Apgar score of 9 or 10. All this will greatly reduce the non-genetic risks of schizophrenia, but is unlikely to modify the genetic risks. However, it has been shown that a combination of both genetic and non-genetic risk factors is associated with a more severe form of schizophrenia.10

Optimal prenatal and postnatal care can be helpful for couples with a family history of schizophrenia (without mov­ing to deliver their baby in a rural village near the equator). However, if their child starts using marijuana during adolescence, all bets are off. The risk of schizophrenia and serious cortical tissue loss increases dramatically when a carrier of risk genes use Cannabis. But that’s another editorial, to be read by clinicians in states where marijuana has been (foolishly, I believe) legalized.

I explained to them that schizophrenia is a neurodevelopmental syndrome that comprises hundreds of different disorders of genetic or non-genetic etiology, all of which share a similar psychotic phenotype. Although the various genetic causes of schizophre­nia are difficult to prevent—but may be prevented in the future with epi­genetic techniques—the many non-genetic (environmental) pathways to schizophrenia can be avoided to significantly reduce the incidence of schizophrenia by 40% to 50%, accord­ing to some estimates.

I will share what I told this couple, because even couples without any family history of psychosis may have a child who develops schizophrenia because of a variety of environmental risk factors.

Genetic risk factors
One-half of the 20,000 genes in the 23 chromosomes of the human genome participate in constructing and sculpt­ing the extremely intricate and com­plex human brain. There are many ways that genetic factors can increase the risk of schizophrenia,1 and only some are transmitted by parents:

Risk genes. More than 30 risk genes have been identified as heritable in schizophrenia. They are spread over many chromosomes and more are likely to be discovered. Most of those risk genes regulate glutamate— not dopamine—pathways, and each increases the risk by 2% to 4%.

Copy number variations (CNVs) are pro­duced via meiosis mishaps, where 1 or 3 alleles of certain genes are formed instead of the usual 2. A high frequency of CNVs have been found in schizophrenia com­pared with the general population—but also are found in autism and bipolar disorders—and are believed to disrupt brain development in various ways.

De novo mutations. Recent studies on large samples of people with schizo­phrenia (50,000 to 100,000) uncovered a much higher rate of mutations (some code for proteins while others are non­sense mutations that code for noth­ing). Obviously, these mutations led to anomalous neurodevelopment.

There are hundreds, maybe thou­sands, of genetic subtypes within the schizophrenia syndrome. Advances in epigenetics, which allow silencing of culprit genes or overexpression of protective genes, one day may enable psychiatric geneticists to prevent schizo­phrenia in fetuses at risk.

Non-genetic risk factors
Just as with the genetic patho-genic heterogeneity, the schizo­phrenia syndrome can be caused by numerous environmental adverse events,2 many of which can be avoided, including:

Older paternal age (>45) at time of conception doubles or triples the risk of schizophrenia3 as well as autism and bipolar disorder. Aging sperm are associated with a higher rate of DNA fragmentation and genetic mutations.

Prenatal complications, especially during the second trimester when CNS development takes place. These adverse prenatal events skew fetal brain development to produce psy­chosis in adulthood and can be mini­mized with optimal prenatal care, which sadly is lacking among the poor. These include:
   • Vaginal infections before preg­nancy,4 such as herpes simplex virus, can cause fetal brain inflammation and increased risk of schizophrenia.
   • Infections during pregnancy— whether bacterial, viral, or proto­zoan (Toxoplasma gondii)—have been shown to significantly increase the risk of schizophrenia in offspring.5 An increase in serum C-reactive protein during pregnancy also is a biomarker of increased risk.
   • Poor diet, especially starva­tion, can double or triple the risk of schizophrenia.
   • Vitamin deficiency, especially folate and vitamin D, are critical for normal brain development.6 Vitamin D is vital to mitigate neuroinflammation.
   Smoking before and during pregnancy.4
   • Medical illness during preg­nancy, especially gestational diabetes, increases the risk of schizophrenia in the fetus by 800%.7
   • Severe stress during pregnancy, such as the death of the spouse, dou­bles the risk of schizophrenia.2
   • Schizophrenia risk is 400% to 500% higher among those born and raised in an urban area, compared with a rural area.8
   • Babies born in northern latitudes, such as in Sweden, Norway, or Canada, have a 10-fold risk of schizophrenia in adulthood compared with babies born near the equator.6 This has been attrib­uted to lack of sunshine and the risk of severe vitamin D deficiency in north­ern latitudes.
   • High maternal body mass index during the first trimester7 increases the child’s risk of schizophrenia.
   Low number of prenatal vis­its is associated with higher risk of schizophrenia.
   • Obstetric complications that cause hypoxia and a low Apgar score after birth increase the risk of schizophrenia. This includes long labor, cord around the neck, meco­nium spillage into the amniotic fluid, and mechanical injury with forceps delivery.
   • Infection in the newborn shortly after birth.

Severe physical or sexual abuse before age 5 is associated with increased risk of schizophrenia in adulthood.2 This may be because of stress-induced epigenetic mechanisms (silencing or overexpressing certain genes).

Migration has been shown to increase the risk of schizophrenia by 3 to 5 fold. The exact reason is unclear, but it could be a combination of social stress, exposure to new types of germs, less sunshine, and even a different diet.

 

 

My advice to the couple? Get a good obstetrician well before conception; get the mother immunized against infections; eat a lot of fish (omega-3 fatty acids); take ade­quate doses of folate and vitamin D, per­haps even choline9; avoid smoking before and during pregnancy; adopt a healthy, balanced diet; avoid excessive weight gain and/or gestational diabetes; avoid contact with people with infections; avoid expo­sure to cat feces (toxoplasmosis); sched­ule frequent prenatal visits; and hope for a smooth and uneventful delivery and a newborn with an Apgar score of 9 or 10. All this will greatly reduce the non-genetic risks of schizophrenia, but is unlikely to modify the genetic risks. However, it has been shown that a combination of both genetic and non-genetic risk factors is associated with a more severe form of schizophrenia.10

Optimal prenatal and postnatal care can be helpful for couples with a family history of schizophrenia (without mov­ing to deliver their baby in a rural village near the equator). However, if their child starts using marijuana during adolescence, all bets are off. The risk of schizophrenia and serious cortical tissue loss increases dramatically when a carrier of risk genes use Cannabis. But that’s another editorial, to be read by clinicians in states where marijuana has been (foolishly, I believe) legalized.

References


1. Rodriguez-Murillo L, Gogos JA, Karayiorgou M. The genetic architecture of schizophrenia: new mutations and emerging paradigms. Annu Rev Med. 2012;63:63-80.
2. van Os J, Kenis G, Rutten BP. The environment and schizophrenia. Nature. 2010;468(7321):203-212.
3. Brown AS, Schefer CA, Wyatt RJ, et al. Paternal age and risk of schizophrenia in adult offspring. Am J Psychiatry. 2002;159(9):1528-1533.
4. Betts KS, Williams GM, Najman JM, et al. Maternal prenatal infection, early susceptibility to illness and adult psychotic experiences: a birth cohort study. Schizophr Res. 2014;156(2- 3):161-167.
5. Brown AS, Derkits EJ. Prental infection and schizophrenia: a review of epidemiologic and translational studies. Am J Psychiatry. 2010;167(3):261-280.
6. Kinney DK, Teixeira P, Hsu D, et al. Relation of schizophrenia prevalence to latitude, climate, fish consumption, infant mortality, and skin color: a role for prenatal vitamin d deficiency and infections? Schizophr Bull. 2009;35(3): 582-595.
7. Kawai M, Minabe Y, Takagai S, et al. Poor maternal care and high maternal body mass index in pregnancy as a risk factor for schizophrenia in offspring. Acta Psychiatry Scand. 2004;110(4):257-263.
8. Kelly BD, O’Callaghan E, Waddington JL, et al. Schizophrenia and the city: a review of literature and prospective study of psychosis and urbanicity in Ireland. Schizophr Res. 2010;116(1):75-89.
9. Ross RG, Hunter SK, McCarthy L, et al. Perinatal choline effects on neonatal pathophysiology related to later schizophrenia risk. Am J Psychiatry. 2013; 170(3):290-298.
10. Maynard TM, Sikich L, Lieberman JA, et al. Neural development, cell-cell signaling, and the “two-hit” hypothesis of schizophrenia. Schizophr Bull. 2001;27(3): 457-476.

References


1. Rodriguez-Murillo L, Gogos JA, Karayiorgou M. The genetic architecture of schizophrenia: new mutations and emerging paradigms. Annu Rev Med. 2012;63:63-80.
2. van Os J, Kenis G, Rutten BP. The environment and schizophrenia. Nature. 2010;468(7321):203-212.
3. Brown AS, Schefer CA, Wyatt RJ, et al. Paternal age and risk of schizophrenia in adult offspring. Am J Psychiatry. 2002;159(9):1528-1533.
4. Betts KS, Williams GM, Najman JM, et al. Maternal prenatal infection, early susceptibility to illness and adult psychotic experiences: a birth cohort study. Schizophr Res. 2014;156(2- 3):161-167.
5. Brown AS, Derkits EJ. Prental infection and schizophrenia: a review of epidemiologic and translational studies. Am J Psychiatry. 2010;167(3):261-280.
6. Kinney DK, Teixeira P, Hsu D, et al. Relation of schizophrenia prevalence to latitude, climate, fish consumption, infant mortality, and skin color: a role for prenatal vitamin d deficiency and infections? Schizophr Bull. 2009;35(3): 582-595.
7. Kawai M, Minabe Y, Takagai S, et al. Poor maternal care and high maternal body mass index in pregnancy as a risk factor for schizophrenia in offspring. Acta Psychiatry Scand. 2004;110(4):257-263.
8. Kelly BD, O’Callaghan E, Waddington JL, et al. Schizophrenia and the city: a review of literature and prospective study of psychosis and urbanicity in Ireland. Schizophr Res. 2010;116(1):75-89.
9. Ross RG, Hunter SK, McCarthy L, et al. Perinatal choline effects on neonatal pathophysiology related to later schizophrenia risk. Am J Psychiatry. 2013; 170(3):290-298.
10. Maynard TM, Sikich L, Lieberman JA, et al. Neural development, cell-cell signaling, and the “two-hit” hypothesis of schizophrenia. Schizophr Bull. 2001;27(3): 457-476.

Issue
Current Psychiatry - 13(8)
Issue
Current Psychiatry - 13(8)
Page Number
11-12, 44
Page Number
11-12, 44
Publications
Publications
Topics
Article Type
Display Headline
For couples seeking to conceive, offer advice on reducing the risk of schizophrenia in their child
Display Headline
For couples seeking to conceive, offer advice on reducing the risk of schizophrenia in their child
Legacy Keywords
schizophrenia, family history of schizophrenia, mitigating risk of schizophrenia, child
Legacy Keywords
schizophrenia, family history of schizophrenia, mitigating risk of schizophrenia, child
Sections
Article PDF Media

Dissociation found to mediate ketamine’s antidepressive effects

Article Type
Changed
Fri, 01/18/2019 - 13:43
Display Headline
Dissociation found to mediate ketamine’s antidepressive effects

HOLLYWOOD, FLA. – Dissociative side effects in patients given a ketamine infusion to treat either major depressive disorder or bipolar disorder predicted a more robust antidepressive response, according to a small secondary analysis.

"Patients who don’t have acute dissociation are more likely not to have antidepressant efficacy in the postinfusion period," Dr. Mark J. Niciu said during an interview discussing his poster presentation at a meeting of the American Society of Clinical Psychopharmacology, formerly known as the New Clinical Drug Evaluation Unit meeting.

"The patients with more disassociation might be the ones to have greater antidepressant efficacy, but they are also the patients we need to keep a closer clinical eye on because they’re having perceptual alterations during the postinfusion period," he said.

Dr. Niciu, a clinical research fellow at the National Institute of Mental Health in Bethesda, Md., reviewed data from 108 treatment-resistant inpatients who met criteria for major depressive disorder or bipolar I or II and were given a subanesthetic ketamine infusion. They examined whether dissociation and psychotic-like experiences, as measured by the Clinician Administered Dissociative States Scale (CADSS), the Brief Psychiatric Rating Scale (BPRS), and the Young Mania Rating Scale (YMRS), and vital sign changes correlated with improvements in the Hamilton Depression Rating Scale (HDRS) at 40 minutes and 230 minutes post infusion, and at 1 and 7 days post infusion.

Pearson correlations indicated that there was a significant association between increased CADSS scores at 40 minutes post infusion and improvement with ketamine in HDRS scores at 230 minutes (r = –0.35, P = .007). Changes in the YMRS or BPRS Positive Symptom score at 40 minutes did not significantly correlate with HDRS improvement at any time point with ketamine. Similarly, none of the vital signs analyzed (changes in systolic or diastolic blood pressure and pulse) significantly correlated to HDRS change.

The question of whether there was an "unblinding" effect was of concern to Dr. Niciu. "The subjects who received ketamine that had greater disassociation might also expect to have greater antidepressive efficacy post infusion," he said. To account for that possibility, he said some researchers are using more active placebos such as midazolam, but he did not think that it was a complete solution because the ideal active placebo would affect glutamate, dopamine, and noradrenaline without having an antidepressant effect.

The overall goal is to discover medications that have keen effects on glutamate receptors such as NMDA (N-methyl-D-aspartate) receptors, as ketamine does, but that do not also have dissociative side effects. "But maybe that’s not possible," Dr. Niciu said. "Maybe we need to have some degree of dissociation as a proxy for the strength of the NMDA receptor blockade because of its antidepressant effects downstream post infusion."

This study was funded by the Intramural Research Program at the National Institute of Mental Health, a NARSAD Independent Investigator Award, and a Brain and Behavior Mood Disorders Research Award. Both of the awards were given to Dr. Carlos A. Zarate.

[email protected]

On Twitter @whitneymcknight

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
Dissociative, ketamine, major depressive disorder, bipolar disorder, antidepressive response, Dr. Mark J. Niciu,
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

HOLLYWOOD, FLA. – Dissociative side effects in patients given a ketamine infusion to treat either major depressive disorder or bipolar disorder predicted a more robust antidepressive response, according to a small secondary analysis.

"Patients who don’t have acute dissociation are more likely not to have antidepressant efficacy in the postinfusion period," Dr. Mark J. Niciu said during an interview discussing his poster presentation at a meeting of the American Society of Clinical Psychopharmacology, formerly known as the New Clinical Drug Evaluation Unit meeting.

"The patients with more disassociation might be the ones to have greater antidepressant efficacy, but they are also the patients we need to keep a closer clinical eye on because they’re having perceptual alterations during the postinfusion period," he said.

Dr. Niciu, a clinical research fellow at the National Institute of Mental Health in Bethesda, Md., reviewed data from 108 treatment-resistant inpatients who met criteria for major depressive disorder or bipolar I or II and were given a subanesthetic ketamine infusion. They examined whether dissociation and psychotic-like experiences, as measured by the Clinician Administered Dissociative States Scale (CADSS), the Brief Psychiatric Rating Scale (BPRS), and the Young Mania Rating Scale (YMRS), and vital sign changes correlated with improvements in the Hamilton Depression Rating Scale (HDRS) at 40 minutes and 230 minutes post infusion, and at 1 and 7 days post infusion.

Pearson correlations indicated that there was a significant association between increased CADSS scores at 40 minutes post infusion and improvement with ketamine in HDRS scores at 230 minutes (r = –0.35, P = .007). Changes in the YMRS or BPRS Positive Symptom score at 40 minutes did not significantly correlate with HDRS improvement at any time point with ketamine. Similarly, none of the vital signs analyzed (changes in systolic or diastolic blood pressure and pulse) significantly correlated to HDRS change.

The question of whether there was an "unblinding" effect was of concern to Dr. Niciu. "The subjects who received ketamine that had greater disassociation might also expect to have greater antidepressive efficacy post infusion," he said. To account for that possibility, he said some researchers are using more active placebos such as midazolam, but he did not think that it was a complete solution because the ideal active placebo would affect glutamate, dopamine, and noradrenaline without having an antidepressant effect.

The overall goal is to discover medications that have keen effects on glutamate receptors such as NMDA (N-methyl-D-aspartate) receptors, as ketamine does, but that do not also have dissociative side effects. "But maybe that’s not possible," Dr. Niciu said. "Maybe we need to have some degree of dissociation as a proxy for the strength of the NMDA receptor blockade because of its antidepressant effects downstream post infusion."

This study was funded by the Intramural Research Program at the National Institute of Mental Health, a NARSAD Independent Investigator Award, and a Brain and Behavior Mood Disorders Research Award. Both of the awards were given to Dr. Carlos A. Zarate.

[email protected]

On Twitter @whitneymcknight

HOLLYWOOD, FLA. – Dissociative side effects in patients given a ketamine infusion to treat either major depressive disorder or bipolar disorder predicted a more robust antidepressive response, according to a small secondary analysis.

"Patients who don’t have acute dissociation are more likely not to have antidepressant efficacy in the postinfusion period," Dr. Mark J. Niciu said during an interview discussing his poster presentation at a meeting of the American Society of Clinical Psychopharmacology, formerly known as the New Clinical Drug Evaluation Unit meeting.

"The patients with more disassociation might be the ones to have greater antidepressant efficacy, but they are also the patients we need to keep a closer clinical eye on because they’re having perceptual alterations during the postinfusion period," he said.

Dr. Niciu, a clinical research fellow at the National Institute of Mental Health in Bethesda, Md., reviewed data from 108 treatment-resistant inpatients who met criteria for major depressive disorder or bipolar I or II and were given a subanesthetic ketamine infusion. They examined whether dissociation and psychotic-like experiences, as measured by the Clinician Administered Dissociative States Scale (CADSS), the Brief Psychiatric Rating Scale (BPRS), and the Young Mania Rating Scale (YMRS), and vital sign changes correlated with improvements in the Hamilton Depression Rating Scale (HDRS) at 40 minutes and 230 minutes post infusion, and at 1 and 7 days post infusion.

Pearson correlations indicated that there was a significant association between increased CADSS scores at 40 minutes post infusion and improvement with ketamine in HDRS scores at 230 minutes (r = –0.35, P = .007). Changes in the YMRS or BPRS Positive Symptom score at 40 minutes did not significantly correlate with HDRS improvement at any time point with ketamine. Similarly, none of the vital signs analyzed (changes in systolic or diastolic blood pressure and pulse) significantly correlated to HDRS change.

The question of whether there was an "unblinding" effect was of concern to Dr. Niciu. "The subjects who received ketamine that had greater disassociation might also expect to have greater antidepressive efficacy post infusion," he said. To account for that possibility, he said some researchers are using more active placebos such as midazolam, but he did not think that it was a complete solution because the ideal active placebo would affect glutamate, dopamine, and noradrenaline without having an antidepressant effect.

The overall goal is to discover medications that have keen effects on glutamate receptors such as NMDA (N-methyl-D-aspartate) receptors, as ketamine does, but that do not also have dissociative side effects. "But maybe that’s not possible," Dr. Niciu said. "Maybe we need to have some degree of dissociation as a proxy for the strength of the NMDA receptor blockade because of its antidepressant effects downstream post infusion."

This study was funded by the Intramural Research Program at the National Institute of Mental Health, a NARSAD Independent Investigator Award, and a Brain and Behavior Mood Disorders Research Award. Both of the awards were given to Dr. Carlos A. Zarate.

[email protected]

On Twitter @whitneymcknight

Publications
Publications
Topics
Article Type
Display Headline
Dissociation found to mediate ketamine’s antidepressive effects
Display Headline
Dissociation found to mediate ketamine’s antidepressive effects
Legacy Keywords
Dissociative, ketamine, major depressive disorder, bipolar disorder, antidepressive response, Dr. Mark J. Niciu,
Legacy Keywords
Dissociative, ketamine, major depressive disorder, bipolar disorder, antidepressive response, Dr. Mark J. Niciu,
Sections
Article Source

AT THE ASCP ANNUAL MEETING

PURLs Copyright

Inside the Article

Vitals

Key clinical point: Some degree of dissociation might be needed "as a proxy for the strength of the NMDA receptor blockade because of its antidepressant effects downstream post infusion."

Major finding: A significant association was found between increased CADSS scores at 40 minutes post infusion and improvement with ketamine in HDRS scores at 230 minutes (r = –0.35, P = .007).

Data source: Secondary analysis of 108 inpatients treated for MDD or BP I or II with ketamine infusion.

Disclosures: This study was funded by the Intramural Research Program at the National Institute of Mental Health, a NARSAD Independent Investigator Award, and a Brain and Behavior Mood Disorders Research Award. Both of the awards were given to Dr. Carlos A. Zarate.

Lurasidone monotherapy improves quality of life in bipolar I

Article Type
Changed
Fri, 01/18/2019 - 13:43
Display Headline
Lurasidone monotherapy improves quality of life in bipolar I

HOLLYWOOD, FLA. – Lurasidone as monotherapy in the dosage range of either 20-60 mg/day* or 80-120 mg/day significantly improved functioning and quality-of-life in patients with bipolar depression, a post hoc analysis has shown.

In a previously published study of 318 patients randomized to either lurasidone dosage groups or placebo, the drug’s association at both dose ranges with reduced Montgomery-sberg Depression Scale total scores and Clinical Global Impressions scale for bipolar depression severity scores from baseline to week 6 was significant. The study also noted that both lurasidone groups had significant improvements in patient-reported measures of quality of life and functional impairment, compared with the placebo group (P less than .001 for each) (Am. J. Psychiatry 2014;171:160-8).

The findings led Dr. Terence A. Ketter, professor of psychiatry and chief of the bipolar disorders clinic at Stanford (Calif.) University, to wonder how the improvements in functionality and quality of life had happened in such relatively short order.

"It kind of makes sense that it’s related to the mood improvement," Dr. Ketter said in an interview during a poster session at a meeting of the American Society of Clinical Psychopharmacology, formerly known as the New Clinical Drug Evaluation Unit meeting.

To investigate further, Dr. Ketter and his colleagues performed a mediation regression analysis using data from phase III of the original trial. The results were that reduced depressive symptoms from baseline to week 6 mediated the effect of lurasidone on Sheehan Disability Scale functional recovery and Quality of Life Enjoyment and Satisfaction Questionnaire scores at week 6 (P less than .05 for each).

"This suggests that, at least in the first 6 weeks, most of the improvement [in functionality and quality of life] is related to the improvement in depressive symptoms," Dr. Ketter said. "Functional recovery is something that takes months, and it may be that beyond this by a month or two, you’re looking at a degree of mood improvement by duration interaction to get functional improvement." He added that there likely would be a "huge improvement" in cognition, simply because of the removal of depressive symptoms.

"Is it enough to be able to go out and get a job? Well, maybe not. Maybe you need things to integrate for months before you can go out and work." But, he added, over time, the less interference with one’s cognitive ability, the greater likelihood that person is employable.

In the original study in bipolar I patients, "the company wanted to know which was the right dose," Dr. Ketter said. "As it turned out, both worked. The higher one was a little bit harder to tolerate, but not horribly so."

As a result, Dr. Ketter said the drug has a "pretty flexible label." In his own practice, he said he starts patients on monotherapy with lurasidone 20 mg/day at dinner time, titrating upward in 20-mg increments each week until his patients reach the dosage that works best for them. "The average dose seems to be about 60 mg/day," he said.

Lurasidone, a second-generation antipsychotic, originally was approved by the Food and Drug Administration in 2010 to treat schizophrenia in adults; the indication was expanded in 2013 to include bipolar I, either as monotherapy or as an adjunct to lithium or valproate.

Because of its relatively low impact on metabolic function and its low sedative effect, compared with other second-generation antipsychotics, Dr. Ketter said he favors using lurasidone in his patients. "It’s like an easier to use quetiapine," he noted.

This study was supported by Sunovion Pharmaceuticals. Dr. Ketter disclosed that he has received funding from Sunovion, as well as from AstraZeneca Pharmaceuticals, Cephalon, Eli Lilly, and others.

[email protected]

On Twitter @whitneymcknight

*Correction, 7/2/2014: An earlier version of this story misstated the lurasidone dosage range.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
Lurasidone, monotherapy, functioning, quality-of-life, bipolar depression,
Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

HOLLYWOOD, FLA. – Lurasidone as monotherapy in the dosage range of either 20-60 mg/day* or 80-120 mg/day significantly improved functioning and quality-of-life in patients with bipolar depression, a post hoc analysis has shown.

In a previously published study of 318 patients randomized to either lurasidone dosage groups or placebo, the drug’s association at both dose ranges with reduced Montgomery-sberg Depression Scale total scores and Clinical Global Impressions scale for bipolar depression severity scores from baseline to week 6 was significant. The study also noted that both lurasidone groups had significant improvements in patient-reported measures of quality of life and functional impairment, compared with the placebo group (P less than .001 for each) (Am. J. Psychiatry 2014;171:160-8).

The findings led Dr. Terence A. Ketter, professor of psychiatry and chief of the bipolar disorders clinic at Stanford (Calif.) University, to wonder how the improvements in functionality and quality of life had happened in such relatively short order.

"It kind of makes sense that it’s related to the mood improvement," Dr. Ketter said in an interview during a poster session at a meeting of the American Society of Clinical Psychopharmacology, formerly known as the New Clinical Drug Evaluation Unit meeting.

To investigate further, Dr. Ketter and his colleagues performed a mediation regression analysis using data from phase III of the original trial. The results were that reduced depressive symptoms from baseline to week 6 mediated the effect of lurasidone on Sheehan Disability Scale functional recovery and Quality of Life Enjoyment and Satisfaction Questionnaire scores at week 6 (P less than .05 for each).

"This suggests that, at least in the first 6 weeks, most of the improvement [in functionality and quality of life] is related to the improvement in depressive symptoms," Dr. Ketter said. "Functional recovery is something that takes months, and it may be that beyond this by a month or two, you’re looking at a degree of mood improvement by duration interaction to get functional improvement." He added that there likely would be a "huge improvement" in cognition, simply because of the removal of depressive symptoms.

"Is it enough to be able to go out and get a job? Well, maybe not. Maybe you need things to integrate for months before you can go out and work." But, he added, over time, the less interference with one’s cognitive ability, the greater likelihood that person is employable.

In the original study in bipolar I patients, "the company wanted to know which was the right dose," Dr. Ketter said. "As it turned out, both worked. The higher one was a little bit harder to tolerate, but not horribly so."

As a result, Dr. Ketter said the drug has a "pretty flexible label." In his own practice, he said he starts patients on monotherapy with lurasidone 20 mg/day at dinner time, titrating upward in 20-mg increments each week until his patients reach the dosage that works best for them. "The average dose seems to be about 60 mg/day," he said.

Lurasidone, a second-generation antipsychotic, originally was approved by the Food and Drug Administration in 2010 to treat schizophrenia in adults; the indication was expanded in 2013 to include bipolar I, either as monotherapy or as an adjunct to lithium or valproate.

Because of its relatively low impact on metabolic function and its low sedative effect, compared with other second-generation antipsychotics, Dr. Ketter said he favors using lurasidone in his patients. "It’s like an easier to use quetiapine," he noted.

This study was supported by Sunovion Pharmaceuticals. Dr. Ketter disclosed that he has received funding from Sunovion, as well as from AstraZeneca Pharmaceuticals, Cephalon, Eli Lilly, and others.

[email protected]

On Twitter @whitneymcknight

*Correction, 7/2/2014: An earlier version of this story misstated the lurasidone dosage range.

HOLLYWOOD, FLA. – Lurasidone as monotherapy in the dosage range of either 20-60 mg/day* or 80-120 mg/day significantly improved functioning and quality-of-life in patients with bipolar depression, a post hoc analysis has shown.

In a previously published study of 318 patients randomized to either lurasidone dosage groups or placebo, the drug’s association at both dose ranges with reduced Montgomery-sberg Depression Scale total scores and Clinical Global Impressions scale for bipolar depression severity scores from baseline to week 6 was significant. The study also noted that both lurasidone groups had significant improvements in patient-reported measures of quality of life and functional impairment, compared with the placebo group (P less than .001 for each) (Am. J. Psychiatry 2014;171:160-8).

The findings led Dr. Terence A. Ketter, professor of psychiatry and chief of the bipolar disorders clinic at Stanford (Calif.) University, to wonder how the improvements in functionality and quality of life had happened in such relatively short order.

"It kind of makes sense that it’s related to the mood improvement," Dr. Ketter said in an interview during a poster session at a meeting of the American Society of Clinical Psychopharmacology, formerly known as the New Clinical Drug Evaluation Unit meeting.

To investigate further, Dr. Ketter and his colleagues performed a mediation regression analysis using data from phase III of the original trial. The results were that reduced depressive symptoms from baseline to week 6 mediated the effect of lurasidone on Sheehan Disability Scale functional recovery and Quality of Life Enjoyment and Satisfaction Questionnaire scores at week 6 (P less than .05 for each).

"This suggests that, at least in the first 6 weeks, most of the improvement [in functionality and quality of life] is related to the improvement in depressive symptoms," Dr. Ketter said. "Functional recovery is something that takes months, and it may be that beyond this by a month or two, you’re looking at a degree of mood improvement by duration interaction to get functional improvement." He added that there likely would be a "huge improvement" in cognition, simply because of the removal of depressive symptoms.

"Is it enough to be able to go out and get a job? Well, maybe not. Maybe you need things to integrate for months before you can go out and work." But, he added, over time, the less interference with one’s cognitive ability, the greater likelihood that person is employable.

In the original study in bipolar I patients, "the company wanted to know which was the right dose," Dr. Ketter said. "As it turned out, both worked. The higher one was a little bit harder to tolerate, but not horribly so."

As a result, Dr. Ketter said the drug has a "pretty flexible label." In his own practice, he said he starts patients on monotherapy with lurasidone 20 mg/day at dinner time, titrating upward in 20-mg increments each week until his patients reach the dosage that works best for them. "The average dose seems to be about 60 mg/day," he said.

Lurasidone, a second-generation antipsychotic, originally was approved by the Food and Drug Administration in 2010 to treat schizophrenia in adults; the indication was expanded in 2013 to include bipolar I, either as monotherapy or as an adjunct to lithium or valproate.

Because of its relatively low impact on metabolic function and its low sedative effect, compared with other second-generation antipsychotics, Dr. Ketter said he favors using lurasidone in his patients. "It’s like an easier to use quetiapine," he noted.

This study was supported by Sunovion Pharmaceuticals. Dr. Ketter disclosed that he has received funding from Sunovion, as well as from AstraZeneca Pharmaceuticals, Cephalon, Eli Lilly, and others.

[email protected]

On Twitter @whitneymcknight

*Correction, 7/2/2014: An earlier version of this story misstated the lurasidone dosage range.

Publications
Publications
Topics
Article Type
Display Headline
Lurasidone monotherapy improves quality of life in bipolar I
Display Headline
Lurasidone monotherapy improves quality of life in bipolar I
Legacy Keywords
Lurasidone, monotherapy, functioning, quality-of-life, bipolar depression,
Legacy Keywords
Lurasidone, monotherapy, functioning, quality-of-life, bipolar depression,
Sections
Article Source

AT THE ASCP ANNUAL MEETING

PURLs Copyright

Inside the Article

Vitals

Key clinical point: Lurasidone appears to reduce depressive symptoms in patients with bipolar I depression without the metabolic complications characteristic of other second-generation antipsychotics.

Major finding: In 318 patients with bipolar I depression, reduced depressive symptoms from baseline to week 6 mediated the effect of lurasidone vs. placebo (P less than .05 in each) on functioning and quality of life.

Data source: A post hoc analysis of phase III data from a randomized, double-blind, placebo controlled trial in 318 intent-to-treat patients with bipolar I.

Disclosures: This study was supported by Sunovion Pharmaceuticals. Dr. Ketter disclosed he has received funding from Sunovion, as well as from AstraZeneca Pharmaceuticals, Cephalon, Eli Lilly, and others.

ADHD or bipolar disorder?

Article Type
Changed
Thu, 03/28/2019 - 15:44
Display Headline
ADHD or bipolar disorder?

Author and Disclosure Information

Dr. Baum is Clinical Assistant Professor of Pediatrics, Nationwide Children’s Hospital, Columbus, Ohio.

Issue
Current Psychiatry - 13(7)
Publications
Topics
Legacy Keywords
ADHD, bipolar disorder, child, children
Sections
Author and Disclosure Information

Dr. Baum is Clinical Assistant Professor of Pediatrics, Nationwide Children’s Hospital, Columbus, Ohio.

Author and Disclosure Information

Dr. Baum is Clinical Assistant Professor of Pediatrics, Nationwide Children’s Hospital, Columbus, Ohio.

Related Articles

Issue
Current Psychiatry - 13(7)
Issue
Current Psychiatry - 13(7)
Publications
Publications
Topics
Article Type
Display Headline
ADHD or bipolar disorder?
Display Headline
ADHD or bipolar disorder?
Legacy Keywords
ADHD, bipolar disorder, child, children
Legacy Keywords
ADHD, bipolar disorder, child, children
Sections
Disallow All Ads
Alternative CME
Use ProPublica

Confused and nearly naked after going on spending sprees

Article Type
Changed
Tue, 12/11/2018 - 15:29
Display Headline
Confused and nearly naked after going on spending sprees

CASE Nearly naked
Mr. A, age 68, is found sitting in his car, wear­ing only a jacket, underpants, and boots. He speaks of spreading a message about Osama bin Laden and “taking a census.” Police officers bring him to a hospital emergency depart­ment for evaluation.

The examining clinician determines that Mr. A is a danger to himself and others because of mental illness, leading to admission to our state psychiatric hospital.

Mr. A’s wife describes recent spend­ing sprees with large purchases. She had obtained a restraining order against her hus­band because of his threatening remarks and behaviors. Within days of the order issuance, he got a home equity loan and purchased a $300,000 house.

The medical history is notable for type 2 diabetes mellitus. Although he is not tak­ing medications, his blood sugar is well controlled. Other than an initial resting heart rate of 116 beats per minute, vital signs are stable and within normal lim­its. Physical examination is unremarkable. Screening laboratory studies are notable for mildly elevated hepatic function, which approaches normal range several days after admission.

Mr. A reports a remote history of alcohol abuse but says he had not been drinking recently, and does not detail his pattern of use. Urine toxicology screen is negative for all substances of abuse.

Mental status examination reveals dishev­eled appearance, motor agitation, pressured speech, labile affect, loosening of associa­tions, grandiose delusions, and auditory hal­lucinations. Mr. A’s thought processes are grossly disorganized, such that we could not gather a meaningful history. He believes God is speaking directly to him about plans to build a carousel at Disney World. He makes strange gestures with his hands throughout the inter­view, as if attempting to trace the shapes of letters and numbers. He frequently speaks of seeing an array of colors. Cognitive examina­tion reveals a score of 5 of 30 on the Montreal Cognitive Assessment (Figure 1), indicating a severe impairment in neurocognitive func­tioning. He demonstrates limited insight and markedly impaired judgment, and denies hav­ing a mental illness.


What should be the next step in managing Mr. A?
a) obtain records from other facilities and collateral history
b) start an antipsychotic
c) order a brain MRI
d) start an alcohol withdrawal protocol


The authors’ observations
Mr. A showed elements of mania, psycho­sis, and delirium. We considered a broad differential diagnosis (Table). Mr. A initially could not provide reliable or accurate information. The least invasive next step was to obtain additional history from his wife and other medical records to refine the differential diagnosis.



HISTORY Bizarre behavior
Mr. A allows staff to speak with his wife and obtain records from a psychiatric hospitaliza­tion 3 years earlier. Mrs. A reports significant and rapid changes in her husband’s behav­ior and personality over 3 months, but does not describe a recent alcohol relapse. Mr. A sleeps very little, remaining awake and active throughout the night. He frequently rear­ranges the furniture in their home for no clear reason. Once, he knocked on the door of a young female neighbor asking if she found him attractive.

Mr. A has a significant criminal history. Approximately 30 years ago, he was charged with attempted murder of his ex-wife and he had faced charges of attempted kidnapping and assaulting a police officer. However, he has no recent legal issues.

Mr. A has a history of episodes that are simi­lar to this presentation. Seven years ago, he impulsively purchased a $650,000 house after his fourth wife died. He then had a $90,000 heart-shaped pool installed. He also drove a tractor through his stepdaughter’s car for no apparent reason. Also, 3 years ago, he displayed symptoms similar to his current presentation, including insomnia, irritability, and grandios­ity. He engaged in strange behaviors, such as dressing up and imitating homeless people at his church.

During the hospitalization 3 years ago, cli­nicians gave Mr. A a diagnosis of bipolar dis­order, current episode manic, and delirium of an unclear cause. A medical workup, includ­ing brain MRI, did not uncover a basis for his delirium. Antipsychotics (risperidone and per­phenazine) and mood stabilizers (lithium and valproic acid), stabilized his condition; after 7 weeks, Mr. A was discharged, but he did not pursue outpatient psychiatric care.


What is the most likely DSM-5 diagnosis?
   a) major neurocognitive disorder (dementia)
   b) alcohol use disorder (eg, Wernicke- Korsakoff syndrome)
   c) delirium secondary to mania
   d) psychotic disorder


The authors’ observations

DSM-51 suggests a stepwise approach to diagnosis, with consideration of:
   • signs and symptoms
   • substance use
   • general medical condition
   • developmental conflict or stage
   • whether a mental disorder is present.

 

 

Mr. A’s age and severe cognitive impair­ment raise the possibility of dementia. Rapid onset, history of similar episodes, and apparent inter-episode recovery make dementia unlikely. The history of alcohol abuse and mildly elevated hepatic func­tion tests suggest a substance use disorder such as Wernicke-Korsakoff syndrome or a withdrawal syndrome. However, there is no evidence of excessive alcohol use over the past several months, toxicology studies were negative, and vital signs were stable. General medical causes for Mr. A’s presen­tation, such as hypoglycemia, head trauma, intracranial infection, and metabolic dis­turbance were considered, but physical examination and laboratory studies did not suggest any condition that would explain his condition.

Mr. A’s previous psychiatric hospitaliza­tion is critical in clarifying the more likely diagnosis. A similar presentation yielded the diagnosis of bipolar disorder, manic phase. Our working diagnosis, therefore, was bipolar disorder with features of delir­ious mania.


Delirious mania
Delirious mania was first described by Luther Bell in 1849 and is characterized by an acute and simultaneous onset of mania— severe insomnia, poor judgment, grandios­ity, excitement, emotional lability, bizarre hallucinations, and delusions—and delir­ium—altered consciousness, disorientation, and confusion.2,3 Although there are no diag­nostic criteria, some authors suggest that delirious mania is characterized by inappro­priate toileting, denudation, profound lack of sleep, and episodic memory impairment that can last hours or days.4 Catatonia fre­quently is seen with delirious mania.5 Initial case descriptions described a high mortality rate, approaching 75% of patients.6 There is little published literature and no classifica­tion of delirious mania in DSM-5.1 Estimates are that delirium is concomitant in 20% to 33% of patients with mania.7,8

Several theories try to clarify the underly­ing etiology of delirious mania. Jacobowski et al9 summarized the etiology and pro­posed that it is:
   • 1 of 3 types of mania, including: acute and delusional manias, as initially pro­posed by Kraeplin
   • a severe form of catatonia
   • a condition akin to, but distinct from, delirium with similar underlying medi­cal causes
   • a primary psychiatric disorder under­lying the cause of delirium.

EVALUATION Brain changes
For several days, Mr. A continues to engage in strange behavior. He tries to take patients’ belongings, is denudative, crawls on floors, licks walls, is unable to feed himself, and exhib­its odd motor movements with purposeless motor activity.

We consult our internal medicine team to iden­tify treatable, medical causes. Results of serum B12, thyroid-stimulating hormone, and rapid plasma reagin studies are within normal limits. Urinalysis is negative. A brain MRI reveals numerous white-matter T2-weighted and FLAIR hyperintensities, indicating small-vessel ischemic changes that are consistent with the findings of an MRI 3 years ago. A sleep-deprived EEG with temporal leads obtained on Day 4 of hospitalization demonstrates a diffusely slow and marginally to poorly organized background, believed to indicate global cerebral dysfunction that is most consistent with nonfocal global encephalopathy. There is no seizure activity. We do not perform a lumbar puncture because of Mr. A’s absence of focal neurologic deficits, lack of fever, and normal white blood cell count.


What is the most appropriate treatment?

   a) electroconvulsive therapy (ECT)
   b) high-dose benzodiazepine
   c) mood stabilizer
   d) antipsychotic


The authors’ observations
We strongly suspect that Mr. A has delirious mania. Symptoms and signs of mania include labile mood, excessive spending, grandios­ity, insomnia, and psychosis together with delirium (marked disorientation, confusion). We ascribed Mr. A’s odd motor behaviors to catatonia, a hallmark of delirious mania. The literature has little description of EEG find­ings in suspected cases of delirious mania; however, abnormal EEG tracings have been reported.10 We also speculated that Mr. A’s EEG reflected effects produced by his pre­scribed antipsychotic regimen.

Treatment
There is no clear consensus on treating deliri­ous mania. Because catatonia is a key feature of delirious mania—whether etiologically or as a prominent sign of the condition—ECT and benzodiazepines are proposed as pri­mary treatments. In a study of 16 patients with delirious mania, Karmacharya et al4 found ECT to be effective, with patients showing improvement after 1 to 4 treat­ments. Lee et al10 reported similar findings. Although a high-dose benzodiazepine is not as effective as ECT, a 1-time oral dose of 3 to 4 mg of lorazepam has been used to treat delirious mania.

The efficacy of antipsychotic and mood-stabilizing pharmacotherapy is not clear. Bond3 described 3 cases in which patients were treated effectively with a typical anti­psychotic (haloperidol or chlorpromazine) and lithium. Jung and Lee11demonstrated the efficacy of atypical antipsychotics, with a marked improvement in symptoms within 1 week. However, other studies do not sup­port these findings. Karmacharya et al4 found that typical antipsychotics 1) make the clinical picture worse by increasing extrapy­ramidal symptoms and 2) produce incon­sistent effects. Mood stabilizers sometimes proved beneficial.

Karmacharya et al4 further argued that the delay in improvement seen with any antipsychotics and mood stabilizers suggest they should not be considered a first-line treatment. These discordant findings are the result of a small number of studies and a lack of understanding of the exact nature of delirious mania.


TREATMENT
Quick Response
Mr. A’s symptoms rapidly resolve with a com­bination of quetiapine, 800 mg/d, haloperidol, 10 mg/d, and lithium, 1,200 mg/d. His mood returns to euthymia and his psychotic symptoms abate. He is able to attend to all activities of daily living. Mental status clears and he is fully oriented and able to hold a logical conversation. He scores 28 out of 30 on a subsequent Montreal Cognitive Assessment, administered 11 days after the ini­tial assessment (Figure 2), indicating normal neurocognitive function. He returns to his baseline level of functioning and is discharged in psychiatrically stable condition. Mr. A has no recollection of the bizarre behaviors he dis­played earlier in his hospitalization.


 

 


The authors’ observations
We started Mr. A on antipsychotics because of his initial level of agitation. In reviewing pharmacotherapy options for Mr. A’s mania and delirium, we contemplated several options. Quetiapine and lithium were cho­sen after a review of outside hospital records demonstrated a combination of a mood sta­bilizer and an antipsychotic was effective in treating a previous similar episode, which led to remission of Mr. A’s symptoms. We chose quetiapine because of it highly sedat­ing properties, suspecting that it would help treat his insomnia. We thought that the risk that lithium would make delirium worse was mitigated by Mr. A’s previous therapeu­tic response to it. Haloperidol was added for treating delirium, given its more potent D2 antagonism. Mr. A responded quickly to these interventions.

We did not consider ECT at the begin­ning of Mr. A’s admission, and we avoided sedative-hypnotic agents because we were concerned that a benzodiazepine might make his delirium worse. In light of avail­able data suggesting that ECT and ben­zodiazepines are preferred treatments for delirious mania, it is noteworthy that Mr. A responded so robustly and rapidly to an antipsychotic and a mood stabilizer.

Bottom Line
Consider delirious mania in any patient who has a history of bipolar disorder presenting with co-occuring symptoms of mania and delirium. Collateral information is vital to establishing a diagnosis. With suspected delirium, rule out concomitant reversible medical problems. Electroconvulsive therapy, high-dose benzodiazepines, antipsychotics, and mood stabilizers have shown efficacy.


Related Resources
• Nunes AL, Cheniaux E. Delirium and mania with catatonic fea­tures in a Brazilian patient: response to ECT. J Neuropsychiatry Clin Neurosci. 2014;26(1):E1-E3.
• Danivas V, Behere RV, Varambally S, et al. Electroconvulsive ther­apy in the treatment of delirious mania: a report of 2 patients. J ECT. 2010;26(4):278-279.

Drug Brand Names
Chlorpromazine • Thorazine               Perphenazine • Trilafon
Haloperidol • Haldol                           Quetiapine • Seroquel
Lithium • Eskalith                              Risperidone • Risperdal
Lorazepam • Ativan                           Valproic acid • Depakene

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

Article PDF
Author and Disclosure Information

Matthew J. Davis, MD
Assistant Professor of Psychiatry
Dartmouth’s Geisel School of Medicine
Staff Psychiatrist
New Hampshire
Hospital, Concord, New Hampshire

Alexander de Nesnera, MD
Associate Professor of Psychiatry
Dartmouth’s Geisel School of Medicine
Associate Medical Director
New Hampshire Hospital
Concord, New Hampshire


David G. Folks, MD
Professor of Psychiatry
Dartmouth’s Geisel School of Medicine
Chief Medical Officer
New Hampshire Hospital, Concord, New Hampshire

Issue
Current Psychiatry - 13(7)
Publications
Topics
Page Number
56-62
Legacy Keywords
delusions, hallucinations, neurocognitive testing, delirious mania, bipolar disorder, manic, delirium
Sections
Author and Disclosure Information

Matthew J. Davis, MD
Assistant Professor of Psychiatry
Dartmouth’s Geisel School of Medicine
Staff Psychiatrist
New Hampshire
Hospital, Concord, New Hampshire

Alexander de Nesnera, MD
Associate Professor of Psychiatry
Dartmouth’s Geisel School of Medicine
Associate Medical Director
New Hampshire Hospital
Concord, New Hampshire


David G. Folks, MD
Professor of Psychiatry
Dartmouth’s Geisel School of Medicine
Chief Medical Officer
New Hampshire Hospital, Concord, New Hampshire

Author and Disclosure Information

Matthew J. Davis, MD
Assistant Professor of Psychiatry
Dartmouth’s Geisel School of Medicine
Staff Psychiatrist
New Hampshire
Hospital, Concord, New Hampshire

Alexander de Nesnera, MD
Associate Professor of Psychiatry
Dartmouth’s Geisel School of Medicine
Associate Medical Director
New Hampshire Hospital
Concord, New Hampshire


David G. Folks, MD
Professor of Psychiatry
Dartmouth’s Geisel School of Medicine
Chief Medical Officer
New Hampshire Hospital, Concord, New Hampshire

Article PDF
Article PDF
Related Articles

CASE Nearly naked
Mr. A, age 68, is found sitting in his car, wear­ing only a jacket, underpants, and boots. He speaks of spreading a message about Osama bin Laden and “taking a census.” Police officers bring him to a hospital emergency depart­ment for evaluation.

The examining clinician determines that Mr. A is a danger to himself and others because of mental illness, leading to admission to our state psychiatric hospital.

Mr. A’s wife describes recent spend­ing sprees with large purchases. She had obtained a restraining order against her hus­band because of his threatening remarks and behaviors. Within days of the order issuance, he got a home equity loan and purchased a $300,000 house.

The medical history is notable for type 2 diabetes mellitus. Although he is not tak­ing medications, his blood sugar is well controlled. Other than an initial resting heart rate of 116 beats per minute, vital signs are stable and within normal lim­its. Physical examination is unremarkable. Screening laboratory studies are notable for mildly elevated hepatic function, which approaches normal range several days after admission.

Mr. A reports a remote history of alcohol abuse but says he had not been drinking recently, and does not detail his pattern of use. Urine toxicology screen is negative for all substances of abuse.

Mental status examination reveals dishev­eled appearance, motor agitation, pressured speech, labile affect, loosening of associa­tions, grandiose delusions, and auditory hal­lucinations. Mr. A’s thought processes are grossly disorganized, such that we could not gather a meaningful history. He believes God is speaking directly to him about plans to build a carousel at Disney World. He makes strange gestures with his hands throughout the inter­view, as if attempting to trace the shapes of letters and numbers. He frequently speaks of seeing an array of colors. Cognitive examina­tion reveals a score of 5 of 30 on the Montreal Cognitive Assessment (Figure 1), indicating a severe impairment in neurocognitive func­tioning. He demonstrates limited insight and markedly impaired judgment, and denies hav­ing a mental illness.


What should be the next step in managing Mr. A?
a) obtain records from other facilities and collateral history
b) start an antipsychotic
c) order a brain MRI
d) start an alcohol withdrawal protocol


The authors’ observations
Mr. A showed elements of mania, psycho­sis, and delirium. We considered a broad differential diagnosis (Table). Mr. A initially could not provide reliable or accurate information. The least invasive next step was to obtain additional history from his wife and other medical records to refine the differential diagnosis.



HISTORY Bizarre behavior
Mr. A allows staff to speak with his wife and obtain records from a psychiatric hospitaliza­tion 3 years earlier. Mrs. A reports significant and rapid changes in her husband’s behav­ior and personality over 3 months, but does not describe a recent alcohol relapse. Mr. A sleeps very little, remaining awake and active throughout the night. He frequently rear­ranges the furniture in their home for no clear reason. Once, he knocked on the door of a young female neighbor asking if she found him attractive.

Mr. A has a significant criminal history. Approximately 30 years ago, he was charged with attempted murder of his ex-wife and he had faced charges of attempted kidnapping and assaulting a police officer. However, he has no recent legal issues.

Mr. A has a history of episodes that are simi­lar to this presentation. Seven years ago, he impulsively purchased a $650,000 house after his fourth wife died. He then had a $90,000 heart-shaped pool installed. He also drove a tractor through his stepdaughter’s car for no apparent reason. Also, 3 years ago, he displayed symptoms similar to his current presentation, including insomnia, irritability, and grandios­ity. He engaged in strange behaviors, such as dressing up and imitating homeless people at his church.

During the hospitalization 3 years ago, cli­nicians gave Mr. A a diagnosis of bipolar dis­order, current episode manic, and delirium of an unclear cause. A medical workup, includ­ing brain MRI, did not uncover a basis for his delirium. Antipsychotics (risperidone and per­phenazine) and mood stabilizers (lithium and valproic acid), stabilized his condition; after 7 weeks, Mr. A was discharged, but he did not pursue outpatient psychiatric care.


What is the most likely DSM-5 diagnosis?
   a) major neurocognitive disorder (dementia)
   b) alcohol use disorder (eg, Wernicke- Korsakoff syndrome)
   c) delirium secondary to mania
   d) psychotic disorder


The authors’ observations

DSM-51 suggests a stepwise approach to diagnosis, with consideration of:
   • signs and symptoms
   • substance use
   • general medical condition
   • developmental conflict or stage
   • whether a mental disorder is present.

 

 

Mr. A’s age and severe cognitive impair­ment raise the possibility of dementia. Rapid onset, history of similar episodes, and apparent inter-episode recovery make dementia unlikely. The history of alcohol abuse and mildly elevated hepatic func­tion tests suggest a substance use disorder such as Wernicke-Korsakoff syndrome or a withdrawal syndrome. However, there is no evidence of excessive alcohol use over the past several months, toxicology studies were negative, and vital signs were stable. General medical causes for Mr. A’s presen­tation, such as hypoglycemia, head trauma, intracranial infection, and metabolic dis­turbance were considered, but physical examination and laboratory studies did not suggest any condition that would explain his condition.

Mr. A’s previous psychiatric hospitaliza­tion is critical in clarifying the more likely diagnosis. A similar presentation yielded the diagnosis of bipolar disorder, manic phase. Our working diagnosis, therefore, was bipolar disorder with features of delir­ious mania.


Delirious mania
Delirious mania was first described by Luther Bell in 1849 and is characterized by an acute and simultaneous onset of mania— severe insomnia, poor judgment, grandios­ity, excitement, emotional lability, bizarre hallucinations, and delusions—and delir­ium—altered consciousness, disorientation, and confusion.2,3 Although there are no diag­nostic criteria, some authors suggest that delirious mania is characterized by inappro­priate toileting, denudation, profound lack of sleep, and episodic memory impairment that can last hours or days.4 Catatonia fre­quently is seen with delirious mania.5 Initial case descriptions described a high mortality rate, approaching 75% of patients.6 There is little published literature and no classifica­tion of delirious mania in DSM-5.1 Estimates are that delirium is concomitant in 20% to 33% of patients with mania.7,8

Several theories try to clarify the underly­ing etiology of delirious mania. Jacobowski et al9 summarized the etiology and pro­posed that it is:
   • 1 of 3 types of mania, including: acute and delusional manias, as initially pro­posed by Kraeplin
   • a severe form of catatonia
   • a condition akin to, but distinct from, delirium with similar underlying medi­cal causes
   • a primary psychiatric disorder under­lying the cause of delirium.

EVALUATION Brain changes
For several days, Mr. A continues to engage in strange behavior. He tries to take patients’ belongings, is denudative, crawls on floors, licks walls, is unable to feed himself, and exhib­its odd motor movements with purposeless motor activity.

We consult our internal medicine team to iden­tify treatable, medical causes. Results of serum B12, thyroid-stimulating hormone, and rapid plasma reagin studies are within normal limits. Urinalysis is negative. A brain MRI reveals numerous white-matter T2-weighted and FLAIR hyperintensities, indicating small-vessel ischemic changes that are consistent with the findings of an MRI 3 years ago. A sleep-deprived EEG with temporal leads obtained on Day 4 of hospitalization demonstrates a diffusely slow and marginally to poorly organized background, believed to indicate global cerebral dysfunction that is most consistent with nonfocal global encephalopathy. There is no seizure activity. We do not perform a lumbar puncture because of Mr. A’s absence of focal neurologic deficits, lack of fever, and normal white blood cell count.


What is the most appropriate treatment?

   a) electroconvulsive therapy (ECT)
   b) high-dose benzodiazepine
   c) mood stabilizer
   d) antipsychotic


The authors’ observations
We strongly suspect that Mr. A has delirious mania. Symptoms and signs of mania include labile mood, excessive spending, grandios­ity, insomnia, and psychosis together with delirium (marked disorientation, confusion). We ascribed Mr. A’s odd motor behaviors to catatonia, a hallmark of delirious mania. The literature has little description of EEG find­ings in suspected cases of delirious mania; however, abnormal EEG tracings have been reported.10 We also speculated that Mr. A’s EEG reflected effects produced by his pre­scribed antipsychotic regimen.

Treatment
There is no clear consensus on treating deliri­ous mania. Because catatonia is a key feature of delirious mania—whether etiologically or as a prominent sign of the condition—ECT and benzodiazepines are proposed as pri­mary treatments. In a study of 16 patients with delirious mania, Karmacharya et al4 found ECT to be effective, with patients showing improvement after 1 to 4 treat­ments. Lee et al10 reported similar findings. Although a high-dose benzodiazepine is not as effective as ECT, a 1-time oral dose of 3 to 4 mg of lorazepam has been used to treat delirious mania.

The efficacy of antipsychotic and mood-stabilizing pharmacotherapy is not clear. Bond3 described 3 cases in which patients were treated effectively with a typical anti­psychotic (haloperidol or chlorpromazine) and lithium. Jung and Lee11demonstrated the efficacy of atypical antipsychotics, with a marked improvement in symptoms within 1 week. However, other studies do not sup­port these findings. Karmacharya et al4 found that typical antipsychotics 1) make the clinical picture worse by increasing extrapy­ramidal symptoms and 2) produce incon­sistent effects. Mood stabilizers sometimes proved beneficial.

Karmacharya et al4 further argued that the delay in improvement seen with any antipsychotics and mood stabilizers suggest they should not be considered a first-line treatment. These discordant findings are the result of a small number of studies and a lack of understanding of the exact nature of delirious mania.


TREATMENT
Quick Response
Mr. A’s symptoms rapidly resolve with a com­bination of quetiapine, 800 mg/d, haloperidol, 10 mg/d, and lithium, 1,200 mg/d. His mood returns to euthymia and his psychotic symptoms abate. He is able to attend to all activities of daily living. Mental status clears and he is fully oriented and able to hold a logical conversation. He scores 28 out of 30 on a subsequent Montreal Cognitive Assessment, administered 11 days after the ini­tial assessment (Figure 2), indicating normal neurocognitive function. He returns to his baseline level of functioning and is discharged in psychiatrically stable condition. Mr. A has no recollection of the bizarre behaviors he dis­played earlier in his hospitalization.


 

 


The authors’ observations
We started Mr. A on antipsychotics because of his initial level of agitation. In reviewing pharmacotherapy options for Mr. A’s mania and delirium, we contemplated several options. Quetiapine and lithium were cho­sen after a review of outside hospital records demonstrated a combination of a mood sta­bilizer and an antipsychotic was effective in treating a previous similar episode, which led to remission of Mr. A’s symptoms. We chose quetiapine because of it highly sedat­ing properties, suspecting that it would help treat his insomnia. We thought that the risk that lithium would make delirium worse was mitigated by Mr. A’s previous therapeu­tic response to it. Haloperidol was added for treating delirium, given its more potent D2 antagonism. Mr. A responded quickly to these interventions.

We did not consider ECT at the begin­ning of Mr. A’s admission, and we avoided sedative-hypnotic agents because we were concerned that a benzodiazepine might make his delirium worse. In light of avail­able data suggesting that ECT and ben­zodiazepines are preferred treatments for delirious mania, it is noteworthy that Mr. A responded so robustly and rapidly to an antipsychotic and a mood stabilizer.

Bottom Line
Consider delirious mania in any patient who has a history of bipolar disorder presenting with co-occuring symptoms of mania and delirium. Collateral information is vital to establishing a diagnosis. With suspected delirium, rule out concomitant reversible medical problems. Electroconvulsive therapy, high-dose benzodiazepines, antipsychotics, and mood stabilizers have shown efficacy.


Related Resources
• Nunes AL, Cheniaux E. Delirium and mania with catatonic fea­tures in a Brazilian patient: response to ECT. J Neuropsychiatry Clin Neurosci. 2014;26(1):E1-E3.
• Danivas V, Behere RV, Varambally S, et al. Electroconvulsive ther­apy in the treatment of delirious mania: a report of 2 patients. J ECT. 2010;26(4):278-279.

Drug Brand Names
Chlorpromazine • Thorazine               Perphenazine • Trilafon
Haloperidol • Haldol                           Quetiapine • Seroquel
Lithium • Eskalith                              Risperidone • Risperdal
Lorazepam • Ativan                           Valproic acid • Depakene

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

CASE Nearly naked
Mr. A, age 68, is found sitting in his car, wear­ing only a jacket, underpants, and boots. He speaks of spreading a message about Osama bin Laden and “taking a census.” Police officers bring him to a hospital emergency depart­ment for evaluation.

The examining clinician determines that Mr. A is a danger to himself and others because of mental illness, leading to admission to our state psychiatric hospital.

Mr. A’s wife describes recent spend­ing sprees with large purchases. She had obtained a restraining order against her hus­band because of his threatening remarks and behaviors. Within days of the order issuance, he got a home equity loan and purchased a $300,000 house.

The medical history is notable for type 2 diabetes mellitus. Although he is not tak­ing medications, his blood sugar is well controlled. Other than an initial resting heart rate of 116 beats per minute, vital signs are stable and within normal lim­its. Physical examination is unremarkable. Screening laboratory studies are notable for mildly elevated hepatic function, which approaches normal range several days after admission.

Mr. A reports a remote history of alcohol abuse but says he had not been drinking recently, and does not detail his pattern of use. Urine toxicology screen is negative for all substances of abuse.

Mental status examination reveals dishev­eled appearance, motor agitation, pressured speech, labile affect, loosening of associa­tions, grandiose delusions, and auditory hal­lucinations. Mr. A’s thought processes are grossly disorganized, such that we could not gather a meaningful history. He believes God is speaking directly to him about plans to build a carousel at Disney World. He makes strange gestures with his hands throughout the inter­view, as if attempting to trace the shapes of letters and numbers. He frequently speaks of seeing an array of colors. Cognitive examina­tion reveals a score of 5 of 30 on the Montreal Cognitive Assessment (Figure 1), indicating a severe impairment in neurocognitive func­tioning. He demonstrates limited insight and markedly impaired judgment, and denies hav­ing a mental illness.


What should be the next step in managing Mr. A?
a) obtain records from other facilities and collateral history
b) start an antipsychotic
c) order a brain MRI
d) start an alcohol withdrawal protocol


The authors’ observations
Mr. A showed elements of mania, psycho­sis, and delirium. We considered a broad differential diagnosis (Table). Mr. A initially could not provide reliable or accurate information. The least invasive next step was to obtain additional history from his wife and other medical records to refine the differential diagnosis.



HISTORY Bizarre behavior
Mr. A allows staff to speak with his wife and obtain records from a psychiatric hospitaliza­tion 3 years earlier. Mrs. A reports significant and rapid changes in her husband’s behav­ior and personality over 3 months, but does not describe a recent alcohol relapse. Mr. A sleeps very little, remaining awake and active throughout the night. He frequently rear­ranges the furniture in their home for no clear reason. Once, he knocked on the door of a young female neighbor asking if she found him attractive.

Mr. A has a significant criminal history. Approximately 30 years ago, he was charged with attempted murder of his ex-wife and he had faced charges of attempted kidnapping and assaulting a police officer. However, he has no recent legal issues.

Mr. A has a history of episodes that are simi­lar to this presentation. Seven years ago, he impulsively purchased a $650,000 house after his fourth wife died. He then had a $90,000 heart-shaped pool installed. He also drove a tractor through his stepdaughter’s car for no apparent reason. Also, 3 years ago, he displayed symptoms similar to his current presentation, including insomnia, irritability, and grandios­ity. He engaged in strange behaviors, such as dressing up and imitating homeless people at his church.

During the hospitalization 3 years ago, cli­nicians gave Mr. A a diagnosis of bipolar dis­order, current episode manic, and delirium of an unclear cause. A medical workup, includ­ing brain MRI, did not uncover a basis for his delirium. Antipsychotics (risperidone and per­phenazine) and mood stabilizers (lithium and valproic acid), stabilized his condition; after 7 weeks, Mr. A was discharged, but he did not pursue outpatient psychiatric care.


What is the most likely DSM-5 diagnosis?
   a) major neurocognitive disorder (dementia)
   b) alcohol use disorder (eg, Wernicke- Korsakoff syndrome)
   c) delirium secondary to mania
   d) psychotic disorder


The authors’ observations

DSM-51 suggests a stepwise approach to diagnosis, with consideration of:
   • signs and symptoms
   • substance use
   • general medical condition
   • developmental conflict or stage
   • whether a mental disorder is present.

 

 

Mr. A’s age and severe cognitive impair­ment raise the possibility of dementia. Rapid onset, history of similar episodes, and apparent inter-episode recovery make dementia unlikely. The history of alcohol abuse and mildly elevated hepatic func­tion tests suggest a substance use disorder such as Wernicke-Korsakoff syndrome or a withdrawal syndrome. However, there is no evidence of excessive alcohol use over the past several months, toxicology studies were negative, and vital signs were stable. General medical causes for Mr. A’s presen­tation, such as hypoglycemia, head trauma, intracranial infection, and metabolic dis­turbance were considered, but physical examination and laboratory studies did not suggest any condition that would explain his condition.

Mr. A’s previous psychiatric hospitaliza­tion is critical in clarifying the more likely diagnosis. A similar presentation yielded the diagnosis of bipolar disorder, manic phase. Our working diagnosis, therefore, was bipolar disorder with features of delir­ious mania.


Delirious mania
Delirious mania was first described by Luther Bell in 1849 and is characterized by an acute and simultaneous onset of mania— severe insomnia, poor judgment, grandios­ity, excitement, emotional lability, bizarre hallucinations, and delusions—and delir­ium—altered consciousness, disorientation, and confusion.2,3 Although there are no diag­nostic criteria, some authors suggest that delirious mania is characterized by inappro­priate toileting, denudation, profound lack of sleep, and episodic memory impairment that can last hours or days.4 Catatonia fre­quently is seen with delirious mania.5 Initial case descriptions described a high mortality rate, approaching 75% of patients.6 There is little published literature and no classifica­tion of delirious mania in DSM-5.1 Estimates are that delirium is concomitant in 20% to 33% of patients with mania.7,8

Several theories try to clarify the underly­ing etiology of delirious mania. Jacobowski et al9 summarized the etiology and pro­posed that it is:
   • 1 of 3 types of mania, including: acute and delusional manias, as initially pro­posed by Kraeplin
   • a severe form of catatonia
   • a condition akin to, but distinct from, delirium with similar underlying medi­cal causes
   • a primary psychiatric disorder under­lying the cause of delirium.

EVALUATION Brain changes
For several days, Mr. A continues to engage in strange behavior. He tries to take patients’ belongings, is denudative, crawls on floors, licks walls, is unable to feed himself, and exhib­its odd motor movements with purposeless motor activity.

We consult our internal medicine team to iden­tify treatable, medical causes. Results of serum B12, thyroid-stimulating hormone, and rapid plasma reagin studies are within normal limits. Urinalysis is negative. A brain MRI reveals numerous white-matter T2-weighted and FLAIR hyperintensities, indicating small-vessel ischemic changes that are consistent with the findings of an MRI 3 years ago. A sleep-deprived EEG with temporal leads obtained on Day 4 of hospitalization demonstrates a diffusely slow and marginally to poorly organized background, believed to indicate global cerebral dysfunction that is most consistent with nonfocal global encephalopathy. There is no seizure activity. We do not perform a lumbar puncture because of Mr. A’s absence of focal neurologic deficits, lack of fever, and normal white blood cell count.


What is the most appropriate treatment?

   a) electroconvulsive therapy (ECT)
   b) high-dose benzodiazepine
   c) mood stabilizer
   d) antipsychotic


The authors’ observations
We strongly suspect that Mr. A has delirious mania. Symptoms and signs of mania include labile mood, excessive spending, grandios­ity, insomnia, and psychosis together with delirium (marked disorientation, confusion). We ascribed Mr. A’s odd motor behaviors to catatonia, a hallmark of delirious mania. The literature has little description of EEG find­ings in suspected cases of delirious mania; however, abnormal EEG tracings have been reported.10 We also speculated that Mr. A’s EEG reflected effects produced by his pre­scribed antipsychotic regimen.

Treatment
There is no clear consensus on treating deliri­ous mania. Because catatonia is a key feature of delirious mania—whether etiologically or as a prominent sign of the condition—ECT and benzodiazepines are proposed as pri­mary treatments. In a study of 16 patients with delirious mania, Karmacharya et al4 found ECT to be effective, with patients showing improvement after 1 to 4 treat­ments. Lee et al10 reported similar findings. Although a high-dose benzodiazepine is not as effective as ECT, a 1-time oral dose of 3 to 4 mg of lorazepam has been used to treat delirious mania.

The efficacy of antipsychotic and mood-stabilizing pharmacotherapy is not clear. Bond3 described 3 cases in which patients were treated effectively with a typical anti­psychotic (haloperidol or chlorpromazine) and lithium. Jung and Lee11demonstrated the efficacy of atypical antipsychotics, with a marked improvement in symptoms within 1 week. However, other studies do not sup­port these findings. Karmacharya et al4 found that typical antipsychotics 1) make the clinical picture worse by increasing extrapy­ramidal symptoms and 2) produce incon­sistent effects. Mood stabilizers sometimes proved beneficial.

Karmacharya et al4 further argued that the delay in improvement seen with any antipsychotics and mood stabilizers suggest they should not be considered a first-line treatment. These discordant findings are the result of a small number of studies and a lack of understanding of the exact nature of delirious mania.


TREATMENT
Quick Response
Mr. A’s symptoms rapidly resolve with a com­bination of quetiapine, 800 mg/d, haloperidol, 10 mg/d, and lithium, 1,200 mg/d. His mood returns to euthymia and his psychotic symptoms abate. He is able to attend to all activities of daily living. Mental status clears and he is fully oriented and able to hold a logical conversation. He scores 28 out of 30 on a subsequent Montreal Cognitive Assessment, administered 11 days after the ini­tial assessment (Figure 2), indicating normal neurocognitive function. He returns to his baseline level of functioning and is discharged in psychiatrically stable condition. Mr. A has no recollection of the bizarre behaviors he dis­played earlier in his hospitalization.


 

 


The authors’ observations
We started Mr. A on antipsychotics because of his initial level of agitation. In reviewing pharmacotherapy options for Mr. A’s mania and delirium, we contemplated several options. Quetiapine and lithium were cho­sen after a review of outside hospital records demonstrated a combination of a mood sta­bilizer and an antipsychotic was effective in treating a previous similar episode, which led to remission of Mr. A’s symptoms. We chose quetiapine because of it highly sedat­ing properties, suspecting that it would help treat his insomnia. We thought that the risk that lithium would make delirium worse was mitigated by Mr. A’s previous therapeu­tic response to it. Haloperidol was added for treating delirium, given its more potent D2 antagonism. Mr. A responded quickly to these interventions.

We did not consider ECT at the begin­ning of Mr. A’s admission, and we avoided sedative-hypnotic agents because we were concerned that a benzodiazepine might make his delirium worse. In light of avail­able data suggesting that ECT and ben­zodiazepines are preferred treatments for delirious mania, it is noteworthy that Mr. A responded so robustly and rapidly to an antipsychotic and a mood stabilizer.

Bottom Line
Consider delirious mania in any patient who has a history of bipolar disorder presenting with co-occuring symptoms of mania and delirium. Collateral information is vital to establishing a diagnosis. With suspected delirium, rule out concomitant reversible medical problems. Electroconvulsive therapy, high-dose benzodiazepines, antipsychotics, and mood stabilizers have shown efficacy.


Related Resources
• Nunes AL, Cheniaux E. Delirium and mania with catatonic fea­tures in a Brazilian patient: response to ECT. J Neuropsychiatry Clin Neurosci. 2014;26(1):E1-E3.
• Danivas V, Behere RV, Varambally S, et al. Electroconvulsive ther­apy in the treatment of delirious mania: a report of 2 patients. J ECT. 2010;26(4):278-279.

Drug Brand Names
Chlorpromazine • Thorazine               Perphenazine • Trilafon
Haloperidol • Haldol                           Quetiapine • Seroquel
Lithium • Eskalith                              Risperidone • Risperdal
Lorazepam • Ativan                           Valproic acid • Depakene

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

Issue
Current Psychiatry - 13(7)
Issue
Current Psychiatry - 13(7)
Page Number
56-62
Page Number
56-62
Publications
Publications
Topics
Article Type
Display Headline
Confused and nearly naked after going on spending sprees
Display Headline
Confused and nearly naked after going on spending sprees
Legacy Keywords
delusions, hallucinations, neurocognitive testing, delirious mania, bipolar disorder, manic, delirium
Legacy Keywords
delusions, hallucinations, neurocognitive testing, delirious mania, bipolar disorder, manic, delirium
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

How should you use the lab to monitor patients taking a mood stabilizer?

Article Type
Changed
Tue, 12/11/2018 - 14:47
Display Headline
How should you use the lab to monitor patients taking a mood stabilizer?

Ms. W, age 27, presents with a chief concern of “depression.” She describes a history of several hypo­manic episodes as well as the current depres­sive episode, prompting a bipolar II disorder diagnosis. She is naïve to all psychotropics. You plan to initiate a mood-stabilizing agent. What would you include in your initial workup before starting treatment and how would you monitor her as she continues treatment?

Mood stabilizers are employed to treat bipolar spectrum disorders (bipolar I, bipolar II, and cyclothymic disorder) and schizoaffective disorder, bipolar type. Some evidence suggests that mood stabiliz­ers also can be used for treatment-resistant depressive disorders and borderline per­sonality disorder.1 Mood stabilizers include lithium, valproate, carbamazepine, oxcar­bazepine, and lamotrigine.2-5

This review focuses on applications and monitoring of mood stabilizers for bipolar I and II disorders. We also will briefly review atypical antipsychotics because they also are used to treat bipo­lar spectrum disorders (see the September 2013 issue of Current Psychiatry at CurrentPsychiatry.com for a more detailed article on monitoring of antipsychotics).6

There are several well-researched guidelines used to guide clinical prac­tice.2-5 Many guidelines recommend base­line and routine monitoring parameters based on the characteristics of the agent used. However, the International Society for Bipolar Disorders (ISBD) guidelines highlight the importance of monitoring medical comorbidities, which are common among patients with bipolar disorder and can affect pharmacotherapy and clinical outcomes. These recommendations are similar to metabolic monitoring guidelines for antipsychotics.5

Reviews of therapeutic monitoring show that only one-third to one-half of patien

 

taking a mood stabilizer are appropriately monitored. Poor adher­ence to guideline recommendations often is observed because of patients’ lack of insight or medication adherence and because psychiatric care generally is segre­gated from other medical care.7-9

Baseline testing
The ISBD guidelines recommend an initial workup for all patients that includes:
  • waist circumference or body mass index (BMI), or both
  • blood pressure
  • complete blood count (CBC)
  • electrolytes
  • blood urea nitrogen (BUN) and creatinine
  • liver function tests (LFTs)
  • fasting glucose
  • fasting lipid profile.

In addition, medical history, cigarette smoking status, alcohol intake, and family history of cardiovascular disease, cerebro­vascular disease, hypertension, dyslip­idemia, and diabetes mellitus should be documented. Rule out pregnancy in women of childbearing potential.2 The Figure describes monitoring parameters based on selected agent.

Agent-specific monitoring
Lithium. Patients beginning lithium ther­apy should undergo thyroid function testing and, for patients age >40, ECG mon­itoring. Educate patients about potential side effects of lithium, signs and symptoms of lithium toxicity, and the importance of avoiding dehydration. Adding or changing certain medications could elevate the serum lithium level (eg, diuretics, angiotensin-converting enzyme [ACE]-inhibitors, nonsteroidal anti-inflammatory drugs [NSAIDs], COX-2 inhibitors).

Lithium can cause weight gain and adverse effects in several organ systems, including:
  • gastrointestinal (GI) (nausea, vomit­ing, abdominal pain, loss of appetite, diarrhea)
  • renal (nephrogenic diabetes insipidus, tubulointerstitial renal disease)
  • neurologic (tremors, cognitive dulling, raised intracranial pressure)
  • endocrine (thyroid and parathyroid dysfunction)
  • cardiac (benign electrocardiographic changes, conduction abnormalities)
  • dermatologic (acne, psoriasis, hair loss)
  • hematologic (benign leukocytosis).

Lithium has a narrow therapeutic index (0.5 to 1.2 mEq/L), which means that small changes in the serum level can result in therapeutic inefficacy or toxic­ity. Lithium toxicity can cause irreversible organ damage or death. Serum lithium levels, symptomatic response, emergence and evolution of adverse drug reactions (ADRs), and the recognition of patient risk factors for toxicity can help guide dosing. From a safety monitoring viewpoint, lith­ium toxicity, renal and endocrine adverse effects, and potential drug interactions are foremost concerns.

Lithium usually is started at a low, divided dosages to minimize side effects, and titrated according to response. Check lithium levels before and after each dose increase. Serum levels reach steady state 5 days after dosage adjustment, but might need to be checked sooner if a rapid increase is necessary, such as when treat­ing acute mania, or if you suspect toxicity.

If the patient has renal insufficiency, it may take longer for the lithium to reach steady state; therefore, delaying a blood level beyond 5 days may be necessary to gauge a true steady state. Also, anytime a medication that interferes with lithium renal elimination, such as diuretics, ACE inhibitors, NSAIDs, COX-2 inhibitors, is added or the dosage is changed, a new lithium level will need to be obtained to reassess the level in 5 days, assuming adequate renal function. In general, renal function and thyroid function should be evaluated once or twice during the first 6 months of lithium treatment.

Subsequently, renal and thyroid func­tion can be checked every 6 months to 1 year in stable patients or when clinically indicated. Check a patient’s weight after 6 months of therapy, then at least annually.2

 

 

Valproic acid (VPA) and its derivatives. The most important initial monitoring for VPA therapy includes LFTs and CBC. Before initiating VPA treatment, take a medical history, with special attention to hepatic, hematologic, and bleeding abnor­malities. Therapeutic blood monitoring can be conducted once steady state is achieved and as clinically necessary thereafter.

VPA can be administered at an ini­tial starting dosage of 20 to 30 mg/kg/d in inpatients. In outpatients it is given in low, divided doses or as once-daily dosing using an extended-release formulation to minimize GI and neurologic toxicity and titrated every few days. Target serum level is 50 to 125 μg/mL.

Side effects of VPA include GI distress (eg, anorexia, nausea, dyspepsia, vomiting, diarrhea), hematologic effects (reversible leukopenia, thrombocytopenia), hair loss, weight gain, tremor, hepatic effects (benign LFT elevations, hepatotoxicity), osteoporo­sis, and sedation. Patients with prior or cur­rent hepatic disease may be at greater risk for hepatotoxicity. There is an association between VPA and polycystic ovarian syn­drome. Rare, idiosyncratic, but potentially fatal adverse events with valproate include irreversible hepatic failure, hemorrhagic pancreatitis, and agranulocytosis.

Older monitoring standards indicated taking LFTs and CBC every 6 months and serum VPA level as clinically indicated. According to ISBD guidelines, weight, CBC, LFTs, and menstrual history should be monitored every 3 months for the first year and then annually; blood pressure, bone status (densitometry), fasting glu­cose, and fasting lipids should be moni­tored only in patients with related risk factors. Routine ammonia levels are not recommended but might be indicated if a patient has sudden mental status changes or change in condition.2

Carbamazepine and oxcarbazepine. The most important initial monitoring for car­bamazepine therapy includes LFTs, renal function, electrolytes, and CBC. Before treatment, take a medical history, with special emphasis on history of blood dys­crasias or liver disease. After initiating car­bamazepine, CBC, LFTs, electrolytes, and renal function should be done monthly for 3 months, then repeated annually.

Carbamazepine is a substrate and an inducer of the cytochrome P450 (CYP) system, so it can reduce levels of many other drugs including other antiepileptics, warfarin, and oral contraceptives. Serum level of carbamazepine can be measured at trough after 5 days, with a target level of 4 to 12 μg/mL. Two levels should be drawn, 4 weeks apart, to establish thera­peutic dosage secondary to autoinduction of the CYP450 system.2

As many as one-half of patients experi­ence side effects with carbamazepine. The most common side effects include fatigue, nausea, and neurologic symptoms (dip­lopia, blurred vision, and ataxia). Less frequent side effects include skin rashes, leukopenia, liver enzyme elevations, thrombocytopenia, hyponatremia, and hypo-osmolality. Rare, potentially fatal side effects include agranulocytosis, aplas­tic anemia, thrombocytopenia, hepatic failure, and exfoliative dermatitis (eg, Stevens-Johnson syndrome).

Patients of Asian descent who are taking carbamazepine should undergo genetic testing for the HLA-B*1502 enzyme because persons with this allele are at higher risk of developing Stevens-Johnson syndrome. Also, patients should be edu­cated about the signs and symptoms of these rare adverse reactions so that medi­cal treatment is not delayed should these adverse events present.

Lamotrigine does not require further lab­oratory monitoring beyond the initial rec­ommended workup. The most important variables to consider are interactions with other medications (especially other antiep­ileptics, such as VPA and carbamazepine) and observing for rash. Titration takes several weeks to minimize risk of develop­ing a rash.2 Similar to carbamazepine, the patient should be educated on the signs and symptoms of exfoliative dermatitis (eg, Stevens-Johnson syndrome) so that medical treatment is sought out should this reaction occur.

Atypical antipsychotics. Baseline workup includes the general monitoring param­eters described above. Atypical anti­psychotics have a lower incidence of extrapyramidal side effects than typical antipsychotics, but are associated with an increased risk of metabolic complications. Other major ADRs to consider are cardiac effects and hyperprolactinemia; clinicians should therefore inquire about a personal or family history of cardiac problems, including congenital long QT syndrome. Patients should be screened for any medi­cations that can prolong the QTc interval or interact with the metabolism of medica­tions known to cause QTc prolongation.

Measure weight monthly for the first 3 months, then every 3 months to monitor for metabolic side effects during ongoing treatment. Obtain blood pressure and fast­ing glucose every 3 months for the first year, then annually. Repeat a fasting lipid profile 3 months after initiating treatment, then annually. Cardiac effects and prolac­tin levels can be monitored as needed if clinically indicated.2

CASE CONTINUED
You discuss with Ms. W choices of a mood sta­bilizing agent to treat her bipolar II disorder; she agrees to start lithium. Before initiating treatment, you obtain her weight (and calcu­late her BMI), blood pressure, CBC, electrolyte levels, BUN and creatinine levels, liver func­tion tests, fasting glucose, fasting lipid profile, and thyroid panel. You also review her medi­cal history, lifestyle factors (cigarette smok­ing status, alcohol intake), and family history. A urine pregnancy screen is negative. The pharmacist assists in screening for potential drug-drug interactions, including over-the-counter medications that Ms. W occasionally takes as needed. She is counseled on the use of NSAIDS because these drugs can increase the lithium level.

 

 

Ms. W tolerates and responds well to lith­ium. No further dosing recommendations are made, based on clinical response. You measure her weight at 6 months, then annu­ally. Renal function and thyroid function are monitored at 3 and 6 months after lithium is initiated, and then annually. One year after starting lithium, she continues to tolerate the medication and has minimal metabolic side effects.

Related Resources
• McInnis MG. Lithium for bipolar disorder: A re-emerging treatment for mood instability. Current Psychiatry. 2014; 13(6):38-44.
• Stahl SM. Stahl’s illustrated mood stabilizers. New York, NY: Cambridge University Press; 2009.

Drug Brand Names
Carbamazepine • Tegretol      Valproic acid • Depacon, Depakote
Lamotrigine • Lamictal           Warfarin • Coumadin
Lithium • Lithobid, Eskalith
Oxcarbazepine • Trileptal

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

References


1. Maglione M, Ruelaz Maher A, Hu J, et al. Off-label use of atypical antipsychotics: an update. Comparative Effectiveness Review No. 43. Rockville, MD: Agency for Healthcare Research and Quality; 2011. http://www.effectivehealthcare.ahrq.gov/ehc/products/150/778/CER43_Off-LabelAntipsychotics_20110928.pdf. Published September 2011. Accessed June 6, 2014.
2. American Psychiatric Association. Practice guideline for the treatment of patients with bipolar disorder (revision). Am J Psychiatry. 2002;159(suppl 4):1-50.
3. Ng F, Mammen OK, Wilting I, et al; International Society for Bipolar Disorders. The International Society for Bipolar Disorders (ISBD) consensus guidelines for the safety monitoring of bipolar disorder treatments. Bipolar Disord. 2009;11(6):559-595.
4. National Institute for Health and Clinical Excellence. Bipolar disorder (CG38). The management of bipolar disorder in adults, children and adolescents, in primary and secondary care. http://www.nice.org.uk/CG038. Updated February 13, 2014. Accessed June 6, 2014.
5. Yatham LN, Kennedy SH, O’Donovan C, et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) guidelines for the management of patients with bipolar disorder: update 2007. Bipolar Disord. 2006;8(6):721-739.
6. Zeier K, Connell R, Resch W, et al. Recommendations for lab monitoring of atypical antipsychotics. Current Psychiatry. 2013; 12(9):51-54.
7. Krishnan KR. Psychiatric and medical comorbidities of bipolar disorder. Psychosom Med. 2005;67(1):1-8.
8. Kilbourne AM, Post EP, Bauer MS, et al. Therapeutic drug and cardiovascular disease risk monitoring in patients with bipolar disorder. J Affect Disord. 2007;102(1-3):145-151.
9. Marcus SC, Olfson M, Pincus HA, et al. Therapeutic drug monitoring of mood stabilizers in Medicaid patients with bipolar disorder. Am J Psychiatry. 1999;156(7):1014-1018.

Article PDF
Author and Disclosure Information

 

Sarah K. Cullison, DO
PGY-4 Psychiatry Resident
Grandview Medical Center/Kettering Health Network
Dayton, Ohio

William J. Resch, DO, FAPA
Director of Osteopathic Psychiatric Residency Program
Chillicothe VAMC
Chillicothe, Ohio


Christopher J. Thomas, PharmD, BCPS, BCPP, CGP
Director of PGY-1 and PGY-2 Pharmacy Residency Programs
Clinical Associate Professor of Pharmacology at Ohio University Heritage College of Osteopathic Medicine
Chillicothe VAMC
Chillicothe, Ohio

 

Vicki L. Ellingrod, PharmD, FCCP
Series Editor

Issue
Current Psychiatry - 13(7)
Publications
Topics
Page Number
51-55
Legacy Keywords
mood stabilizers, lithium, bipolar disorder, antipsychotic
Sections
Author and Disclosure Information

 

Sarah K. Cullison, DO
PGY-4 Psychiatry Resident
Grandview Medical Center/Kettering Health Network
Dayton, Ohio

William J. Resch, DO, FAPA
Director of Osteopathic Psychiatric Residency Program
Chillicothe VAMC
Chillicothe, Ohio


Christopher J. Thomas, PharmD, BCPS, BCPP, CGP
Director of PGY-1 and PGY-2 Pharmacy Residency Programs
Clinical Associate Professor of Pharmacology at Ohio University Heritage College of Osteopathic Medicine
Chillicothe VAMC
Chillicothe, Ohio

 

Vicki L. Ellingrod, PharmD, FCCP
Series Editor

Author and Disclosure Information

 

Sarah K. Cullison, DO
PGY-4 Psychiatry Resident
Grandview Medical Center/Kettering Health Network
Dayton, Ohio

William J. Resch, DO, FAPA
Director of Osteopathic Psychiatric Residency Program
Chillicothe VAMC
Chillicothe, Ohio


Christopher J. Thomas, PharmD, BCPS, BCPP, CGP
Director of PGY-1 and PGY-2 Pharmacy Residency Programs
Clinical Associate Professor of Pharmacology at Ohio University Heritage College of Osteopathic Medicine
Chillicothe VAMC
Chillicothe, Ohio

 

Vicki L. Ellingrod, PharmD, FCCP
Series Editor

Article PDF
Article PDF
Related Articles

Ms. W, age 27, presents with a chief concern of “depression.” She describes a history of several hypo­manic episodes as well as the current depres­sive episode, prompting a bipolar II disorder diagnosis. She is naïve to all psychotropics. You plan to initiate a mood-stabilizing agent. What would you include in your initial workup before starting treatment and how would you monitor her as she continues treatment?

Mood stabilizers are employed to treat bipolar spectrum disorders (bipolar I, bipolar II, and cyclothymic disorder) and schizoaffective disorder, bipolar type. Some evidence suggests that mood stabiliz­ers also can be used for treatment-resistant depressive disorders and borderline per­sonality disorder.1 Mood stabilizers include lithium, valproate, carbamazepine, oxcar­bazepine, and lamotrigine.2-5

This review focuses on applications and monitoring of mood stabilizers for bipolar I and II disorders. We also will briefly review atypical antipsychotics because they also are used to treat bipo­lar spectrum disorders (see the September 2013 issue of Current Psychiatry at CurrentPsychiatry.com for a more detailed article on monitoring of antipsychotics).6

There are several well-researched guidelines used to guide clinical prac­tice.2-5 Many guidelines recommend base­line and routine monitoring parameters based on the characteristics of the agent used. However, the International Society for Bipolar Disorders (ISBD) guidelines highlight the importance of monitoring medical comorbidities, which are common among patients with bipolar disorder and can affect pharmacotherapy and clinical outcomes. These recommendations are similar to metabolic monitoring guidelines for antipsychotics.5

Reviews of therapeutic monitoring show that only one-third to one-half of patien

 

taking a mood stabilizer are appropriately monitored. Poor adher­ence to guideline recommendations often is observed because of patients’ lack of insight or medication adherence and because psychiatric care generally is segre­gated from other medical care.7-9

Baseline testing
The ISBD guidelines recommend an initial workup for all patients that includes:
  • waist circumference or body mass index (BMI), or both
  • blood pressure
  • complete blood count (CBC)
  • electrolytes
  • blood urea nitrogen (BUN) and creatinine
  • liver function tests (LFTs)
  • fasting glucose
  • fasting lipid profile.

In addition, medical history, cigarette smoking status, alcohol intake, and family history of cardiovascular disease, cerebro­vascular disease, hypertension, dyslip­idemia, and diabetes mellitus should be documented. Rule out pregnancy in women of childbearing potential.2 The Figure describes monitoring parameters based on selected agent.

Agent-specific monitoring
Lithium. Patients beginning lithium ther­apy should undergo thyroid function testing and, for patients age >40, ECG mon­itoring. Educate patients about potential side effects of lithium, signs and symptoms of lithium toxicity, and the importance of avoiding dehydration. Adding or changing certain medications could elevate the serum lithium level (eg, diuretics, angiotensin-converting enzyme [ACE]-inhibitors, nonsteroidal anti-inflammatory drugs [NSAIDs], COX-2 inhibitors).

Lithium can cause weight gain and adverse effects in several organ systems, including:
  • gastrointestinal (GI) (nausea, vomit­ing, abdominal pain, loss of appetite, diarrhea)
  • renal (nephrogenic diabetes insipidus, tubulointerstitial renal disease)
  • neurologic (tremors, cognitive dulling, raised intracranial pressure)
  • endocrine (thyroid and parathyroid dysfunction)
  • cardiac (benign electrocardiographic changes, conduction abnormalities)
  • dermatologic (acne, psoriasis, hair loss)
  • hematologic (benign leukocytosis).

Lithium has a narrow therapeutic index (0.5 to 1.2 mEq/L), which means that small changes in the serum level can result in therapeutic inefficacy or toxic­ity. Lithium toxicity can cause irreversible organ damage or death. Serum lithium levels, symptomatic response, emergence and evolution of adverse drug reactions (ADRs), and the recognition of patient risk factors for toxicity can help guide dosing. From a safety monitoring viewpoint, lith­ium toxicity, renal and endocrine adverse effects, and potential drug interactions are foremost concerns.

Lithium usually is started at a low, divided dosages to minimize side effects, and titrated according to response. Check lithium levels before and after each dose increase. Serum levels reach steady state 5 days after dosage adjustment, but might need to be checked sooner if a rapid increase is necessary, such as when treat­ing acute mania, or if you suspect toxicity.

If the patient has renal insufficiency, it may take longer for the lithium to reach steady state; therefore, delaying a blood level beyond 5 days may be necessary to gauge a true steady state. Also, anytime a medication that interferes with lithium renal elimination, such as diuretics, ACE inhibitors, NSAIDs, COX-2 inhibitors, is added or the dosage is changed, a new lithium level will need to be obtained to reassess the level in 5 days, assuming adequate renal function. In general, renal function and thyroid function should be evaluated once or twice during the first 6 months of lithium treatment.

Subsequently, renal and thyroid func­tion can be checked every 6 months to 1 year in stable patients or when clinically indicated. Check a patient’s weight after 6 months of therapy, then at least annually.2

 

 

Valproic acid (VPA) and its derivatives. The most important initial monitoring for VPA therapy includes LFTs and CBC. Before initiating VPA treatment, take a medical history, with special attention to hepatic, hematologic, and bleeding abnor­malities. Therapeutic blood monitoring can be conducted once steady state is achieved and as clinically necessary thereafter.

VPA can be administered at an ini­tial starting dosage of 20 to 30 mg/kg/d in inpatients. In outpatients it is given in low, divided doses or as once-daily dosing using an extended-release formulation to minimize GI and neurologic toxicity and titrated every few days. Target serum level is 50 to 125 μg/mL.

Side effects of VPA include GI distress (eg, anorexia, nausea, dyspepsia, vomiting, diarrhea), hematologic effects (reversible leukopenia, thrombocytopenia), hair loss, weight gain, tremor, hepatic effects (benign LFT elevations, hepatotoxicity), osteoporo­sis, and sedation. Patients with prior or cur­rent hepatic disease may be at greater risk for hepatotoxicity. There is an association between VPA and polycystic ovarian syn­drome. Rare, idiosyncratic, but potentially fatal adverse events with valproate include irreversible hepatic failure, hemorrhagic pancreatitis, and agranulocytosis.

Older monitoring standards indicated taking LFTs and CBC every 6 months and serum VPA level as clinically indicated. According to ISBD guidelines, weight, CBC, LFTs, and menstrual history should be monitored every 3 months for the first year and then annually; blood pressure, bone status (densitometry), fasting glu­cose, and fasting lipids should be moni­tored only in patients with related risk factors. Routine ammonia levels are not recommended but might be indicated if a patient has sudden mental status changes or change in condition.2

Carbamazepine and oxcarbazepine. The most important initial monitoring for car­bamazepine therapy includes LFTs, renal function, electrolytes, and CBC. Before treatment, take a medical history, with special emphasis on history of blood dys­crasias or liver disease. After initiating car­bamazepine, CBC, LFTs, electrolytes, and renal function should be done monthly for 3 months, then repeated annually.

Carbamazepine is a substrate and an inducer of the cytochrome P450 (CYP) system, so it can reduce levels of many other drugs including other antiepileptics, warfarin, and oral contraceptives. Serum level of carbamazepine can be measured at trough after 5 days, with a target level of 4 to 12 μg/mL. Two levels should be drawn, 4 weeks apart, to establish thera­peutic dosage secondary to autoinduction of the CYP450 system.2

As many as one-half of patients experi­ence side effects with carbamazepine. The most common side effects include fatigue, nausea, and neurologic symptoms (dip­lopia, blurred vision, and ataxia). Less frequent side effects include skin rashes, leukopenia, liver enzyme elevations, thrombocytopenia, hyponatremia, and hypo-osmolality. Rare, potentially fatal side effects include agranulocytosis, aplas­tic anemia, thrombocytopenia, hepatic failure, and exfoliative dermatitis (eg, Stevens-Johnson syndrome).

Patients of Asian descent who are taking carbamazepine should undergo genetic testing for the HLA-B*1502 enzyme because persons with this allele are at higher risk of developing Stevens-Johnson syndrome. Also, patients should be edu­cated about the signs and symptoms of these rare adverse reactions so that medi­cal treatment is not delayed should these adverse events present.

Lamotrigine does not require further lab­oratory monitoring beyond the initial rec­ommended workup. The most important variables to consider are interactions with other medications (especially other antiep­ileptics, such as VPA and carbamazepine) and observing for rash. Titration takes several weeks to minimize risk of develop­ing a rash.2 Similar to carbamazepine, the patient should be educated on the signs and symptoms of exfoliative dermatitis (eg, Stevens-Johnson syndrome) so that medical treatment is sought out should this reaction occur.

Atypical antipsychotics. Baseline workup includes the general monitoring param­eters described above. Atypical anti­psychotics have a lower incidence of extrapyramidal side effects than typical antipsychotics, but are associated with an increased risk of metabolic complications. Other major ADRs to consider are cardiac effects and hyperprolactinemia; clinicians should therefore inquire about a personal or family history of cardiac problems, including congenital long QT syndrome. Patients should be screened for any medi­cations that can prolong the QTc interval or interact with the metabolism of medica­tions known to cause QTc prolongation.

Measure weight monthly for the first 3 months, then every 3 months to monitor for metabolic side effects during ongoing treatment. Obtain blood pressure and fast­ing glucose every 3 months for the first year, then annually. Repeat a fasting lipid profile 3 months after initiating treatment, then annually. Cardiac effects and prolac­tin levels can be monitored as needed if clinically indicated.2

CASE CONTINUED
You discuss with Ms. W choices of a mood sta­bilizing agent to treat her bipolar II disorder; she agrees to start lithium. Before initiating treatment, you obtain her weight (and calcu­late her BMI), blood pressure, CBC, electrolyte levels, BUN and creatinine levels, liver func­tion tests, fasting glucose, fasting lipid profile, and thyroid panel. You also review her medi­cal history, lifestyle factors (cigarette smok­ing status, alcohol intake), and family history. A urine pregnancy screen is negative. The pharmacist assists in screening for potential drug-drug interactions, including over-the-counter medications that Ms. W occasionally takes as needed. She is counseled on the use of NSAIDS because these drugs can increase the lithium level.

 

 

Ms. W tolerates and responds well to lith­ium. No further dosing recommendations are made, based on clinical response. You measure her weight at 6 months, then annu­ally. Renal function and thyroid function are monitored at 3 and 6 months after lithium is initiated, and then annually. One year after starting lithium, she continues to tolerate the medication and has minimal metabolic side effects.

Related Resources
• McInnis MG. Lithium for bipolar disorder: A re-emerging treatment for mood instability. Current Psychiatry. 2014; 13(6):38-44.
• Stahl SM. Stahl’s illustrated mood stabilizers. New York, NY: Cambridge University Press; 2009.

Drug Brand Names
Carbamazepine • Tegretol      Valproic acid • Depacon, Depakote
Lamotrigine • Lamictal           Warfarin • Coumadin
Lithium • Lithobid, Eskalith
Oxcarbazepine • Trileptal

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

Ms. W, age 27, presents with a chief concern of “depression.” She describes a history of several hypo­manic episodes as well as the current depres­sive episode, prompting a bipolar II disorder diagnosis. She is naïve to all psychotropics. You plan to initiate a mood-stabilizing agent. What would you include in your initial workup before starting treatment and how would you monitor her as she continues treatment?

Mood stabilizers are employed to treat bipolar spectrum disorders (bipolar I, bipolar II, and cyclothymic disorder) and schizoaffective disorder, bipolar type. Some evidence suggests that mood stabiliz­ers also can be used for treatment-resistant depressive disorders and borderline per­sonality disorder.1 Mood stabilizers include lithium, valproate, carbamazepine, oxcar­bazepine, and lamotrigine.2-5

This review focuses on applications and monitoring of mood stabilizers for bipolar I and II disorders. We also will briefly review atypical antipsychotics because they also are used to treat bipo­lar spectrum disorders (see the September 2013 issue of Current Psychiatry at CurrentPsychiatry.com for a more detailed article on monitoring of antipsychotics).6

There are several well-researched guidelines used to guide clinical prac­tice.2-5 Many guidelines recommend base­line and routine monitoring parameters based on the characteristics of the agent used. However, the International Society for Bipolar Disorders (ISBD) guidelines highlight the importance of monitoring medical comorbidities, which are common among patients with bipolar disorder and can affect pharmacotherapy and clinical outcomes. These recommendations are similar to metabolic monitoring guidelines for antipsychotics.5

Reviews of therapeutic monitoring show that only one-third to one-half of patien

 

taking a mood stabilizer are appropriately monitored. Poor adher­ence to guideline recommendations often is observed because of patients’ lack of insight or medication adherence and because psychiatric care generally is segre­gated from other medical care.7-9

Baseline testing
The ISBD guidelines recommend an initial workup for all patients that includes:
  • waist circumference or body mass index (BMI), or both
  • blood pressure
  • complete blood count (CBC)
  • electrolytes
  • blood urea nitrogen (BUN) and creatinine
  • liver function tests (LFTs)
  • fasting glucose
  • fasting lipid profile.

In addition, medical history, cigarette smoking status, alcohol intake, and family history of cardiovascular disease, cerebro­vascular disease, hypertension, dyslip­idemia, and diabetes mellitus should be documented. Rule out pregnancy in women of childbearing potential.2 The Figure describes monitoring parameters based on selected agent.

Agent-specific monitoring
Lithium. Patients beginning lithium ther­apy should undergo thyroid function testing and, for patients age >40, ECG mon­itoring. Educate patients about potential side effects of lithium, signs and symptoms of lithium toxicity, and the importance of avoiding dehydration. Adding or changing certain medications could elevate the serum lithium level (eg, diuretics, angiotensin-converting enzyme [ACE]-inhibitors, nonsteroidal anti-inflammatory drugs [NSAIDs], COX-2 inhibitors).

Lithium can cause weight gain and adverse effects in several organ systems, including:
  • gastrointestinal (GI) (nausea, vomit­ing, abdominal pain, loss of appetite, diarrhea)
  • renal (nephrogenic diabetes insipidus, tubulointerstitial renal disease)
  • neurologic (tremors, cognitive dulling, raised intracranial pressure)
  • endocrine (thyroid and parathyroid dysfunction)
  • cardiac (benign electrocardiographic changes, conduction abnormalities)
  • dermatologic (acne, psoriasis, hair loss)
  • hematologic (benign leukocytosis).

Lithium has a narrow therapeutic index (0.5 to 1.2 mEq/L), which means that small changes in the serum level can result in therapeutic inefficacy or toxic­ity. Lithium toxicity can cause irreversible organ damage or death. Serum lithium levels, symptomatic response, emergence and evolution of adverse drug reactions (ADRs), and the recognition of patient risk factors for toxicity can help guide dosing. From a safety monitoring viewpoint, lith­ium toxicity, renal and endocrine adverse effects, and potential drug interactions are foremost concerns.

Lithium usually is started at a low, divided dosages to minimize side effects, and titrated according to response. Check lithium levels before and after each dose increase. Serum levels reach steady state 5 days after dosage adjustment, but might need to be checked sooner if a rapid increase is necessary, such as when treat­ing acute mania, or if you suspect toxicity.

If the patient has renal insufficiency, it may take longer for the lithium to reach steady state; therefore, delaying a blood level beyond 5 days may be necessary to gauge a true steady state. Also, anytime a medication that interferes with lithium renal elimination, such as diuretics, ACE inhibitors, NSAIDs, COX-2 inhibitors, is added or the dosage is changed, a new lithium level will need to be obtained to reassess the level in 5 days, assuming adequate renal function. In general, renal function and thyroid function should be evaluated once or twice during the first 6 months of lithium treatment.

Subsequently, renal and thyroid func­tion can be checked every 6 months to 1 year in stable patients or when clinically indicated. Check a patient’s weight after 6 months of therapy, then at least annually.2

 

 

Valproic acid (VPA) and its derivatives. The most important initial monitoring for VPA therapy includes LFTs and CBC. Before initiating VPA treatment, take a medical history, with special attention to hepatic, hematologic, and bleeding abnor­malities. Therapeutic blood monitoring can be conducted once steady state is achieved and as clinically necessary thereafter.

VPA can be administered at an ini­tial starting dosage of 20 to 30 mg/kg/d in inpatients. In outpatients it is given in low, divided doses or as once-daily dosing using an extended-release formulation to minimize GI and neurologic toxicity and titrated every few days. Target serum level is 50 to 125 μg/mL.

Side effects of VPA include GI distress (eg, anorexia, nausea, dyspepsia, vomiting, diarrhea), hematologic effects (reversible leukopenia, thrombocytopenia), hair loss, weight gain, tremor, hepatic effects (benign LFT elevations, hepatotoxicity), osteoporo­sis, and sedation. Patients with prior or cur­rent hepatic disease may be at greater risk for hepatotoxicity. There is an association between VPA and polycystic ovarian syn­drome. Rare, idiosyncratic, but potentially fatal adverse events with valproate include irreversible hepatic failure, hemorrhagic pancreatitis, and agranulocytosis.

Older monitoring standards indicated taking LFTs and CBC every 6 months and serum VPA level as clinically indicated. According to ISBD guidelines, weight, CBC, LFTs, and menstrual history should be monitored every 3 months for the first year and then annually; blood pressure, bone status (densitometry), fasting glu­cose, and fasting lipids should be moni­tored only in patients with related risk factors. Routine ammonia levels are not recommended but might be indicated if a patient has sudden mental status changes or change in condition.2

Carbamazepine and oxcarbazepine. The most important initial monitoring for car­bamazepine therapy includes LFTs, renal function, electrolytes, and CBC. Before treatment, take a medical history, with special emphasis on history of blood dys­crasias or liver disease. After initiating car­bamazepine, CBC, LFTs, electrolytes, and renal function should be done monthly for 3 months, then repeated annually.

Carbamazepine is a substrate and an inducer of the cytochrome P450 (CYP) system, so it can reduce levels of many other drugs including other antiepileptics, warfarin, and oral contraceptives. Serum level of carbamazepine can be measured at trough after 5 days, with a target level of 4 to 12 μg/mL. Two levels should be drawn, 4 weeks apart, to establish thera­peutic dosage secondary to autoinduction of the CYP450 system.2

As many as one-half of patients experi­ence side effects with carbamazepine. The most common side effects include fatigue, nausea, and neurologic symptoms (dip­lopia, blurred vision, and ataxia). Less frequent side effects include skin rashes, leukopenia, liver enzyme elevations, thrombocytopenia, hyponatremia, and hypo-osmolality. Rare, potentially fatal side effects include agranulocytosis, aplas­tic anemia, thrombocytopenia, hepatic failure, and exfoliative dermatitis (eg, Stevens-Johnson syndrome).

Patients of Asian descent who are taking carbamazepine should undergo genetic testing for the HLA-B*1502 enzyme because persons with this allele are at higher risk of developing Stevens-Johnson syndrome. Also, patients should be edu­cated about the signs and symptoms of these rare adverse reactions so that medi­cal treatment is not delayed should these adverse events present.

Lamotrigine does not require further lab­oratory monitoring beyond the initial rec­ommended workup. The most important variables to consider are interactions with other medications (especially other antiep­ileptics, such as VPA and carbamazepine) and observing for rash. Titration takes several weeks to minimize risk of develop­ing a rash.2 Similar to carbamazepine, the patient should be educated on the signs and symptoms of exfoliative dermatitis (eg, Stevens-Johnson syndrome) so that medical treatment is sought out should this reaction occur.

Atypical antipsychotics. Baseline workup includes the general monitoring param­eters described above. Atypical anti­psychotics have a lower incidence of extrapyramidal side effects than typical antipsychotics, but are associated with an increased risk of metabolic complications. Other major ADRs to consider are cardiac effects and hyperprolactinemia; clinicians should therefore inquire about a personal or family history of cardiac problems, including congenital long QT syndrome. Patients should be screened for any medi­cations that can prolong the QTc interval or interact with the metabolism of medica­tions known to cause QTc prolongation.

Measure weight monthly for the first 3 months, then every 3 months to monitor for metabolic side effects during ongoing treatment. Obtain blood pressure and fast­ing glucose every 3 months for the first year, then annually. Repeat a fasting lipid profile 3 months after initiating treatment, then annually. Cardiac effects and prolac­tin levels can be monitored as needed if clinically indicated.2

CASE CONTINUED
You discuss with Ms. W choices of a mood sta­bilizing agent to treat her bipolar II disorder; she agrees to start lithium. Before initiating treatment, you obtain her weight (and calcu­late her BMI), blood pressure, CBC, electrolyte levels, BUN and creatinine levels, liver func­tion tests, fasting glucose, fasting lipid profile, and thyroid panel. You also review her medi­cal history, lifestyle factors (cigarette smok­ing status, alcohol intake), and family history. A urine pregnancy screen is negative. The pharmacist assists in screening for potential drug-drug interactions, including over-the-counter medications that Ms. W occasionally takes as needed. She is counseled on the use of NSAIDS because these drugs can increase the lithium level.

 

 

Ms. W tolerates and responds well to lith­ium. No further dosing recommendations are made, based on clinical response. You measure her weight at 6 months, then annu­ally. Renal function and thyroid function are monitored at 3 and 6 months after lithium is initiated, and then annually. One year after starting lithium, she continues to tolerate the medication and has minimal metabolic side effects.

Related Resources
• McInnis MG. Lithium for bipolar disorder: A re-emerging treatment for mood instability. Current Psychiatry. 2014; 13(6):38-44.
• Stahl SM. Stahl’s illustrated mood stabilizers. New York, NY: Cambridge University Press; 2009.

Drug Brand Names
Carbamazepine • Tegretol      Valproic acid • Depacon, Depakote
Lamotrigine • Lamictal           Warfarin • Coumadin
Lithium • Lithobid, Eskalith
Oxcarbazepine • Trileptal

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

References


1. Maglione M, Ruelaz Maher A, Hu J, et al. Off-label use of atypical antipsychotics: an update. Comparative Effectiveness Review No. 43. Rockville, MD: Agency for Healthcare Research and Quality; 2011. http://www.effectivehealthcare.ahrq.gov/ehc/products/150/778/CER43_Off-LabelAntipsychotics_20110928.pdf. Published September 2011. Accessed June 6, 2014.
2. American Psychiatric Association. Practice guideline for the treatment of patients with bipolar disorder (revision). Am J Psychiatry. 2002;159(suppl 4):1-50.
3. Ng F, Mammen OK, Wilting I, et al; International Society for Bipolar Disorders. The International Society for Bipolar Disorders (ISBD) consensus guidelines for the safety monitoring of bipolar disorder treatments. Bipolar Disord. 2009;11(6):559-595.
4. National Institute for Health and Clinical Excellence. Bipolar disorder (CG38). The management of bipolar disorder in adults, children and adolescents, in primary and secondary care. http://www.nice.org.uk/CG038. Updated February 13, 2014. Accessed June 6, 2014.
5. Yatham LN, Kennedy SH, O’Donovan C, et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) guidelines for the management of patients with bipolar disorder: update 2007. Bipolar Disord. 2006;8(6):721-739.
6. Zeier K, Connell R, Resch W, et al. Recommendations for lab monitoring of atypical antipsychotics. Current Psychiatry. 2013; 12(9):51-54.
7. Krishnan KR. Psychiatric and medical comorbidities of bipolar disorder. Psychosom Med. 2005;67(1):1-8.
8. Kilbourne AM, Post EP, Bauer MS, et al. Therapeutic drug and cardiovascular disease risk monitoring in patients with bipolar disorder. J Affect Disord. 2007;102(1-3):145-151.
9. Marcus SC, Olfson M, Pincus HA, et al. Therapeutic drug monitoring of mood stabilizers in Medicaid patients with bipolar disorder. Am J Psychiatry. 1999;156(7):1014-1018.

References


1. Maglione M, Ruelaz Maher A, Hu J, et al. Off-label use of atypical antipsychotics: an update. Comparative Effectiveness Review No. 43. Rockville, MD: Agency for Healthcare Research and Quality; 2011. http://www.effectivehealthcare.ahrq.gov/ehc/products/150/778/CER43_Off-LabelAntipsychotics_20110928.pdf. Published September 2011. Accessed June 6, 2014.
2. American Psychiatric Association. Practice guideline for the treatment of patients with bipolar disorder (revision). Am J Psychiatry. 2002;159(suppl 4):1-50.
3. Ng F, Mammen OK, Wilting I, et al; International Society for Bipolar Disorders. The International Society for Bipolar Disorders (ISBD) consensus guidelines for the safety monitoring of bipolar disorder treatments. Bipolar Disord. 2009;11(6):559-595.
4. National Institute for Health and Clinical Excellence. Bipolar disorder (CG38). The management of bipolar disorder in adults, children and adolescents, in primary and secondary care. http://www.nice.org.uk/CG038. Updated February 13, 2014. Accessed June 6, 2014.
5. Yatham LN, Kennedy SH, O’Donovan C, et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) guidelines for the management of patients with bipolar disorder: update 2007. Bipolar Disord. 2006;8(6):721-739.
6. Zeier K, Connell R, Resch W, et al. Recommendations for lab monitoring of atypical antipsychotics. Current Psychiatry. 2013; 12(9):51-54.
7. Krishnan KR. Psychiatric and medical comorbidities of bipolar disorder. Psychosom Med. 2005;67(1):1-8.
8. Kilbourne AM, Post EP, Bauer MS, et al. Therapeutic drug and cardiovascular disease risk monitoring in patients with bipolar disorder. J Affect Disord. 2007;102(1-3):145-151.
9. Marcus SC, Olfson M, Pincus HA, et al. Therapeutic drug monitoring of mood stabilizers in Medicaid patients with bipolar disorder. Am J Psychiatry. 1999;156(7):1014-1018.

Issue
Current Psychiatry - 13(7)
Issue
Current Psychiatry - 13(7)
Page Number
51-55
Page Number
51-55
Publications
Publications
Topics
Article Type
Display Headline
How should you use the lab to monitor patients taking a mood stabilizer?
Display Headline
How should you use the lab to monitor patients taking a mood stabilizer?
Legacy Keywords
mood stabilizers, lithium, bipolar disorder, antipsychotic
Legacy Keywords
mood stabilizers, lithium, bipolar disorder, antipsychotic
Sections
Disallow All Ads
Alternative CME
Article PDF Media

Four questions address stigma

Article Type
Changed
Mon, 04/16/2018 - 13:31
Display Headline
Four questions address stigma

Naomi is a 61-year-old woman who has lived with bipolar disorder and its stigma for 30 years. After a major manic episode and hospitalization, she entered into family treatment at the urging of her three daughters. Previously, her husband had been the primary force in guiding her psychiatric care, and she had been in treatment with a psychiatrist who is his professional colleague.

The patient’s first depressive episode began in the postpartum period, but she did not seek help at that time because she thought that her feelings were normal for a new mother. She did not receive any psychiatric attention until she cycled into mania and called the police for fear her child was being poisoned by neighbors. Her most recent manic episode occurred after she stopped her medications because of concerns about side effects. She was too embarrassed to tell her husband or doctor. She routinely fails to tell her other medical doctors that she is on mood stabilizers, because she does not want them to know she has bipolar disorder.


As Naomi recovers from the most recent manic episode and settles into family treatment, she is struggling with the consequences of her actions to her family. In family therapy in the past, her husband has revealed his belief that he has been protecting the family from Naomi’s mania and protecting Naomi from "embarrassing herself." This is difficult for Naomi to hear as she has always prided herself on being a good mother and protecting her daughters. Naomi’s situation illustrates the difficulty of coping with a diagnosis of bipolar disorder, the consequences of the illness on the family, and the importance of addressing stigma.

How stigma gets in the way

As discussed previously by Dr. Alison M. Heru ("Mental illness stigma is a family affair," Clinical Psychiatry News, April 2014, p. 8), stigma, when internalized or self-directed, can lead to psychological distress, decreased self-esteem and life satisfaction, and increased depression and suicidality (Compr. Psychiatry 2010;51:603-6). Close family members of those with mental disorders are affected by stigma, commonly referred to as "stigma by association" or "courtesy stigma."

Up to 92% of caregivers of people with psychiatric disorders have reported internalized stigma (J. Psychiatr. Ment. Health Nurs. 2012;19:665-71). These family members become distant and avoidant, resulting in a reduced quality of life and an impaired ability to provide critical support for their loved ones. Caregiver anxiety is inversely related to patient anxiety, stigma, and poor alliance (J. Nerv. Ment. Disease 2011;199:18-24).

As a result of these factors, while people with psychiatric disorders have to cope with their own mental illness as well as the public and self-stigma that alienate them from society, they also are at risk of losing their family connections.

In order to confront stigma, the Family Center for Bipolar Disorder in New York City, for example, uses a Family Inclusive Treatment (FIT) model. The FIT model includes an engagement period at the initiation of treatment that is focused on psychoeducation and relapse prevention planning. FIT is unique in that every patient is required to sign a release of information giving permission for full, open communication at all times between the patient’s clinician and a treatment partner of their choice. After the initial engagement period, there are quarterly family visits to supplement regular individual treatment sessions. FIT treatment promotes open communication about symptoms and medications. FIT strives to minimize patient isolation from families; they can talk openly with one another and their clinician.

Dr. Amy Mednick

After seeing many families enter treatment, FIT staff noticed the prominence of stigma.

We have begun to ask about stigma directly. Do people with more stigma do worse in treatment? Do they adhere more poorly to treatment? Do their families tend to become less involved over time? To begin, Dr. Mednick and staff examined demographic data looking for factors that might predispose a person to experience increased stigma.

In terms of diagnosis, people with more internalizing disorders such as depression and anxiety disorders tend to experience more stigma. Distress is experienced internally. As Dr. Bassirnia and her colleagues wrote in a poster presented at the recent American Psychiatric Association meeting, people with externalizing disorders, such as substance abuse and antisocial disorders, are more likely to express their distress outwardly and are less likely to suffer from stigma ("The relationship between personality traits and perceived internalized stigma in bipolar patients and their caregivers," 2014).

Meanwhile, two systematic review studies have reported moderate to high levels of internalized stigma in people with bipolar disorder. In these studies, a higher level of internalized stigma had a negative correlation with self-esteem, social adjustment, and perceived social support, and positive correlation with severity of symptoms, functional impairment, and rehospitalization. In spite of having more severe symptoms; people with higher levels of self-stigma are less likely to seek professional help and adhere to their treatment. Stigma by association and its negative consequences in caregivers of people with mental disorders also have been reported (J. Affect. Disord. 2013;150:181-91).

 

 

A useful and easy to administer scale that helps to identify stigma is the "Perceived Criticism Scale" (J. Abnorm. Psychol. 1989;98:225-35). By asking four questions, the clinician can get a good sense of family dynamics and can monitor the progress and change over time. The questions rate perception on a scale of 1-10, where "X" is the other person involved in treatment, either patient or caregiver. Here are the questions:

1. How critical do you think you are of X?

2. How critical do you think X is of you?

3. When X criticizes you, how upset do you get?

4. When you criticize X, how upset does he/she get?

For families with high scores, follow-up is needed. The Internalized Stigma of Mental Illness (ISMI) scale (Psychiatry Res. 2003;121:31-49) can be used. The ISMI scale makes statements about stigma for which participants rate their agreement on a Likert scale, such as:

• I don’t talk about myself much because I don’t want to burden others with my mental illness.

• Being around people who don’t have a mental illness makes me feel out of place or inadequate.

• People can tell that I have a mental illness by the way I look.

• Mentally ill people tend to be violent.

• I feel out of place in the world because I have a mental illness.

The ISMI scale contains 29 short, simple statements like the ones above and can be completed in less than 10 minutes. The statements are designed to avoid hypothetical situations, stay focused in the present, and address the participant’s own identity and experience.

Using the tools in practice

Naomi entered family treatment with her husband and daughters. Using the ISMI to measure the stigma of mental illness that each family member was experiencing, Naomi was shocked to see that her daughters felt far less stigma about having a mother with mental illness than she had assumed. In turn, her daughters were shocked at how much stigma Naomi was experiencing. Naomi’s husband scored between them. This data paved the way for an open family conversation about how Naomi’s illness had affected their lives, and especially how Naomi’s husband and his perceptions of her illness had affected her treatment course.

Caregivers play a very important role in bipolar disorder. After all, the illness can lead to difficulty functioning and can threaten the family’s stability. Sometimes caregivers can serve as a source of strength and a beacon of stability in the occasional storm. It is hard for the family between the storms, when the same flashing beacon can be a constant reminder to the patient of their illness. Often, well intentioned concerns become constant checking up, making the patient feel stigmatized and expected to fail.

"Good" caregivers will be aware of the stigma and the impact it has on their loved one and on themselves, without becoming a source of stigma.

Dr. Mednick is an attending psychiatrist at the Family Center for Bipolar at Mount Sinai Beth Israel in New York City. Dr. Bassirnia is a second-year psychiatry resident at Mount Sinai Beth Israel. Scan the QR code to read more Families in Psychiatry columns at clinicalpsychiatrynews.com.

Name
DR. MEDNICKDR. BASSIRNIA
Author and Disclosure Information

Publications
Topics
Legacy Keywords
bipolar disorder, stigma, manic episode, hospitalization, treatment, psychiatric care,
Sections
Author and Disclosure Information

Author and Disclosure Information

Name
DR. MEDNICKDR. BASSIRNIA
Name
DR. MEDNICKDR. BASSIRNIA

Naomi is a 61-year-old woman who has lived with bipolar disorder and its stigma for 30 years. After a major manic episode and hospitalization, she entered into family treatment at the urging of her three daughters. Previously, her husband had been the primary force in guiding her psychiatric care, and she had been in treatment with a psychiatrist who is his professional colleague.

The patient’s first depressive episode began in the postpartum period, but she did not seek help at that time because she thought that her feelings were normal for a new mother. She did not receive any psychiatric attention until she cycled into mania and called the police for fear her child was being poisoned by neighbors. Her most recent manic episode occurred after she stopped her medications because of concerns about side effects. She was too embarrassed to tell her husband or doctor. She routinely fails to tell her other medical doctors that she is on mood stabilizers, because she does not want them to know she has bipolar disorder.


As Naomi recovers from the most recent manic episode and settles into family treatment, she is struggling with the consequences of her actions to her family. In family therapy in the past, her husband has revealed his belief that he has been protecting the family from Naomi’s mania and protecting Naomi from "embarrassing herself." This is difficult for Naomi to hear as she has always prided herself on being a good mother and protecting her daughters. Naomi’s situation illustrates the difficulty of coping with a diagnosis of bipolar disorder, the consequences of the illness on the family, and the importance of addressing stigma.

How stigma gets in the way

As discussed previously by Dr. Alison M. Heru ("Mental illness stigma is a family affair," Clinical Psychiatry News, April 2014, p. 8), stigma, when internalized or self-directed, can lead to psychological distress, decreased self-esteem and life satisfaction, and increased depression and suicidality (Compr. Psychiatry 2010;51:603-6). Close family members of those with mental disorders are affected by stigma, commonly referred to as "stigma by association" or "courtesy stigma."

Up to 92% of caregivers of people with psychiatric disorders have reported internalized stigma (J. Psychiatr. Ment. Health Nurs. 2012;19:665-71). These family members become distant and avoidant, resulting in a reduced quality of life and an impaired ability to provide critical support for their loved ones. Caregiver anxiety is inversely related to patient anxiety, stigma, and poor alliance (J. Nerv. Ment. Disease 2011;199:18-24).

As a result of these factors, while people with psychiatric disorders have to cope with their own mental illness as well as the public and self-stigma that alienate them from society, they also are at risk of losing their family connections.

In order to confront stigma, the Family Center for Bipolar Disorder in New York City, for example, uses a Family Inclusive Treatment (FIT) model. The FIT model includes an engagement period at the initiation of treatment that is focused on psychoeducation and relapse prevention planning. FIT is unique in that every patient is required to sign a release of information giving permission for full, open communication at all times between the patient’s clinician and a treatment partner of their choice. After the initial engagement period, there are quarterly family visits to supplement regular individual treatment sessions. FIT treatment promotes open communication about symptoms and medications. FIT strives to minimize patient isolation from families; they can talk openly with one another and their clinician.

Dr. Amy Mednick

After seeing many families enter treatment, FIT staff noticed the prominence of stigma.

We have begun to ask about stigma directly. Do people with more stigma do worse in treatment? Do they adhere more poorly to treatment? Do their families tend to become less involved over time? To begin, Dr. Mednick and staff examined demographic data looking for factors that might predispose a person to experience increased stigma.

In terms of diagnosis, people with more internalizing disorders such as depression and anxiety disorders tend to experience more stigma. Distress is experienced internally. As Dr. Bassirnia and her colleagues wrote in a poster presented at the recent American Psychiatric Association meeting, people with externalizing disorders, such as substance abuse and antisocial disorders, are more likely to express their distress outwardly and are less likely to suffer from stigma ("The relationship between personality traits and perceived internalized stigma in bipolar patients and their caregivers," 2014).

Meanwhile, two systematic review studies have reported moderate to high levels of internalized stigma in people with bipolar disorder. In these studies, a higher level of internalized stigma had a negative correlation with self-esteem, social adjustment, and perceived social support, and positive correlation with severity of symptoms, functional impairment, and rehospitalization. In spite of having more severe symptoms; people with higher levels of self-stigma are less likely to seek professional help and adhere to their treatment. Stigma by association and its negative consequences in caregivers of people with mental disorders also have been reported (J. Affect. Disord. 2013;150:181-91).

 

 

A useful and easy to administer scale that helps to identify stigma is the "Perceived Criticism Scale" (J. Abnorm. Psychol. 1989;98:225-35). By asking four questions, the clinician can get a good sense of family dynamics and can monitor the progress and change over time. The questions rate perception on a scale of 1-10, where "X" is the other person involved in treatment, either patient or caregiver. Here are the questions:

1. How critical do you think you are of X?

2. How critical do you think X is of you?

3. When X criticizes you, how upset do you get?

4. When you criticize X, how upset does he/she get?

For families with high scores, follow-up is needed. The Internalized Stigma of Mental Illness (ISMI) scale (Psychiatry Res. 2003;121:31-49) can be used. The ISMI scale makes statements about stigma for which participants rate their agreement on a Likert scale, such as:

• I don’t talk about myself much because I don’t want to burden others with my mental illness.

• Being around people who don’t have a mental illness makes me feel out of place or inadequate.

• People can tell that I have a mental illness by the way I look.

• Mentally ill people tend to be violent.

• I feel out of place in the world because I have a mental illness.

The ISMI scale contains 29 short, simple statements like the ones above and can be completed in less than 10 minutes. The statements are designed to avoid hypothetical situations, stay focused in the present, and address the participant’s own identity and experience.

Using the tools in practice

Naomi entered family treatment with her husband and daughters. Using the ISMI to measure the stigma of mental illness that each family member was experiencing, Naomi was shocked to see that her daughters felt far less stigma about having a mother with mental illness than she had assumed. In turn, her daughters were shocked at how much stigma Naomi was experiencing. Naomi’s husband scored between them. This data paved the way for an open family conversation about how Naomi’s illness had affected their lives, and especially how Naomi’s husband and his perceptions of her illness had affected her treatment course.

Caregivers play a very important role in bipolar disorder. After all, the illness can lead to difficulty functioning and can threaten the family’s stability. Sometimes caregivers can serve as a source of strength and a beacon of stability in the occasional storm. It is hard for the family between the storms, when the same flashing beacon can be a constant reminder to the patient of their illness. Often, well intentioned concerns become constant checking up, making the patient feel stigmatized and expected to fail.

"Good" caregivers will be aware of the stigma and the impact it has on their loved one and on themselves, without becoming a source of stigma.

Dr. Mednick is an attending psychiatrist at the Family Center for Bipolar at Mount Sinai Beth Israel in New York City. Dr. Bassirnia is a second-year psychiatry resident at Mount Sinai Beth Israel. Scan the QR code to read more Families in Psychiatry columns at clinicalpsychiatrynews.com.

Naomi is a 61-year-old woman who has lived with bipolar disorder and its stigma for 30 years. After a major manic episode and hospitalization, she entered into family treatment at the urging of her three daughters. Previously, her husband had been the primary force in guiding her psychiatric care, and she had been in treatment with a psychiatrist who is his professional colleague.

The patient’s first depressive episode began in the postpartum period, but she did not seek help at that time because she thought that her feelings were normal for a new mother. She did not receive any psychiatric attention until she cycled into mania and called the police for fear her child was being poisoned by neighbors. Her most recent manic episode occurred after she stopped her medications because of concerns about side effects. She was too embarrassed to tell her husband or doctor. She routinely fails to tell her other medical doctors that she is on mood stabilizers, because she does not want them to know she has bipolar disorder.


As Naomi recovers from the most recent manic episode and settles into family treatment, she is struggling with the consequences of her actions to her family. In family therapy in the past, her husband has revealed his belief that he has been protecting the family from Naomi’s mania and protecting Naomi from "embarrassing herself." This is difficult for Naomi to hear as she has always prided herself on being a good mother and protecting her daughters. Naomi’s situation illustrates the difficulty of coping with a diagnosis of bipolar disorder, the consequences of the illness on the family, and the importance of addressing stigma.

How stigma gets in the way

As discussed previously by Dr. Alison M. Heru ("Mental illness stigma is a family affair," Clinical Psychiatry News, April 2014, p. 8), stigma, when internalized or self-directed, can lead to psychological distress, decreased self-esteem and life satisfaction, and increased depression and suicidality (Compr. Psychiatry 2010;51:603-6). Close family members of those with mental disorders are affected by stigma, commonly referred to as "stigma by association" or "courtesy stigma."

Up to 92% of caregivers of people with psychiatric disorders have reported internalized stigma (J. Psychiatr. Ment. Health Nurs. 2012;19:665-71). These family members become distant and avoidant, resulting in a reduced quality of life and an impaired ability to provide critical support for their loved ones. Caregiver anxiety is inversely related to patient anxiety, stigma, and poor alliance (J. Nerv. Ment. Disease 2011;199:18-24).

As a result of these factors, while people with psychiatric disorders have to cope with their own mental illness as well as the public and self-stigma that alienate them from society, they also are at risk of losing their family connections.

In order to confront stigma, the Family Center for Bipolar Disorder in New York City, for example, uses a Family Inclusive Treatment (FIT) model. The FIT model includes an engagement period at the initiation of treatment that is focused on psychoeducation and relapse prevention planning. FIT is unique in that every patient is required to sign a release of information giving permission for full, open communication at all times between the patient’s clinician and a treatment partner of their choice. After the initial engagement period, there are quarterly family visits to supplement regular individual treatment sessions. FIT treatment promotes open communication about symptoms and medications. FIT strives to minimize patient isolation from families; they can talk openly with one another and their clinician.

Dr. Amy Mednick

After seeing many families enter treatment, FIT staff noticed the prominence of stigma.

We have begun to ask about stigma directly. Do people with more stigma do worse in treatment? Do they adhere more poorly to treatment? Do their families tend to become less involved over time? To begin, Dr. Mednick and staff examined demographic data looking for factors that might predispose a person to experience increased stigma.

In terms of diagnosis, people with more internalizing disorders such as depression and anxiety disorders tend to experience more stigma. Distress is experienced internally. As Dr. Bassirnia and her colleagues wrote in a poster presented at the recent American Psychiatric Association meeting, people with externalizing disorders, such as substance abuse and antisocial disorders, are more likely to express their distress outwardly and are less likely to suffer from stigma ("The relationship between personality traits and perceived internalized stigma in bipolar patients and their caregivers," 2014).

Meanwhile, two systematic review studies have reported moderate to high levels of internalized stigma in people with bipolar disorder. In these studies, a higher level of internalized stigma had a negative correlation with self-esteem, social adjustment, and perceived social support, and positive correlation with severity of symptoms, functional impairment, and rehospitalization. In spite of having more severe symptoms; people with higher levels of self-stigma are less likely to seek professional help and adhere to their treatment. Stigma by association and its negative consequences in caregivers of people with mental disorders also have been reported (J. Affect. Disord. 2013;150:181-91).

 

 

A useful and easy to administer scale that helps to identify stigma is the "Perceived Criticism Scale" (J. Abnorm. Psychol. 1989;98:225-35). By asking four questions, the clinician can get a good sense of family dynamics and can monitor the progress and change over time. The questions rate perception on a scale of 1-10, where "X" is the other person involved in treatment, either patient or caregiver. Here are the questions:

1. How critical do you think you are of X?

2. How critical do you think X is of you?

3. When X criticizes you, how upset do you get?

4. When you criticize X, how upset does he/she get?

For families with high scores, follow-up is needed. The Internalized Stigma of Mental Illness (ISMI) scale (Psychiatry Res. 2003;121:31-49) can be used. The ISMI scale makes statements about stigma for which participants rate their agreement on a Likert scale, such as:

• I don’t talk about myself much because I don’t want to burden others with my mental illness.

• Being around people who don’t have a mental illness makes me feel out of place or inadequate.

• People can tell that I have a mental illness by the way I look.

• Mentally ill people tend to be violent.

• I feel out of place in the world because I have a mental illness.

The ISMI scale contains 29 short, simple statements like the ones above and can be completed in less than 10 minutes. The statements are designed to avoid hypothetical situations, stay focused in the present, and address the participant’s own identity and experience.

Using the tools in practice

Naomi entered family treatment with her husband and daughters. Using the ISMI to measure the stigma of mental illness that each family member was experiencing, Naomi was shocked to see that her daughters felt far less stigma about having a mother with mental illness than she had assumed. In turn, her daughters were shocked at how much stigma Naomi was experiencing. Naomi’s husband scored between them. This data paved the way for an open family conversation about how Naomi’s illness had affected their lives, and especially how Naomi’s husband and his perceptions of her illness had affected her treatment course.

Caregivers play a very important role in bipolar disorder. After all, the illness can lead to difficulty functioning and can threaten the family’s stability. Sometimes caregivers can serve as a source of strength and a beacon of stability in the occasional storm. It is hard for the family between the storms, when the same flashing beacon can be a constant reminder to the patient of their illness. Often, well intentioned concerns become constant checking up, making the patient feel stigmatized and expected to fail.

"Good" caregivers will be aware of the stigma and the impact it has on their loved one and on themselves, without becoming a source of stigma.

Dr. Mednick is an attending psychiatrist at the Family Center for Bipolar at Mount Sinai Beth Israel in New York City. Dr. Bassirnia is a second-year psychiatry resident at Mount Sinai Beth Israel. Scan the QR code to read more Families in Psychiatry columns at clinicalpsychiatrynews.com.

Publications
Publications
Topics
Article Type
Display Headline
Four questions address stigma
Display Headline
Four questions address stigma
Legacy Keywords
bipolar disorder, stigma, manic episode, hospitalization, treatment, psychiatric care,
Legacy Keywords
bipolar disorder, stigma, manic episode, hospitalization, treatment, psychiatric care,
Sections
Article Source

PURLs Copyright

Inside the Article

Lithium for bipolar disorder

Article Type
Changed
Thu, 03/28/2019 - 15:45
Display Headline
Lithium for bipolar disorder

Author and Disclosure Information

Dr. McInnis is Thomas B. and Nancy Upjohn Woodworth Professor of Bipolar Disorder and Depression, Department of Psychiatry, University of Michigan Depression Center, Ann Arbor, Michigan.

Issue
Current Psychiatry - 13(6)
Publications
Topics
Legacy Keywords
clozapine, schizophrenia, practice trends
Sections
Author and Disclosure Information

Dr. McInnis is Thomas B. and Nancy Upjohn Woodworth Professor of Bipolar Disorder and Depression, Department of Psychiatry, University of Michigan Depression Center, Ann Arbor, Michigan.

Author and Disclosure Information

Dr. McInnis is Thomas B. and Nancy Upjohn Woodworth Professor of Bipolar Disorder and Depression, Department of Psychiatry, University of Michigan Depression Center, Ann Arbor, Michigan.

Related Articles

Issue
Current Psychiatry - 13(6)
Issue
Current Psychiatry - 13(6)
Publications
Publications
Topics
Article Type
Display Headline
Lithium for bipolar disorder
Display Headline
Lithium for bipolar disorder
Legacy Keywords
clozapine, schizophrenia, practice trends
Legacy Keywords
clozapine, schizophrenia, practice trends
Sections
Disallow All Ads
Alternative CME
Use ProPublica

Lithium for bipolar disorder: A re-emerging treatment for mood instability

Article Type
Changed
Tue, 12/11/2018 - 15:02
Display Headline
Lithium for bipolar disorder: A re-emerging treatment for mood instability

Lithium is among the most effective therapies for bipolar disorder (BD), and enthusiasm for this simple molecule is waxing. The his­tory of lithium is fascinating,1 and recent considerations include that this element, the third on the periodic table, has few, if any, indus­try champions. The recent renaissance is caused by a groundswell of appreciation for the clinical efficacy of lithium and an increasing num­ber of providers who are willing to manage patients with lithium.

Target: Bipolar disorder
The target illness for lithium is BD, a spectrum of mood disorders with characteristic features of unstable mood and affect. Shifts in mood include recurrent episodes of mania, which are pathologically ener­gized states with misguided volition and behavior with intoxicating euphoria (or irritability).2 Psychomotor activity is elevated and out of character; speech and body movements are revved up, with a dimin­ished need for sleep. The social, personal, and vocational consequences often are disastrous.

The most common mood state of BD is depression. Depressive episodes consist of pathologically compromised energy and volition with a slowing of bodily functions, most prominently cognition and concentration; a pervasive depressed or sad mood is common but not always present. Presence of mixed states, when features of depression and mania are present simultaneously, is one of the many challenges of treating BD; an elevated volitional or energized state may occur with a depressed, dysphoric mood.

Evidence for lithium
Efficacy studies of lithium have focused on managing mood disorders, treating mania and depression, and prevention or main­tenance care.3 Most were performed dur­ing the 1970s and 1980s,3 but recent studies have been comparing lithium with other mood stabilizers4-7 and searching for a genetic basis for lithium response.8-10 Other researchers have examined the use of lith­ium to prevent suicide.11 Some have sug­gested a neuroprotective effect of lithium, which may have profound implications for neuropsychiatry if valid.12-14 Results of additional studies, which are at differ­ent stages of completion, will clarify lith­ium use,15,16 and characterize the genetic makeup of individuals who respond to lithium.17 The primary evidence for lith­ium, however, is for maintenance treat­ment of BD and for preventing manic and depressive episodes.

Biochemistry and physiology of lithium. The biochemical and physiological effects of lithium are complex, wide-ranging, and likely to affect hundreds, if not thou­sands, of genes and gene products. The mechanisms of action remain a focus of aca­demic pursuit (for a review of hypotheses related to these mechanisms see Goodwin and Jamison2 and Can et al18) Lithium is involved in cell signaling pathways that involve complex molecular mechanisms of inter- and intracellular communica­tion19; some neural receptors are down-regulated20 and others show inhibition,21 which is thought to be a mechanism of lithium. The hypothesized neuroprotective effect of lithium22 may be mediated through an increased level of brain-derived neuro­trophic factor in brain tissue.14 Recently, investigators using induced pluripotent stem cell derived neurons have shown that patterns of calcium-related cell signaling in bipolar neurons are affected specifically by lithium in the culture media.23 There likely are many mechanisms through which lithi­um’s effects are mediated, including a series of dynamic pathways that vary over time and in reaction to the internal and external environments of the cell and person.

The lithium renaissance
In the past decade, there has been an increase in interest and use of lithium because clinicians recognize its efficacy and advantages and can monitor serum levels and gauge the patient’s response and side effects24 against the lithium level. This is important because balancing effi­ cacy and side effects depends on the serum level. Efficacy often is not immediate, although side effects may emerge early. All systems of the body may show effects that could be related to lithium use. It is helpful to be aware of the side effects in chronolog­ical order, because some immediate effects may be associated with starting at higher dosages (Table 1). Common side effects in the short term include:

   • GI distress, such as nausea, vomiting, diarrhea, and abdominal discomfort
   • a fine neurologic tremor, which may be seen with accentuation upon deliber­ate movement
   • prominent thirst with polyuria
   • drowsiness and clouded thinking, which can be upsetting to the patient and family.

In the longer term, adverse effects on kidney and thyroid function are common. Management must include monitoring of the serum level.

Lithium is FDA-approved for acute and maintenance treatment of mania in BD. There are reports that discuss most vari­ants of mood disorders, including BD I, BD II, unipolar depression, rapid cycling, and even alcohol abuse.25-29 Lithium could help manage mood dysregulation in the context of temperament and personal­ity.30 There is evidence that lithium has an antidepressant effect31-33 and has shown efficacy as an adjunctive treatment for depression.31-33 There are data that sug­gest that lithium, with its neuroprotective mechanisms, may prevent progression of mild cognitive impairment.34

 

 

Is there an ideal lithium candidate?
Mood instability is the characteristic fea­ture of a lithium responder. The instability may be over the course of the day, such as a dysregulated temperament that often is associated with DSM-IV personality cat­egories, shorter-term fluctuations (within days with BD II), or in the context of epi­sodic shifts of mood states over weeks and months, which are characteristic of BD I. The hallmark of mood instability is fluctuation from depression to elevated mood states and charged emotions with increased energy.

The patient considered ideal for lith­ium treatment has BD I with recurrent severe euphoric manic episodes, absence of significant comorbid disorders such as substance abuse, and a family history of lithium response. However, any patient with a clinically significant and unstable mood disorder, regardless of the DSM diagnosis, should be considered for lith­ium treatment.

When considering a lithium trial for a patient with significant mood instability, it is critical to establish the target symptoms and behavior that will help you gauge the efficacy of the intervention. Measurement-based care utilizes clinician and self-report instruments to provide data on the ill­ness course and response to interven­tion. Commonly used clinician driven assessments include the Young Mania Rating Scale35 and the Quick Inventory of Depressive Symptoms,36 while the self-report assessments are the Patient Health Questionnaire37 and the Altman Self- Rating Mania Scale.38

During acute mania or depression, lithium often is used in combination with another medications such as an antipsy­chotic or antidepressant. Used in the out­patient and non-acute setting, lithium may be an “add-on” or monotherapy for pre­venting recurrence of episodes. Response in early acute manic symptoms are predic­tive of later response and remission.39

Dosing strategies
An initial problem with lithium is side effects that emerge when beginning treat­ment, which may discourage the patient and family from using this agent. Starting with 150 mg/d for the first 2 or 3 doses is unlikely to produce any adverse effects and can show the patient that there is a high likelihood that he will be able to tol­erate the medication. Gradual titration over several days—or even weeks—to the target dosage and serum levels will enhance patient compliance. Rate of dos­age increase is best guided by tolerance to the medication. The general consensus is that lithium is most effective at levels of 0.6 to 0.8 mEq/L,40 although a lower level (0.5 mEq/L) over a 2-year period also can be effective.41 Lithium may be used in to treat acute mania at higher serum levels (0.8 to 1.2 mEq/L), however, the acute phase often requires urgent management, usually with an antipsychotic.

Emerging consensus
Although there is a need to gather and analyze longer observational periods to clarify the clinical and biological charac­teristics of persons who respond to lith­ium, there are several points of consensus. Management will be guided by patient characteristics such as age, comorbidities, and other therapies. Most studies that address the effect of lithium level focus on high vs low serum levels. There are 3 cat­egories of lithium serum levels, low (<0.6 mEq/L), mid-range (0.6 to 0.8 mEq/L), and high (>0.8 mEq/L), each has risk-benefit considerations.

The LiTMUS study42 compared low-level lithium augmentation with opti­mized personal treatment without lithium. Both groups had similar outcomes but the lithium-treated group had significantly lower use of atypical antipsychotics. This may be important when considering the long-term risk of the metabolic syndrome because the tolerability and side-effect profile of lithium at lower levels is more favorable than that of atypical antipsy­chotics. As lithium levels increase, there seems to be concomitant increase in effi­cacy and side effects. Many patients will benefit with low-level lithium use; yet clearly some individuals require higher dosages for effective maintenance therapy.

Dosing and monitoring. In patients age >50 or those with comorbid medical conditions, use a lower level of lithium (<0.6 mEq/L). Most individuals with BD likely will benefit from the mid-range level strategy (0.6 to 0.8 mEq/L); however, there will be those who require a higher level. When beginning lithium, start at a low dosage (150 mg/d) and increase as tolerated to the desired serum level. With acute mania, temporary use of an antipsychotic will be required.

There are no tests available to determine whether a patient will do well at any of these lithium serum levels. Breakthrough mania in an adherent patient with a serum lithium level of 0.7 mEq/L indicates the need to obtain a higher lithium level. A major deficit in lithium research is the lack of long-term data (>5 years) on out­comes, clinical and biological features with lithium levels because of a lack of pharma­ceutical company support.3,17 Monitoring mood symptoms using detailed mood charts, whether clinician-administered or self-reported, is an effective way to moni­tor outcomes, provided the clinician uses the same scales or methods to record a patient’s moods. If a patient wants to dis­continue lithium, taper the drug over an extended period (months) to minimize the likelihood of emerging manic or depressive episodes related to drug discontinuation.

Managing side effects
Consider lithium’s side effects in the con­text of their short-, intermediate-, and long-term presence (Table 2). Gradually increasing the lithium dosage often will prevent side effects that manifest in the short term. If side effects emerge at low dosages, proceed slowly with lithium and manage symptoms with other medica­tions. When a patient shows a change in side effects, obtain lithium and electrolytes levels; a change in mental status with con­fusion will require an acute lithium level.

 

 

A diary of symptoms or clinically rel­evant matters such as fluid intake or fre­quency of GI- or neurological-related events will help the clinician monitor the frequency and severity of side effects. The patient and clinician should not be dis­couraged by emerging side effects in the short term, because they may dissipate or become minimally intrusive.

Several strategies can alleviate immedi­ate GI effects, such as dosing with meals, enteric-coated formulations, multiple dose strategies, and short-term use of antidiar­rheal medicine as needed. Side effects that disrupt a patient’s fluid and electrolyte balance (diabetes insipidus) to the point of clouding mental status will require discon­tinuing the medication until mental sta­tus improves, then reconsideration of the treatment regime, which will include man­aging diabetes insipidus with amiloride. Managing side effects may require consulta­tion with specialty services. Likewise, some patients might experience neurologic side effects, such as profound tremor, that inter­feres with their ability to function. However, many side effects can be managed symp­tomatically with practical strategies (eg, a sugar-free lozenge for dry mouth or dysgeu­sia). Consider lower lithium dosages and serum levels because patients may experi­ence benefits with lower therapeutic levels.

Long-term side effects include decreased renal function, hypothyroidism, persistent tremor, and dermatologic effects of acne and alopecia. Monitor renal and thyroid function annually in stable patients and more frequently when making changes in the treatment plan.

Before discontinuing lithium, consider discussing the medical issues with a spe­cialist who has experience with complica­tions of lithium.

Bottom Line
Lithium is an effective and under used medication for managing bipolar disorder. Initial prejudices and side effects often deter patients and prescribers from proceeding with a therapeutic trial of lithium. Although the mid-range lithium level of 0.6 to 0.8 mEq/L is desirable, many patients will experience significant benefits with lower levels. Initial strategies include the use of low-dose preparations that are unlikely to have uncomfortable side effects.

Related Resources
• Andreasen A, Ellingrod VL. Lithium-induced diabetes in­sipidus: prevention and management. Current Psychiatry. 2013;12(7):42-45.
• Cipriani A, Hawton K, Stockton S, et al. Lithium in the pre­vention of suicide in mood disorders: updated system­atic review and meta-analysis. BMJ. 2013;346:f3646. doi: 10.1136/bmj.f3646.

Drug Brand Names
Amiloride • Midamor                Lithium • Eskalith, Lithobid


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

References


1. Shorter E. The history of lithium therapy. Bipolar Disord. 2009;11(11 suppl 2):4-9.
2. Goodwin FK, Jamison KR. Manic-depressive illness: bipolar disorders and recurrent depression. 2nd ed. New York, NY: Oxford University Press; 2007.
3. Burgess S, Geddes J, Hawton K, et al. Lithium for maintenance treatment of mood disorders. Cochrane Database Syst Rev. 2001:CD003013.
4. Bowden CL, Calabrese JR, McElroy SL, et al. A randomized, placebo-controlled 12-month trial of divalproex and lithium in treatment of outpatients with bipolar I disorder. Divalproex maintenance study group. Arch Gen Psychiatry. 2000;57(5):481-489.
5. Bowden CL, Calabrese JR, Sachs G, et al; Lamictal 606 Study Group. A placebo-controlled 18-month trial of lamotrigine and lithium maintenance treatment in recently manic or hypomanic patients with bipolar I disorder. Arch Gen Psychiatry. 2003;60(4):392-400.
6. Swann AC, Bowden CL, Calabrese JR, et al. Pattern of response to divalproex, lithium, or placebo in four naturalistic subtypes of mania. Neuropsychopharmacology. 2002;26(4):530-536.
7. 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.
8. Perlis RH, Smoller JW, Ferreira MA, et al. A genomewide association study of response to lithium for prevention of recurrence in bipolar disorder. Am J Psychiatry. 2009; 166(6):718-725.
9. Grof P, Duffy A, Cavazzoni P, et al. Is response to prophylactic lithium a familial trait? J Clin Psychiatry. 2002;63(10): 942-947.
10. Duffy A, Alda M, Kutcher S, et al. A prospective study of the offspring of bipolar parents responsive and nonresponsive to lithium treatment. J Clin Psychiatry. 2002;63(12): 1171-1178.
11. 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.
12. Quiroz JA, Machado-Vieira R, Zarate CA Jr, et al. Novel insights into lithium’s mechanism of action: neurotrophic and neuroprotective effects. Neuropsychobiology. 2010; 62(1):50-60.
13. Forlenza OV, Diniz BS, Radanovic M, et al. Disease-modifying properties of long-term lithium treatment for amnestic mild cognitive impairment: randomised controlled trial. Br J Psychiatry. 2011;198(5):351-356.
14. de Sousa RT, van de Bilt MT, Diniz BS, et al. Lithium increases plasma brain-derived neurotrophic factor in acute bipolar mania: a preliminary 4-week study. Neurosci Lett. 2011;494(1):54-56.
15. Nierenberg AA, Sylvia LG, Leon AC, et al; LiTMUS Study Group. Lithium treatment–moderate dose use study (LiTMUS) for bipolar disorder: rationale and design. Clin Trials. 2009;6(6):637-648.
16. Sylvia LG, Reilly-Harrington NA, Leon AC, et al. Methods to limit attrition in longitudinal comparative effectiveness trials: lessons from the Lithium Treatment - Moderate dose Use Study (LiTMUS) for bipolar disorder. Clin Trials. 2012;9(1):94-101.
17. McCarthy MJ, Leckband SG, Kelsoe JR. Pharmacogenetics of lithium response in bipolar disorder. Pharmacogenomics. 2010;11(10):1439-1465.
18. Can A, Schulze TG, Gould TD. Molecular actions and clinical pharmacogenetics of lithium therapy [published online February 15, 2014]. Pharmacol Biochem Behav. doi: 10.1016/j.pbb.2014.02.004.
19. Berridge MJ. Unlocking the secrets of cell signaling. Annu Rev Physiol. 2005;67:1-21.
20. Devaki R, Shankar Rao S, Nadgir SM. The effect of lithium on the adrenoceptor-mediated second messenger system in the rat brain. J Psychiatry Neurosci. 2006;31(4):246-252.
21. Pan JQ, Lewis MC, Ketterman JK, et al. AKT kinase activity is required for lithium to modulate mood-related behaviors in mice. Neuropsychopharmacology. 2011;36(7):1397-1411.
22. Hu LW, Kawamoto EM, Brietzke E, et al. The role of Wnt signaling and its interaction with diverse mechanisms of cellular apoptosis in the pathophysiology of bipolar disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35(1):11-17.
23. 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 [published online March 25, 2014]. Transl Psychiatry. doi:10.1038/tp.2014.12.
24. Jefferson JW. Lithium. In: Aronson JK, ed. Side effects of drugs annual, volume 26. Amsterdam, The Netherlands: Elsevier Science; 2003:19-29.
25. Baldessarini RJ, Tondo L, Floris G, et al. Effects of rapid cycling on response to lithium maintenance treatment in 360 bipolar I and II disorder patients. J Affect Disord. 2000;61(2):13-22.
26. Baldessarini RJ, Tondo L, Hennen J, et al. Latency and episodes before treatment: response to lithium maintenance in bipolar I and II disorders. Bipolar Disord. 1999;1(2): 91-97.
27. Fieve RR, Kumbaraci T, Dunner DL. Lithium prophylaxis of depression in bipolar I, bipolar II, and unipolar patients. Am J Psychiatry. 1976;133(8):925-929.
28. Peck CC, Pond SM, Becker CE, et al. An evaluation of the effects of lithium in the treatment of chronic alcoholism. II. Assessment of the two-period crossover design. Alcohol Clin Exp Res. 1981;5(2):252-255.
29. Peselow ED, Dunner DL, Fieve RR, et al. Lithium prophylaxis of depression in unipolar, bipolar II, and cyclothymic patients. Am J Psychiatry. 1982;139(6):747-752.
30. Bellino S, Paradiso E, Bogetto F. Efficacy and tolerability of pharmacotherapies for borderline personality disorder. CNS Drugs. 2008;22(8):671-692.
31. Alevizos B, Alevizos E, Leonardou A, et al. Low dosage lithium augmentation in venlafaxine resistant depression: an open-label study. Psychiatrike. 2012;23(2):143-148.
32. Goldberg JF, Sacks MH, Kocsis JH. Low-dose lithium augmentation of divalproex in geriatric mania. J Clin Psychiatry. 2000;61(4):304.
33. Saunders KE, Goodwin GM. New approaches in the treatment of bipolar depression. Curr Top Behav Neurosci. 2013;14:291-307.
34. Forlenza OV, Diniz BS, Radanovic M, et al. Disease-modifying properties of long-term lithium treatment for amnestic mild cognitive impairment: randomised controlled trial. Br J Psychiatry. 2011;198(5):351-356.
35. Young RC, Biggs JT, Ziegler VE, et al. A rating scale for mania: reliability, validity and sensitivity. Br J Psychiatry. 1978;133:429-435.
36. Trivedi MH, Rush AJ, Ibrahim HM, et al. The Inventory of Depressive Symptomatology, Clinician Rating (IDS-C) and Self-Report (IDS-SR), and the Quick Inventory of Depressive Symptomatology, Clinician Rating (QIDS-C) and Self-Report (QIDS-SR) in public sector patients with mood disorders: a psychometric evaluation. Psychol Med. 2004;34(1):73-82.
37. Kroenke K, Spitzer RL, Williams JB. The PHQ-9: validity of a brief depression severity measure. J Gen Intern Med. 2001;16(9):606-613.
38. Altman EG, Hedeker D, Peterson JL, et al. The Altman Self- Rating Mania Scale. Biol Psychiatry. 1997;42(10):948-955.
39. Machado-Vieira R, Luckenbaugh DA, Soeiro-de-Souza MG, et al. Early improvement with lithium in classic mania and its association with later response. J Affect Disord. 2013;144(1-2):160-164.
40. Severus WE, Lipkovich IA, Licht RW, et al. In search of optimal lithium levels and olanzapine doses in the long-term treatment of bipolar I disorder. A post-hoc analysis of the maintenance study by Tohen et al. 2005. Eur Psychiatry. 2010;25(8):443-449.
41. Vestergaard P, Licht RW, Brodersen A, et al. Outcome of lithium prophylaxis: a prospective follow-up of affective disorder patients assigned to high and low serum lithium levels. Acta Psychiatr Scand. 1998;98(4):310-315.
42. Nierenberg AA, Friedman ES, Bowden CL, et al. Lithium treatment moderate-dose use study (LiTMUS) for bipolar disorder: a randomized comparative effectiveness trial of optimized personalized treatment with and without lithium. Am J Psychiatry. 2013;170(1):102-111.

Article PDF
Author and Disclosure Information

Melvin G. McInnis, MD, FRCPsych
Thomas B. and Nancy Upjohn Woodworth
Professor of Bipolar Disorder and Depression
University of Michigan Depression Center
Department of Psychiatry
Ann Arbor, Michigan

Audio / Podcast
Issue
Current Psychiatry - 13(6)
Publications
Topics
Page Number
38-44
Legacy Keywords
lithium, bipolar disorder, lithium dosages, depression, mood disorders, mood instability
Sections
Audio / Podcast
Audio / Podcast
Author and Disclosure Information

Melvin G. McInnis, MD, FRCPsych
Thomas B. and Nancy Upjohn Woodworth
Professor of Bipolar Disorder and Depression
University of Michigan Depression Center
Department of Psychiatry
Ann Arbor, Michigan

Author and Disclosure Information

Melvin G. McInnis, MD, FRCPsych
Thomas B. and Nancy Upjohn Woodworth
Professor of Bipolar Disorder and Depression
University of Michigan Depression Center
Department of Psychiatry
Ann Arbor, Michigan

Article PDF
Article PDF
Related Articles

Lithium is among the most effective therapies for bipolar disorder (BD), and enthusiasm for this simple molecule is waxing. The his­tory of lithium is fascinating,1 and recent considerations include that this element, the third on the periodic table, has few, if any, indus­try champions. The recent renaissance is caused by a groundswell of appreciation for the clinical efficacy of lithium and an increasing num­ber of providers who are willing to manage patients with lithium.

Target: Bipolar disorder
The target illness for lithium is BD, a spectrum of mood disorders with characteristic features of unstable mood and affect. Shifts in mood include recurrent episodes of mania, which are pathologically ener­gized states with misguided volition and behavior with intoxicating euphoria (or irritability).2 Psychomotor activity is elevated and out of character; speech and body movements are revved up, with a dimin­ished need for sleep. The social, personal, and vocational consequences often are disastrous.

The most common mood state of BD is depression. Depressive episodes consist of pathologically compromised energy and volition with a slowing of bodily functions, most prominently cognition and concentration; a pervasive depressed or sad mood is common but not always present. Presence of mixed states, when features of depression and mania are present simultaneously, is one of the many challenges of treating BD; an elevated volitional or energized state may occur with a depressed, dysphoric mood.

Evidence for lithium
Efficacy studies of lithium have focused on managing mood disorders, treating mania and depression, and prevention or main­tenance care.3 Most were performed dur­ing the 1970s and 1980s,3 but recent studies have been comparing lithium with other mood stabilizers4-7 and searching for a genetic basis for lithium response.8-10 Other researchers have examined the use of lith­ium to prevent suicide.11 Some have sug­gested a neuroprotective effect of lithium, which may have profound implications for neuropsychiatry if valid.12-14 Results of additional studies, which are at differ­ent stages of completion, will clarify lith­ium use,15,16 and characterize the genetic makeup of individuals who respond to lithium.17 The primary evidence for lith­ium, however, is for maintenance treat­ment of BD and for preventing manic and depressive episodes.

Biochemistry and physiology of lithium. The biochemical and physiological effects of lithium are complex, wide-ranging, and likely to affect hundreds, if not thou­sands, of genes and gene products. The mechanisms of action remain a focus of aca­demic pursuit (for a review of hypotheses related to these mechanisms see Goodwin and Jamison2 and Can et al18) Lithium is involved in cell signaling pathways that involve complex molecular mechanisms of inter- and intracellular communica­tion19; some neural receptors are down-regulated20 and others show inhibition,21 which is thought to be a mechanism of lithium. The hypothesized neuroprotective effect of lithium22 may be mediated through an increased level of brain-derived neuro­trophic factor in brain tissue.14 Recently, investigators using induced pluripotent stem cell derived neurons have shown that patterns of calcium-related cell signaling in bipolar neurons are affected specifically by lithium in the culture media.23 There likely are many mechanisms through which lithi­um’s effects are mediated, including a series of dynamic pathways that vary over time and in reaction to the internal and external environments of the cell and person.

The lithium renaissance
In the past decade, there has been an increase in interest and use of lithium because clinicians recognize its efficacy and advantages and can monitor serum levels and gauge the patient’s response and side effects24 against the lithium level. This is important because balancing effi­ cacy and side effects depends on the serum level. Efficacy often is not immediate, although side effects may emerge early. All systems of the body may show effects that could be related to lithium use. It is helpful to be aware of the side effects in chronolog­ical order, because some immediate effects may be associated with starting at higher dosages (Table 1). Common side effects in the short term include:

   • GI distress, such as nausea, vomiting, diarrhea, and abdominal discomfort
   • a fine neurologic tremor, which may be seen with accentuation upon deliber­ate movement
   • prominent thirst with polyuria
   • drowsiness and clouded thinking, which can be upsetting to the patient and family.

In the longer term, adverse effects on kidney and thyroid function are common. Management must include monitoring of the serum level.

Lithium is FDA-approved for acute and maintenance treatment of mania in BD. There are reports that discuss most vari­ants of mood disorders, including BD I, BD II, unipolar depression, rapid cycling, and even alcohol abuse.25-29 Lithium could help manage mood dysregulation in the context of temperament and personal­ity.30 There is evidence that lithium has an antidepressant effect31-33 and has shown efficacy as an adjunctive treatment for depression.31-33 There are data that sug­gest that lithium, with its neuroprotective mechanisms, may prevent progression of mild cognitive impairment.34

 

 

Is there an ideal lithium candidate?
Mood instability is the characteristic fea­ture of a lithium responder. The instability may be over the course of the day, such as a dysregulated temperament that often is associated with DSM-IV personality cat­egories, shorter-term fluctuations (within days with BD II), or in the context of epi­sodic shifts of mood states over weeks and months, which are characteristic of BD I. The hallmark of mood instability is fluctuation from depression to elevated mood states and charged emotions with increased energy.

The patient considered ideal for lith­ium treatment has BD I with recurrent severe euphoric manic episodes, absence of significant comorbid disorders such as substance abuse, and a family history of lithium response. However, any patient with a clinically significant and unstable mood disorder, regardless of the DSM diagnosis, should be considered for lith­ium treatment.

When considering a lithium trial for a patient with significant mood instability, it is critical to establish the target symptoms and behavior that will help you gauge the efficacy of the intervention. Measurement-based care utilizes clinician and self-report instruments to provide data on the ill­ness course and response to interven­tion. Commonly used clinician driven assessments include the Young Mania Rating Scale35 and the Quick Inventory of Depressive Symptoms,36 while the self-report assessments are the Patient Health Questionnaire37 and the Altman Self- Rating Mania Scale.38

During acute mania or depression, lithium often is used in combination with another medications such as an antipsy­chotic or antidepressant. Used in the out­patient and non-acute setting, lithium may be an “add-on” or monotherapy for pre­venting recurrence of episodes. Response in early acute manic symptoms are predic­tive of later response and remission.39

Dosing strategies
An initial problem with lithium is side effects that emerge when beginning treat­ment, which may discourage the patient and family from using this agent. Starting with 150 mg/d for the first 2 or 3 doses is unlikely to produce any adverse effects and can show the patient that there is a high likelihood that he will be able to tol­erate the medication. Gradual titration over several days—or even weeks—to the target dosage and serum levels will enhance patient compliance. Rate of dos­age increase is best guided by tolerance to the medication. The general consensus is that lithium is most effective at levels of 0.6 to 0.8 mEq/L,40 although a lower level (0.5 mEq/L) over a 2-year period also can be effective.41 Lithium may be used in to treat acute mania at higher serum levels (0.8 to 1.2 mEq/L), however, the acute phase often requires urgent management, usually with an antipsychotic.

Emerging consensus
Although there is a need to gather and analyze longer observational periods to clarify the clinical and biological charac­teristics of persons who respond to lith­ium, there are several points of consensus. Management will be guided by patient characteristics such as age, comorbidities, and other therapies. Most studies that address the effect of lithium level focus on high vs low serum levels. There are 3 cat­egories of lithium serum levels, low (<0.6 mEq/L), mid-range (0.6 to 0.8 mEq/L), and high (>0.8 mEq/L), each has risk-benefit considerations.

The LiTMUS study42 compared low-level lithium augmentation with opti­mized personal treatment without lithium. Both groups had similar outcomes but the lithium-treated group had significantly lower use of atypical antipsychotics. This may be important when considering the long-term risk of the metabolic syndrome because the tolerability and side-effect profile of lithium at lower levels is more favorable than that of atypical antipsy­chotics. As lithium levels increase, there seems to be concomitant increase in effi­cacy and side effects. Many patients will benefit with low-level lithium use; yet clearly some individuals require higher dosages for effective maintenance therapy.

Dosing and monitoring. In patients age >50 or those with comorbid medical conditions, use a lower level of lithium (<0.6 mEq/L). Most individuals with BD likely will benefit from the mid-range level strategy (0.6 to 0.8 mEq/L); however, there will be those who require a higher level. When beginning lithium, start at a low dosage (150 mg/d) and increase as tolerated to the desired serum level. With acute mania, temporary use of an antipsychotic will be required.

There are no tests available to determine whether a patient will do well at any of these lithium serum levels. Breakthrough mania in an adherent patient with a serum lithium level of 0.7 mEq/L indicates the need to obtain a higher lithium level. A major deficit in lithium research is the lack of long-term data (>5 years) on out­comes, clinical and biological features with lithium levels because of a lack of pharma­ceutical company support.3,17 Monitoring mood symptoms using detailed mood charts, whether clinician-administered or self-reported, is an effective way to moni­tor outcomes, provided the clinician uses the same scales or methods to record a patient’s moods. If a patient wants to dis­continue lithium, taper the drug over an extended period (months) to minimize the likelihood of emerging manic or depressive episodes related to drug discontinuation.

Managing side effects
Consider lithium’s side effects in the con­text of their short-, intermediate-, and long-term presence (Table 2). Gradually increasing the lithium dosage often will prevent side effects that manifest in the short term. If side effects emerge at low dosages, proceed slowly with lithium and manage symptoms with other medica­tions. When a patient shows a change in side effects, obtain lithium and electrolytes levels; a change in mental status with con­fusion will require an acute lithium level.

 

 

A diary of symptoms or clinically rel­evant matters such as fluid intake or fre­quency of GI- or neurological-related events will help the clinician monitor the frequency and severity of side effects. The patient and clinician should not be dis­couraged by emerging side effects in the short term, because they may dissipate or become minimally intrusive.

Several strategies can alleviate immedi­ate GI effects, such as dosing with meals, enteric-coated formulations, multiple dose strategies, and short-term use of antidiar­rheal medicine as needed. Side effects that disrupt a patient’s fluid and electrolyte balance (diabetes insipidus) to the point of clouding mental status will require discon­tinuing the medication until mental sta­tus improves, then reconsideration of the treatment regime, which will include man­aging diabetes insipidus with amiloride. Managing side effects may require consulta­tion with specialty services. Likewise, some patients might experience neurologic side effects, such as profound tremor, that inter­feres with their ability to function. However, many side effects can be managed symp­tomatically with practical strategies (eg, a sugar-free lozenge for dry mouth or dysgeu­sia). Consider lower lithium dosages and serum levels because patients may experi­ence benefits with lower therapeutic levels.

Long-term side effects include decreased renal function, hypothyroidism, persistent tremor, and dermatologic effects of acne and alopecia. Monitor renal and thyroid function annually in stable patients and more frequently when making changes in the treatment plan.

Before discontinuing lithium, consider discussing the medical issues with a spe­cialist who has experience with complica­tions of lithium.

Bottom Line
Lithium is an effective and under used medication for managing bipolar disorder. Initial prejudices and side effects often deter patients and prescribers from proceeding with a therapeutic trial of lithium. Although the mid-range lithium level of 0.6 to 0.8 mEq/L is desirable, many patients will experience significant benefits with lower levels. Initial strategies include the use of low-dose preparations that are unlikely to have uncomfortable side effects.

Related Resources
• Andreasen A, Ellingrod VL. Lithium-induced diabetes in­sipidus: prevention and management. Current Psychiatry. 2013;12(7):42-45.
• Cipriani A, Hawton K, Stockton S, et al. Lithium in the pre­vention of suicide in mood disorders: updated system­atic review and meta-analysis. BMJ. 2013;346:f3646. doi: 10.1136/bmj.f3646.

Drug Brand Names
Amiloride • Midamor                Lithium • Eskalith, Lithobid


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

Lithium is among the most effective therapies for bipolar disorder (BD), and enthusiasm for this simple molecule is waxing. The his­tory of lithium is fascinating,1 and recent considerations include that this element, the third on the periodic table, has few, if any, indus­try champions. The recent renaissance is caused by a groundswell of appreciation for the clinical efficacy of lithium and an increasing num­ber of providers who are willing to manage patients with lithium.

Target: Bipolar disorder
The target illness for lithium is BD, a spectrum of mood disorders with characteristic features of unstable mood and affect. Shifts in mood include recurrent episodes of mania, which are pathologically ener­gized states with misguided volition and behavior with intoxicating euphoria (or irritability).2 Psychomotor activity is elevated and out of character; speech and body movements are revved up, with a dimin­ished need for sleep. The social, personal, and vocational consequences often are disastrous.

The most common mood state of BD is depression. Depressive episodes consist of pathologically compromised energy and volition with a slowing of bodily functions, most prominently cognition and concentration; a pervasive depressed or sad mood is common but not always present. Presence of mixed states, when features of depression and mania are present simultaneously, is one of the many challenges of treating BD; an elevated volitional or energized state may occur with a depressed, dysphoric mood.

Evidence for lithium
Efficacy studies of lithium have focused on managing mood disorders, treating mania and depression, and prevention or main­tenance care.3 Most were performed dur­ing the 1970s and 1980s,3 but recent studies have been comparing lithium with other mood stabilizers4-7 and searching for a genetic basis for lithium response.8-10 Other researchers have examined the use of lith­ium to prevent suicide.11 Some have sug­gested a neuroprotective effect of lithium, which may have profound implications for neuropsychiatry if valid.12-14 Results of additional studies, which are at differ­ent stages of completion, will clarify lith­ium use,15,16 and characterize the genetic makeup of individuals who respond to lithium.17 The primary evidence for lith­ium, however, is for maintenance treat­ment of BD and for preventing manic and depressive episodes.

Biochemistry and physiology of lithium. The biochemical and physiological effects of lithium are complex, wide-ranging, and likely to affect hundreds, if not thou­sands, of genes and gene products. The mechanisms of action remain a focus of aca­demic pursuit (for a review of hypotheses related to these mechanisms see Goodwin and Jamison2 and Can et al18) Lithium is involved in cell signaling pathways that involve complex molecular mechanisms of inter- and intracellular communica­tion19; some neural receptors are down-regulated20 and others show inhibition,21 which is thought to be a mechanism of lithium. The hypothesized neuroprotective effect of lithium22 may be mediated through an increased level of brain-derived neuro­trophic factor in brain tissue.14 Recently, investigators using induced pluripotent stem cell derived neurons have shown that patterns of calcium-related cell signaling in bipolar neurons are affected specifically by lithium in the culture media.23 There likely are many mechanisms through which lithi­um’s effects are mediated, including a series of dynamic pathways that vary over time and in reaction to the internal and external environments of the cell and person.

The lithium renaissance
In the past decade, there has been an increase in interest and use of lithium because clinicians recognize its efficacy and advantages and can monitor serum levels and gauge the patient’s response and side effects24 against the lithium level. This is important because balancing effi­ cacy and side effects depends on the serum level. Efficacy often is not immediate, although side effects may emerge early. All systems of the body may show effects that could be related to lithium use. It is helpful to be aware of the side effects in chronolog­ical order, because some immediate effects may be associated with starting at higher dosages (Table 1). Common side effects in the short term include:

   • GI distress, such as nausea, vomiting, diarrhea, and abdominal discomfort
   • a fine neurologic tremor, which may be seen with accentuation upon deliber­ate movement
   • prominent thirst with polyuria
   • drowsiness and clouded thinking, which can be upsetting to the patient and family.

In the longer term, adverse effects on kidney and thyroid function are common. Management must include monitoring of the serum level.

Lithium is FDA-approved for acute and maintenance treatment of mania in BD. There are reports that discuss most vari­ants of mood disorders, including BD I, BD II, unipolar depression, rapid cycling, and even alcohol abuse.25-29 Lithium could help manage mood dysregulation in the context of temperament and personal­ity.30 There is evidence that lithium has an antidepressant effect31-33 and has shown efficacy as an adjunctive treatment for depression.31-33 There are data that sug­gest that lithium, with its neuroprotective mechanisms, may prevent progression of mild cognitive impairment.34

 

 

Is there an ideal lithium candidate?
Mood instability is the characteristic fea­ture of a lithium responder. The instability may be over the course of the day, such as a dysregulated temperament that often is associated with DSM-IV personality cat­egories, shorter-term fluctuations (within days with BD II), or in the context of epi­sodic shifts of mood states over weeks and months, which are characteristic of BD I. The hallmark of mood instability is fluctuation from depression to elevated mood states and charged emotions with increased energy.

The patient considered ideal for lith­ium treatment has BD I with recurrent severe euphoric manic episodes, absence of significant comorbid disorders such as substance abuse, and a family history of lithium response. However, any patient with a clinically significant and unstable mood disorder, regardless of the DSM diagnosis, should be considered for lith­ium treatment.

When considering a lithium trial for a patient with significant mood instability, it is critical to establish the target symptoms and behavior that will help you gauge the efficacy of the intervention. Measurement-based care utilizes clinician and self-report instruments to provide data on the ill­ness course and response to interven­tion. Commonly used clinician driven assessments include the Young Mania Rating Scale35 and the Quick Inventory of Depressive Symptoms,36 while the self-report assessments are the Patient Health Questionnaire37 and the Altman Self- Rating Mania Scale.38

During acute mania or depression, lithium often is used in combination with another medications such as an antipsy­chotic or antidepressant. Used in the out­patient and non-acute setting, lithium may be an “add-on” or monotherapy for pre­venting recurrence of episodes. Response in early acute manic symptoms are predic­tive of later response and remission.39

Dosing strategies
An initial problem with lithium is side effects that emerge when beginning treat­ment, which may discourage the patient and family from using this agent. Starting with 150 mg/d for the first 2 or 3 doses is unlikely to produce any adverse effects and can show the patient that there is a high likelihood that he will be able to tol­erate the medication. Gradual titration over several days—or even weeks—to the target dosage and serum levels will enhance patient compliance. Rate of dos­age increase is best guided by tolerance to the medication. The general consensus is that lithium is most effective at levels of 0.6 to 0.8 mEq/L,40 although a lower level (0.5 mEq/L) over a 2-year period also can be effective.41 Lithium may be used in to treat acute mania at higher serum levels (0.8 to 1.2 mEq/L), however, the acute phase often requires urgent management, usually with an antipsychotic.

Emerging consensus
Although there is a need to gather and analyze longer observational periods to clarify the clinical and biological charac­teristics of persons who respond to lith­ium, there are several points of consensus. Management will be guided by patient characteristics such as age, comorbidities, and other therapies. Most studies that address the effect of lithium level focus on high vs low serum levels. There are 3 cat­egories of lithium serum levels, low (<0.6 mEq/L), mid-range (0.6 to 0.8 mEq/L), and high (>0.8 mEq/L), each has risk-benefit considerations.

The LiTMUS study42 compared low-level lithium augmentation with opti­mized personal treatment without lithium. Both groups had similar outcomes but the lithium-treated group had significantly lower use of atypical antipsychotics. This may be important when considering the long-term risk of the metabolic syndrome because the tolerability and side-effect profile of lithium at lower levels is more favorable than that of atypical antipsy­chotics. As lithium levels increase, there seems to be concomitant increase in effi­cacy and side effects. Many patients will benefit with low-level lithium use; yet clearly some individuals require higher dosages for effective maintenance therapy.

Dosing and monitoring. In patients age >50 or those with comorbid medical conditions, use a lower level of lithium (<0.6 mEq/L). Most individuals with BD likely will benefit from the mid-range level strategy (0.6 to 0.8 mEq/L); however, there will be those who require a higher level. When beginning lithium, start at a low dosage (150 mg/d) and increase as tolerated to the desired serum level. With acute mania, temporary use of an antipsychotic will be required.

There are no tests available to determine whether a patient will do well at any of these lithium serum levels. Breakthrough mania in an adherent patient with a serum lithium level of 0.7 mEq/L indicates the need to obtain a higher lithium level. A major deficit in lithium research is the lack of long-term data (>5 years) on out­comes, clinical and biological features with lithium levels because of a lack of pharma­ceutical company support.3,17 Monitoring mood symptoms using detailed mood charts, whether clinician-administered or self-reported, is an effective way to moni­tor outcomes, provided the clinician uses the same scales or methods to record a patient’s moods. If a patient wants to dis­continue lithium, taper the drug over an extended period (months) to minimize the likelihood of emerging manic or depressive episodes related to drug discontinuation.

Managing side effects
Consider lithium’s side effects in the con­text of their short-, intermediate-, and long-term presence (Table 2). Gradually increasing the lithium dosage often will prevent side effects that manifest in the short term. If side effects emerge at low dosages, proceed slowly with lithium and manage symptoms with other medica­tions. When a patient shows a change in side effects, obtain lithium and electrolytes levels; a change in mental status with con­fusion will require an acute lithium level.

 

 

A diary of symptoms or clinically rel­evant matters such as fluid intake or fre­quency of GI- or neurological-related events will help the clinician monitor the frequency and severity of side effects. The patient and clinician should not be dis­couraged by emerging side effects in the short term, because they may dissipate or become minimally intrusive.

Several strategies can alleviate immedi­ate GI effects, such as dosing with meals, enteric-coated formulations, multiple dose strategies, and short-term use of antidiar­rheal medicine as needed. Side effects that disrupt a patient’s fluid and electrolyte balance (diabetes insipidus) to the point of clouding mental status will require discon­tinuing the medication until mental sta­tus improves, then reconsideration of the treatment regime, which will include man­aging diabetes insipidus with amiloride. Managing side effects may require consulta­tion with specialty services. Likewise, some patients might experience neurologic side effects, such as profound tremor, that inter­feres with their ability to function. However, many side effects can be managed symp­tomatically with practical strategies (eg, a sugar-free lozenge for dry mouth or dysgeu­sia). Consider lower lithium dosages and serum levels because patients may experi­ence benefits with lower therapeutic levels.

Long-term side effects include decreased renal function, hypothyroidism, persistent tremor, and dermatologic effects of acne and alopecia. Monitor renal and thyroid function annually in stable patients and more frequently when making changes in the treatment plan.

Before discontinuing lithium, consider discussing the medical issues with a spe­cialist who has experience with complica­tions of lithium.

Bottom Line
Lithium is an effective and under used medication for managing bipolar disorder. Initial prejudices and side effects often deter patients and prescribers from proceeding with a therapeutic trial of lithium. Although the mid-range lithium level of 0.6 to 0.8 mEq/L is desirable, many patients will experience significant benefits with lower levels. Initial strategies include the use of low-dose preparations that are unlikely to have uncomfortable side effects.

Related Resources
• Andreasen A, Ellingrod VL. Lithium-induced diabetes in­sipidus: prevention and management. Current Psychiatry. 2013;12(7):42-45.
• Cipriani A, Hawton K, Stockton S, et al. Lithium in the pre­vention of suicide in mood disorders: updated system­atic review and meta-analysis. BMJ. 2013;346:f3646. doi: 10.1136/bmj.f3646.

Drug Brand Names
Amiloride • Midamor                Lithium • Eskalith, Lithobid


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

References


1. Shorter E. The history of lithium therapy. Bipolar Disord. 2009;11(11 suppl 2):4-9.
2. Goodwin FK, Jamison KR. Manic-depressive illness: bipolar disorders and recurrent depression. 2nd ed. New York, NY: Oxford University Press; 2007.
3. Burgess S, Geddes J, Hawton K, et al. Lithium for maintenance treatment of mood disorders. Cochrane Database Syst Rev. 2001:CD003013.
4. Bowden CL, Calabrese JR, McElroy SL, et al. A randomized, placebo-controlled 12-month trial of divalproex and lithium in treatment of outpatients with bipolar I disorder. Divalproex maintenance study group. Arch Gen Psychiatry. 2000;57(5):481-489.
5. Bowden CL, Calabrese JR, Sachs G, et al; Lamictal 606 Study Group. A placebo-controlled 18-month trial of lamotrigine and lithium maintenance treatment in recently manic or hypomanic patients with bipolar I disorder. Arch Gen Psychiatry. 2003;60(4):392-400.
6. Swann AC, Bowden CL, Calabrese JR, et al. Pattern of response to divalproex, lithium, or placebo in four naturalistic subtypes of mania. Neuropsychopharmacology. 2002;26(4):530-536.
7. 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.
8. Perlis RH, Smoller JW, Ferreira MA, et al. A genomewide association study of response to lithium for prevention of recurrence in bipolar disorder. Am J Psychiatry. 2009; 166(6):718-725.
9. Grof P, Duffy A, Cavazzoni P, et al. Is response to prophylactic lithium a familial trait? J Clin Psychiatry. 2002;63(10): 942-947.
10. Duffy A, Alda M, Kutcher S, et al. A prospective study of the offspring of bipolar parents responsive and nonresponsive to lithium treatment. J Clin Psychiatry. 2002;63(12): 1171-1178.
11. 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.
12. Quiroz JA, Machado-Vieira R, Zarate CA Jr, et al. Novel insights into lithium’s mechanism of action: neurotrophic and neuroprotective effects. Neuropsychobiology. 2010; 62(1):50-60.
13. Forlenza OV, Diniz BS, Radanovic M, et al. Disease-modifying properties of long-term lithium treatment for amnestic mild cognitive impairment: randomised controlled trial. Br J Psychiatry. 2011;198(5):351-356.
14. de Sousa RT, van de Bilt MT, Diniz BS, et al. Lithium increases plasma brain-derived neurotrophic factor in acute bipolar mania: a preliminary 4-week study. Neurosci Lett. 2011;494(1):54-56.
15. Nierenberg AA, Sylvia LG, Leon AC, et al; LiTMUS Study Group. Lithium treatment–moderate dose use study (LiTMUS) for bipolar disorder: rationale and design. Clin Trials. 2009;6(6):637-648.
16. Sylvia LG, Reilly-Harrington NA, Leon AC, et al. Methods to limit attrition in longitudinal comparative effectiveness trials: lessons from the Lithium Treatment - Moderate dose Use Study (LiTMUS) for bipolar disorder. Clin Trials. 2012;9(1):94-101.
17. McCarthy MJ, Leckband SG, Kelsoe JR. Pharmacogenetics of lithium response in bipolar disorder. Pharmacogenomics. 2010;11(10):1439-1465.
18. Can A, Schulze TG, Gould TD. Molecular actions and clinical pharmacogenetics of lithium therapy [published online February 15, 2014]. Pharmacol Biochem Behav. doi: 10.1016/j.pbb.2014.02.004.
19. Berridge MJ. Unlocking the secrets of cell signaling. Annu Rev Physiol. 2005;67:1-21.
20. Devaki R, Shankar Rao S, Nadgir SM. The effect of lithium on the adrenoceptor-mediated second messenger system in the rat brain. J Psychiatry Neurosci. 2006;31(4):246-252.
21. Pan JQ, Lewis MC, Ketterman JK, et al. AKT kinase activity is required for lithium to modulate mood-related behaviors in mice. Neuropsychopharmacology. 2011;36(7):1397-1411.
22. Hu LW, Kawamoto EM, Brietzke E, et al. The role of Wnt signaling and its interaction with diverse mechanisms of cellular apoptosis in the pathophysiology of bipolar disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35(1):11-17.
23. 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 [published online March 25, 2014]. Transl Psychiatry. doi:10.1038/tp.2014.12.
24. Jefferson JW. Lithium. In: Aronson JK, ed. Side effects of drugs annual, volume 26. Amsterdam, The Netherlands: Elsevier Science; 2003:19-29.
25. Baldessarini RJ, Tondo L, Floris G, et al. Effects of rapid cycling on response to lithium maintenance treatment in 360 bipolar I and II disorder patients. J Affect Disord. 2000;61(2):13-22.
26. Baldessarini RJ, Tondo L, Hennen J, et al. Latency and episodes before treatment: response to lithium maintenance in bipolar I and II disorders. Bipolar Disord. 1999;1(2): 91-97.
27. Fieve RR, Kumbaraci T, Dunner DL. Lithium prophylaxis of depression in bipolar I, bipolar II, and unipolar patients. Am J Psychiatry. 1976;133(8):925-929.
28. Peck CC, Pond SM, Becker CE, et al. An evaluation of the effects of lithium in the treatment of chronic alcoholism. II. Assessment of the two-period crossover design. Alcohol Clin Exp Res. 1981;5(2):252-255.
29. Peselow ED, Dunner DL, Fieve RR, et al. Lithium prophylaxis of depression in unipolar, bipolar II, and cyclothymic patients. Am J Psychiatry. 1982;139(6):747-752.
30. Bellino S, Paradiso E, Bogetto F. Efficacy and tolerability of pharmacotherapies for borderline personality disorder. CNS Drugs. 2008;22(8):671-692.
31. Alevizos B, Alevizos E, Leonardou A, et al. Low dosage lithium augmentation in venlafaxine resistant depression: an open-label study. Psychiatrike. 2012;23(2):143-148.
32. Goldberg JF, Sacks MH, Kocsis JH. Low-dose lithium augmentation of divalproex in geriatric mania. J Clin Psychiatry. 2000;61(4):304.
33. Saunders KE, Goodwin GM. New approaches in the treatment of bipolar depression. Curr Top Behav Neurosci. 2013;14:291-307.
34. Forlenza OV, Diniz BS, Radanovic M, et al. Disease-modifying properties of long-term lithium treatment for amnestic mild cognitive impairment: randomised controlled trial. Br J Psychiatry. 2011;198(5):351-356.
35. Young RC, Biggs JT, Ziegler VE, et al. A rating scale for mania: reliability, validity and sensitivity. Br J Psychiatry. 1978;133:429-435.
36. Trivedi MH, Rush AJ, Ibrahim HM, et al. The Inventory of Depressive Symptomatology, Clinician Rating (IDS-C) and Self-Report (IDS-SR), and the Quick Inventory of Depressive Symptomatology, Clinician Rating (QIDS-C) and Self-Report (QIDS-SR) in public sector patients with mood disorders: a psychometric evaluation. Psychol Med. 2004;34(1):73-82.
37. Kroenke K, Spitzer RL, Williams JB. The PHQ-9: validity of a brief depression severity measure. J Gen Intern Med. 2001;16(9):606-613.
38. Altman EG, Hedeker D, Peterson JL, et al. The Altman Self- Rating Mania Scale. Biol Psychiatry. 1997;42(10):948-955.
39. Machado-Vieira R, Luckenbaugh DA, Soeiro-de-Souza MG, et al. Early improvement with lithium in classic mania and its association with later response. J Affect Disord. 2013;144(1-2):160-164.
40. Severus WE, Lipkovich IA, Licht RW, et al. In search of optimal lithium levels and olanzapine doses in the long-term treatment of bipolar I disorder. A post-hoc analysis of the maintenance study by Tohen et al. 2005. Eur Psychiatry. 2010;25(8):443-449.
41. Vestergaard P, Licht RW, Brodersen A, et al. Outcome of lithium prophylaxis: a prospective follow-up of affective disorder patients assigned to high and low serum lithium levels. Acta Psychiatr Scand. 1998;98(4):310-315.
42. Nierenberg AA, Friedman ES, Bowden CL, et al. Lithium treatment moderate-dose use study (LiTMUS) for bipolar disorder: a randomized comparative effectiveness trial of optimized personalized treatment with and without lithium. Am J Psychiatry. 2013;170(1):102-111.

References


1. Shorter E. The history of lithium therapy. Bipolar Disord. 2009;11(11 suppl 2):4-9.
2. Goodwin FK, Jamison KR. Manic-depressive illness: bipolar disorders and recurrent depression. 2nd ed. New York, NY: Oxford University Press; 2007.
3. Burgess S, Geddes J, Hawton K, et al. Lithium for maintenance treatment of mood disorders. Cochrane Database Syst Rev. 2001:CD003013.
4. Bowden CL, Calabrese JR, McElroy SL, et al. A randomized, placebo-controlled 12-month trial of divalproex and lithium in treatment of outpatients with bipolar I disorder. Divalproex maintenance study group. Arch Gen Psychiatry. 2000;57(5):481-489.
5. Bowden CL, Calabrese JR, Sachs G, et al; Lamictal 606 Study Group. A placebo-controlled 18-month trial of lamotrigine and lithium maintenance treatment in recently manic or hypomanic patients with bipolar I disorder. Arch Gen Psychiatry. 2003;60(4):392-400.
6. Swann AC, Bowden CL, Calabrese JR, et al. Pattern of response to divalproex, lithium, or placebo in four naturalistic subtypes of mania. Neuropsychopharmacology. 2002;26(4):530-536.
7. 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.
8. Perlis RH, Smoller JW, Ferreira MA, et al. A genomewide association study of response to lithium for prevention of recurrence in bipolar disorder. Am J Psychiatry. 2009; 166(6):718-725.
9. Grof P, Duffy A, Cavazzoni P, et al. Is response to prophylactic lithium a familial trait? J Clin Psychiatry. 2002;63(10): 942-947.
10. Duffy A, Alda M, Kutcher S, et al. A prospective study of the offspring of bipolar parents responsive and nonresponsive to lithium treatment. J Clin Psychiatry. 2002;63(12): 1171-1178.
11. 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.
12. Quiroz JA, Machado-Vieira R, Zarate CA Jr, et al. Novel insights into lithium’s mechanism of action: neurotrophic and neuroprotective effects. Neuropsychobiology. 2010; 62(1):50-60.
13. Forlenza OV, Diniz BS, Radanovic M, et al. Disease-modifying properties of long-term lithium treatment for amnestic mild cognitive impairment: randomised controlled trial. Br J Psychiatry. 2011;198(5):351-356.
14. de Sousa RT, van de Bilt MT, Diniz BS, et al. Lithium increases plasma brain-derived neurotrophic factor in acute bipolar mania: a preliminary 4-week study. Neurosci Lett. 2011;494(1):54-56.
15. Nierenberg AA, Sylvia LG, Leon AC, et al; LiTMUS Study Group. Lithium treatment–moderate dose use study (LiTMUS) for bipolar disorder: rationale and design. Clin Trials. 2009;6(6):637-648.
16. Sylvia LG, Reilly-Harrington NA, Leon AC, et al. Methods to limit attrition in longitudinal comparative effectiveness trials: lessons from the Lithium Treatment - Moderate dose Use Study (LiTMUS) for bipolar disorder. Clin Trials. 2012;9(1):94-101.
17. McCarthy MJ, Leckband SG, Kelsoe JR. Pharmacogenetics of lithium response in bipolar disorder. Pharmacogenomics. 2010;11(10):1439-1465.
18. Can A, Schulze TG, Gould TD. Molecular actions and clinical pharmacogenetics of lithium therapy [published online February 15, 2014]. Pharmacol Biochem Behav. doi: 10.1016/j.pbb.2014.02.004.
19. Berridge MJ. Unlocking the secrets of cell signaling. Annu Rev Physiol. 2005;67:1-21.
20. Devaki R, Shankar Rao S, Nadgir SM. The effect of lithium on the adrenoceptor-mediated second messenger system in the rat brain. J Psychiatry Neurosci. 2006;31(4):246-252.
21. Pan JQ, Lewis MC, Ketterman JK, et al. AKT kinase activity is required for lithium to modulate mood-related behaviors in mice. Neuropsychopharmacology. 2011;36(7):1397-1411.
22. Hu LW, Kawamoto EM, Brietzke E, et al. The role of Wnt signaling and its interaction with diverse mechanisms of cellular apoptosis in the pathophysiology of bipolar disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35(1):11-17.
23. 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 [published online March 25, 2014]. Transl Psychiatry. doi:10.1038/tp.2014.12.
24. Jefferson JW. Lithium. In: Aronson JK, ed. Side effects of drugs annual, volume 26. Amsterdam, The Netherlands: Elsevier Science; 2003:19-29.
25. Baldessarini RJ, Tondo L, Floris G, et al. Effects of rapid cycling on response to lithium maintenance treatment in 360 bipolar I and II disorder patients. J Affect Disord. 2000;61(2):13-22.
26. Baldessarini RJ, Tondo L, Hennen J, et al. Latency and episodes before treatment: response to lithium maintenance in bipolar I and II disorders. Bipolar Disord. 1999;1(2): 91-97.
27. Fieve RR, Kumbaraci T, Dunner DL. Lithium prophylaxis of depression in bipolar I, bipolar II, and unipolar patients. Am J Psychiatry. 1976;133(8):925-929.
28. Peck CC, Pond SM, Becker CE, et al. An evaluation of the effects of lithium in the treatment of chronic alcoholism. II. Assessment of the two-period crossover design. Alcohol Clin Exp Res. 1981;5(2):252-255.
29. Peselow ED, Dunner DL, Fieve RR, et al. Lithium prophylaxis of depression in unipolar, bipolar II, and cyclothymic patients. Am J Psychiatry. 1982;139(6):747-752.
30. Bellino S, Paradiso E, Bogetto F. Efficacy and tolerability of pharmacotherapies for borderline personality disorder. CNS Drugs. 2008;22(8):671-692.
31. Alevizos B, Alevizos E, Leonardou A, et al. Low dosage lithium augmentation in venlafaxine resistant depression: an open-label study. Psychiatrike. 2012;23(2):143-148.
32. Goldberg JF, Sacks MH, Kocsis JH. Low-dose lithium augmentation of divalproex in geriatric mania. J Clin Psychiatry. 2000;61(4):304.
33. Saunders KE, Goodwin GM. New approaches in the treatment of bipolar depression. Curr Top Behav Neurosci. 2013;14:291-307.
34. Forlenza OV, Diniz BS, Radanovic M, et al. Disease-modifying properties of long-term lithium treatment for amnestic mild cognitive impairment: randomised controlled trial. Br J Psychiatry. 2011;198(5):351-356.
35. Young RC, Biggs JT, Ziegler VE, et al. A rating scale for mania: reliability, validity and sensitivity. Br J Psychiatry. 1978;133:429-435.
36. Trivedi MH, Rush AJ, Ibrahim HM, et al. The Inventory of Depressive Symptomatology, Clinician Rating (IDS-C) and Self-Report (IDS-SR), and the Quick Inventory of Depressive Symptomatology, Clinician Rating (QIDS-C) and Self-Report (QIDS-SR) in public sector patients with mood disorders: a psychometric evaluation. Psychol Med. 2004;34(1):73-82.
37. Kroenke K, Spitzer RL, Williams JB. The PHQ-9: validity of a brief depression severity measure. J Gen Intern Med. 2001;16(9):606-613.
38. Altman EG, Hedeker D, Peterson JL, et al. The Altman Self- Rating Mania Scale. Biol Psychiatry. 1997;42(10):948-955.
39. Machado-Vieira R, Luckenbaugh DA, Soeiro-de-Souza MG, et al. Early improvement with lithium in classic mania and its association with later response. J Affect Disord. 2013;144(1-2):160-164.
40. Severus WE, Lipkovich IA, Licht RW, et al. In search of optimal lithium levels and olanzapine doses in the long-term treatment of bipolar I disorder. A post-hoc analysis of the maintenance study by Tohen et al. 2005. Eur Psychiatry. 2010;25(8):443-449.
41. Vestergaard P, Licht RW, Brodersen A, et al. Outcome of lithium prophylaxis: a prospective follow-up of affective disorder patients assigned to high and low serum lithium levels. Acta Psychiatr Scand. 1998;98(4):310-315.
42. Nierenberg AA, Friedman ES, Bowden CL, et al. Lithium treatment moderate-dose use study (LiTMUS) for bipolar disorder: a randomized comparative effectiveness trial of optimized personalized treatment with and without lithium. Am J Psychiatry. 2013;170(1):102-111.

Issue
Current Psychiatry - 13(6)
Issue
Current Psychiatry - 13(6)
Page Number
38-44
Page Number
38-44
Publications
Publications
Topics
Article Type
Display Headline
Lithium for bipolar disorder: A re-emerging treatment for mood instability
Display Headline
Lithium for bipolar disorder: A re-emerging treatment for mood instability
Legacy Keywords
lithium, bipolar disorder, lithium dosages, depression, mood disorders, mood instability
Legacy Keywords
lithium, bipolar disorder, lithium dosages, depression, mood disorders, mood instability
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

BPD and the broader landscape of neuropsychiatric illness

Article Type
Changed
Mon, 04/16/2018 - 14:33
Display Headline
BPD and the broader landscape of neuropsychiatric illness

Dr. Henry A. Nasrallah’s recent Editorial on borderline personality disorder (BPD) (Current Psychiatry, From the Editor, April 2014, p. 19-20, 32 [http://bit.ly/1e8yAwE]) describes BPD as a heritable brain disease. I have been arguing this point for many years, often finding support from my colleague, Hagop Akiskal, MD, and opposition from my psychoanalytic colleagues.

In recent papers1,2 on brain changes in BPD and the connection between BPD and bipolar disorders, I wrote that there often is a heritable aspect to the condition. There are exceptions to such heritability, as in the setting of a horrific environment (eg, father-daughter incest, parental brutality), where the same symptoms seen in BPD develop primarily from post-natal influences. Dr. Akiskal and I were discussing this a long time back, before MRI. Now I feel vindicated, with generous help from someone of Dr. Nasrallah’s prestige and influence.

There also is electrophysiological (including evoked potential) evi­dence for neural pathology in BPD, as well as data derived from single photon emission CT scanning. The burgeoning literature on MRI and functional MRI studies of BPD is in good agreement about the brain changes most relevant to BPD and that are found with regularity in this condition.

Particularly when BPD is diag­nosed in people (usually women) who do not have a history of neglect, sexual molestation, parental humili­ation or cruelty, or head injury, what else is there, if not genetically pre­disposed alterations in the fronto­limbic structures (and maybe the periaqueductal gray) that underlie the so-called “personality disorder,” and, not surprisingly, bipolar dis­orders, especially bipolar II disor­der, which often is the other side of the coin as BPD, and amenable to the same combination of medication and psychotherapy?

Michael H. Stone, MD
Professor of Clinical Psychiatry Columbia College of Physicians and Surgeons
New York, New York

----------------------------------------------------------------------------------------------------------

As a psychiatrist/psychoanalyst who works with BPD patients, I read Dr. Nasrallah’s April 2014 Editorial with great interest and enthusiasm. Over the past 10 years, I have been impressed with the number of patients with BPD whose nonverbal learning disorders and auditory and visual pro­cessing disorders have gone undiag­nosed. Recently, I lectured on this topic to the staff of a school for children with a range of neuropsychiatric disorders; the staff found my observations about such comorbidity consistent with their observations. These dysfunctions, or neurological variations—unknown to the parent and the child—interfere with early object-relation formation, attachment capacity, and learning. Neuropsychiatry and psychological development are, in fact, part of the same system.

An example: For 12 years, I have been treating a patient who has audi­tory processing and working memory problems, meaning that she could not process the connections among different ideas. This difficulty frus­trated her parents, who, in their frus­tration, criticized her for not paying attention. She was labeled “bad” and assumed the role of the “black sheep” in her family. Although she was intelligent, she was often wrong in her judgments and choices, and easily frustrated. In therapy, as I realized what part of her prob­lem was, I changed my technique.

When my patient asked me to tell her the sequence of understandings that we had just put together, I invited her to take my pad and write down her sense of it. As she described each part of that sequence to me, we would discuss it and I would remind her of lost fragments. Gradually, she learned to put ideas together; however, I also watched her struggle to hold these ideas in working memory and to use them.

Over time, she has improved and is more functional. After several years of dis­ability, she returned to work, although she still struggles interpersonally.

With many of such patients, I have had to modify traditional techniques of psy­chotherapy. I am fascinated by, and enjoy, such intensive psychotherapy. I am also amazed to see the impact of previously unknown neuropathologic variations on development. The more I learn about the impact of neuropsychiatry on psychologi­cal development, the more I can help my patients.

Howard Wishnie, MD
Cambridge, Massachusetts

Dr. Nasrallah responds
I appreciate Dr. Stone’s kind words and concur­rence with my thinking about BPD. It would have been appropriate to include discussion of neuro­physiological findings in my Editorial, but I opted to use my limited space to focus on structural and functional neuroimaging and genetics.

Henry A. Nasrallah, MD
Professor and Chairman Department of Neurology & Psychiatry
Saint Louis University School of Medicine
St. Louis, Missouri

References


1. Stone MH. The spectrum of borderline personality disorder: a neurophysiological view. Neuropsychiatric Electrophysiology. In press.
2. Stone MH. A new look at borderline personality disorder and related disorders: hyper-activity in the limbic system and lower centers. Psychodyn Psychiatry. 2013;41(3):437-466.

Article PDF
Author and Disclosure Information

Issue
Current Psychiatry - 13(6)
Publications
Topics
Page Number
37, 53
Legacy Keywords
letters to the editor, borderline personality disorder, neuropsychiatric illness, bipolar disorder
Sections
Author and Disclosure Information

Author and Disclosure Information

Article PDF
Article PDF
Related Articles

Dr. Henry A. Nasrallah’s recent Editorial on borderline personality disorder (BPD) (Current Psychiatry, From the Editor, April 2014, p. 19-20, 32 [http://bit.ly/1e8yAwE]) describes BPD as a heritable brain disease. I have been arguing this point for many years, often finding support from my colleague, Hagop Akiskal, MD, and opposition from my psychoanalytic colleagues.

In recent papers1,2 on brain changes in BPD and the connection between BPD and bipolar disorders, I wrote that there often is a heritable aspect to the condition. There are exceptions to such heritability, as in the setting of a horrific environment (eg, father-daughter incest, parental brutality), where the same symptoms seen in BPD develop primarily from post-natal influences. Dr. Akiskal and I were discussing this a long time back, before MRI. Now I feel vindicated, with generous help from someone of Dr. Nasrallah’s prestige and influence.

There also is electrophysiological (including evoked potential) evi­dence for neural pathology in BPD, as well as data derived from single photon emission CT scanning. The burgeoning literature on MRI and functional MRI studies of BPD is in good agreement about the brain changes most relevant to BPD and that are found with regularity in this condition.

Particularly when BPD is diag­nosed in people (usually women) who do not have a history of neglect, sexual molestation, parental humili­ation or cruelty, or head injury, what else is there, if not genetically pre­disposed alterations in the fronto­limbic structures (and maybe the periaqueductal gray) that underlie the so-called “personality disorder,” and, not surprisingly, bipolar dis­orders, especially bipolar II disor­der, which often is the other side of the coin as BPD, and amenable to the same combination of medication and psychotherapy?

Michael H. Stone, MD
Professor of Clinical Psychiatry Columbia College of Physicians and Surgeons
New York, New York

----------------------------------------------------------------------------------------------------------

As a psychiatrist/psychoanalyst who works with BPD patients, I read Dr. Nasrallah’s April 2014 Editorial with great interest and enthusiasm. Over the past 10 years, I have been impressed with the number of patients with BPD whose nonverbal learning disorders and auditory and visual pro­cessing disorders have gone undiag­nosed. Recently, I lectured on this topic to the staff of a school for children with a range of neuropsychiatric disorders; the staff found my observations about such comorbidity consistent with their observations. These dysfunctions, or neurological variations—unknown to the parent and the child—interfere with early object-relation formation, attachment capacity, and learning. Neuropsychiatry and psychological development are, in fact, part of the same system.

An example: For 12 years, I have been treating a patient who has audi­tory processing and working memory problems, meaning that she could not process the connections among different ideas. This difficulty frus­trated her parents, who, in their frus­tration, criticized her for not paying attention. She was labeled “bad” and assumed the role of the “black sheep” in her family. Although she was intelligent, she was often wrong in her judgments and choices, and easily frustrated. In therapy, as I realized what part of her prob­lem was, I changed my technique.

When my patient asked me to tell her the sequence of understandings that we had just put together, I invited her to take my pad and write down her sense of it. As she described each part of that sequence to me, we would discuss it and I would remind her of lost fragments. Gradually, she learned to put ideas together; however, I also watched her struggle to hold these ideas in working memory and to use them.

Over time, she has improved and is more functional. After several years of dis­ability, she returned to work, although she still struggles interpersonally.

With many of such patients, I have had to modify traditional techniques of psy­chotherapy. I am fascinated by, and enjoy, such intensive psychotherapy. I am also amazed to see the impact of previously unknown neuropathologic variations on development. The more I learn about the impact of neuropsychiatry on psychologi­cal development, the more I can help my patients.

Howard Wishnie, MD
Cambridge, Massachusetts

Dr. Nasrallah responds
I appreciate Dr. Stone’s kind words and concur­rence with my thinking about BPD. It would have been appropriate to include discussion of neuro­physiological findings in my Editorial, but I opted to use my limited space to focus on structural and functional neuroimaging and genetics.

Henry A. Nasrallah, MD
Professor and Chairman Department of Neurology & Psychiatry
Saint Louis University School of Medicine
St. Louis, Missouri

Dr. Henry A. Nasrallah’s recent Editorial on borderline personality disorder (BPD) (Current Psychiatry, From the Editor, April 2014, p. 19-20, 32 [http://bit.ly/1e8yAwE]) describes BPD as a heritable brain disease. I have been arguing this point for many years, often finding support from my colleague, Hagop Akiskal, MD, and opposition from my psychoanalytic colleagues.

In recent papers1,2 on brain changes in BPD and the connection between BPD and bipolar disorders, I wrote that there often is a heritable aspect to the condition. There are exceptions to such heritability, as in the setting of a horrific environment (eg, father-daughter incest, parental brutality), where the same symptoms seen in BPD develop primarily from post-natal influences. Dr. Akiskal and I were discussing this a long time back, before MRI. Now I feel vindicated, with generous help from someone of Dr. Nasrallah’s prestige and influence.

There also is electrophysiological (including evoked potential) evi­dence for neural pathology in BPD, as well as data derived from single photon emission CT scanning. The burgeoning literature on MRI and functional MRI studies of BPD is in good agreement about the brain changes most relevant to BPD and that are found with regularity in this condition.

Particularly when BPD is diag­nosed in people (usually women) who do not have a history of neglect, sexual molestation, parental humili­ation or cruelty, or head injury, what else is there, if not genetically pre­disposed alterations in the fronto­limbic structures (and maybe the periaqueductal gray) that underlie the so-called “personality disorder,” and, not surprisingly, bipolar dis­orders, especially bipolar II disor­der, which often is the other side of the coin as BPD, and amenable to the same combination of medication and psychotherapy?

Michael H. Stone, MD
Professor of Clinical Psychiatry Columbia College of Physicians and Surgeons
New York, New York

----------------------------------------------------------------------------------------------------------

As a psychiatrist/psychoanalyst who works with BPD patients, I read Dr. Nasrallah’s April 2014 Editorial with great interest and enthusiasm. Over the past 10 years, I have been impressed with the number of patients with BPD whose nonverbal learning disorders and auditory and visual pro­cessing disorders have gone undiag­nosed. Recently, I lectured on this topic to the staff of a school for children with a range of neuropsychiatric disorders; the staff found my observations about such comorbidity consistent with their observations. These dysfunctions, or neurological variations—unknown to the parent and the child—interfere with early object-relation formation, attachment capacity, and learning. Neuropsychiatry and psychological development are, in fact, part of the same system.

An example: For 12 years, I have been treating a patient who has audi­tory processing and working memory problems, meaning that she could not process the connections among different ideas. This difficulty frus­trated her parents, who, in their frus­tration, criticized her for not paying attention. She was labeled “bad” and assumed the role of the “black sheep” in her family. Although she was intelligent, she was often wrong in her judgments and choices, and easily frustrated. In therapy, as I realized what part of her prob­lem was, I changed my technique.

When my patient asked me to tell her the sequence of understandings that we had just put together, I invited her to take my pad and write down her sense of it. As she described each part of that sequence to me, we would discuss it and I would remind her of lost fragments. Gradually, she learned to put ideas together; however, I also watched her struggle to hold these ideas in working memory and to use them.

Over time, she has improved and is more functional. After several years of dis­ability, she returned to work, although she still struggles interpersonally.

With many of such patients, I have had to modify traditional techniques of psy­chotherapy. I am fascinated by, and enjoy, such intensive psychotherapy. I am also amazed to see the impact of previously unknown neuropathologic variations on development. The more I learn about the impact of neuropsychiatry on psychologi­cal development, the more I can help my patients.

Howard Wishnie, MD
Cambridge, Massachusetts

Dr. Nasrallah responds
I appreciate Dr. Stone’s kind words and concur­rence with my thinking about BPD. It would have been appropriate to include discussion of neuro­physiological findings in my Editorial, but I opted to use my limited space to focus on structural and functional neuroimaging and genetics.

Henry A. Nasrallah, MD
Professor and Chairman Department of Neurology & Psychiatry
Saint Louis University School of Medicine
St. Louis, Missouri

References


1. Stone MH. The spectrum of borderline personality disorder: a neurophysiological view. Neuropsychiatric Electrophysiology. In press.
2. Stone MH. A new look at borderline personality disorder and related disorders: hyper-activity in the limbic system and lower centers. Psychodyn Psychiatry. 2013;41(3):437-466.

References


1. Stone MH. The spectrum of borderline personality disorder: a neurophysiological view. Neuropsychiatric Electrophysiology. In press.
2. Stone MH. A new look at borderline personality disorder and related disorders: hyper-activity in the limbic system and lower centers. Psychodyn Psychiatry. 2013;41(3):437-466.

Issue
Current Psychiatry - 13(6)
Issue
Current Psychiatry - 13(6)
Page Number
37, 53
Page Number
37, 53
Publications
Publications
Topics
Article Type
Display Headline
BPD and the broader landscape of neuropsychiatric illness
Display Headline
BPD and the broader landscape of neuropsychiatric illness
Legacy Keywords
letters to the editor, borderline personality disorder, neuropsychiatric illness, bipolar disorder
Legacy Keywords
letters to the editor, borderline personality disorder, neuropsychiatric illness, bipolar disorder
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media