User login
Four genetic markers associated with five psychiatric disorders
Genetic variants at four chromosomal positions have now been linked to five diverse childhood- and adult-onset psychiatric illnesses: schizophrenia, autism spectrum disorders, attention-deficit/hyperactivity disorder, bipolar disorder, and major depressive disorder.
Two of the four loci encode for transmembranal calcium ion transport, a physiologic finding that could become a treatment target, Dr. Jordan W. Smoller and his colleagues wrote in the Feb. 28 online issue of the Lancet (2013 Feb. 28 [http://dx.doi.org/10.1016/S0140-6736(12)62129-1]).
"The finding that genetic variants have cross-disorder effects is an empirical step toward helping clinicians understand the common co-occurrence of clinical phenotypes in individual patients," wrote Dr. Smoller of the Massachusetts General Hospital, Boston, and his coauthors. "Our results implicate a specific biological pathway – voltage-gated calcium-channel signaling – as a contributor to the pathogenesis of several psychiatric disorders, and support the potential of this pathway as a therapeutic target for psychiatric disease."
The findings also could further the goal of "moving beyond descriptive syndromes in psychiatry and towards a nosology informed by disease cause," the investigators noted.
The genome-wide association study – the largest yet of its kind – analyzed single nucleotide polymorphisms (SNPs) for the five disorders among 33,332 cases and 27,888 controls, all of European ancestry. Four independent regions contained SNPs that were significantly related to the disorders. Three were related to all five disorders, and one to only bipolar disorder and schizophrenia.
The analysis also identified additional cross-disorder associations with a number of loci previously identified only with schizophrenia, although these associations were not as strong as those seen with the four primary regions.
Two of the four loci are related to voltage-gated transmembranal calcium channels, the authors noted. One of these has been previously identified as a risk gene for schizophrenia, bipolar disorder, and major depressive disorder. The gene facilitates the passage of calcium ions into the plasma membrane.
"Thus, our results suggest that voltage-gated calcium signaling, and, more broadly, calcium channel activity, could be an important biological process in psychiatric disorders," Dr. Smoller and his colleagues wrote. "Alterations in calcium channel signaling could represent a fundamental mechanism contributing to a broad vulnerability to psychopathology."
The authors cited several possible limitations. For example, diagnostic misclassification in cases of schizophrenia and bipolar disorder could produce "spurious evidence of genetic overlap between disorders." In addition, because their analyses were restricted to people of European ancestry, it is unclear whether their findings would apply to other populations.
Nevertheless, they said these result could provide "insights into the shared causation of psychiatric disorders."
Dr. Smoller and his coauthors are members of the Cross-Disorder Group of the Psychiatric Genomics Consortium. Their work was sponsored by the National Institutes of Health; none of the authors had any financial disclosures.
While the genetic findings will contribute to a more logical classification system of psychotropic disorders, the physiologic associations hold out a tantalizing promise for the future, Dr. Alessandro Serretti, lead author, wrote in an accompanying editorial.
"In addition to methodological issues, which are pertinent to researchers, genetic studies should provide translational value for clinicians," wrote Dr. Serretti of the University of Bologna, Italy. "With this perspective, the present study might contribute to future nosographic systems, which could be based not only on statistically determined clinical categories, but also on biological pathogenic factors that are pivotal to the identification of suitable treatments."
Calcium signaling is a critical regulator of neuronal growth and development, he said. Prior studies have confirmed the antidepressant effects of voltage-dependent calcium channel agonists in mice; a mutation that blocks such channels would be a logical underpinning for an inherited prediction toward depression and, perhaps, other disorders.
"[Single nucleotide polymorphisms] associated previously with different psychiatric disorders identified convergence of pathways in synaptogenesis, axonal guidance, and synaptic plasticity, and now calcium signaling, which is pivotal in the mechanisms of all these biological processes."
Not all patients with these genetic markers will develop one of the associated disorders. "We agree about the presence of some transdiagnostic risk factors, but many genes and polymorphisms are expected to confer a liability to individual psychiatric diseases." Prenatal and postnatal environments – both their negative and positive environmental conditions – modulate the expression of any genetic predilection.
But genetic predilections present an invaluable look into both the development and treatment of disease. "We therefore believe that genetics, possibly thanks to more comprehensive phenotype and endophenotype assessments, can contribute to prediction and prevention of psychiatric diseases, along with the identification of molecular targets for new generations of psychotropic drugs."
Dr. Serretti is a neuropsychiatrist at the University of Bologna, Italy. He had no financial disclosures.
While the genetic findings will contribute to a more logical classification system of psychotropic disorders, the physiologic associations hold out a tantalizing promise for the future, Dr. Alessandro Serretti, lead author, wrote in an accompanying editorial.
"In addition to methodological issues, which are pertinent to researchers, genetic studies should provide translational value for clinicians," wrote Dr. Serretti of the University of Bologna, Italy. "With this perspective, the present study might contribute to future nosographic systems, which could be based not only on statistically determined clinical categories, but also on biological pathogenic factors that are pivotal to the identification of suitable treatments."
Calcium signaling is a critical regulator of neuronal growth and development, he said. Prior studies have confirmed the antidepressant effects of voltage-dependent calcium channel agonists in mice; a mutation that blocks such channels would be a logical underpinning for an inherited prediction toward depression and, perhaps, other disorders.
"[Single nucleotide polymorphisms] associated previously with different psychiatric disorders identified convergence of pathways in synaptogenesis, axonal guidance, and synaptic plasticity, and now calcium signaling, which is pivotal in the mechanisms of all these biological processes."
Not all patients with these genetic markers will develop one of the associated disorders. "We agree about the presence of some transdiagnostic risk factors, but many genes and polymorphisms are expected to confer a liability to individual psychiatric diseases." Prenatal and postnatal environments – both their negative and positive environmental conditions – modulate the expression of any genetic predilection.
But genetic predilections present an invaluable look into both the development and treatment of disease. "We therefore believe that genetics, possibly thanks to more comprehensive phenotype and endophenotype assessments, can contribute to prediction and prevention of psychiatric diseases, along with the identification of molecular targets for new generations of psychotropic drugs."
Dr. Serretti is a neuropsychiatrist at the University of Bologna, Italy. He had no financial disclosures.
While the genetic findings will contribute to a more logical classification system of psychotropic disorders, the physiologic associations hold out a tantalizing promise for the future, Dr. Alessandro Serretti, lead author, wrote in an accompanying editorial.
"In addition to methodological issues, which are pertinent to researchers, genetic studies should provide translational value for clinicians," wrote Dr. Serretti of the University of Bologna, Italy. "With this perspective, the present study might contribute to future nosographic systems, which could be based not only on statistically determined clinical categories, but also on biological pathogenic factors that are pivotal to the identification of suitable treatments."
Calcium signaling is a critical regulator of neuronal growth and development, he said. Prior studies have confirmed the antidepressant effects of voltage-dependent calcium channel agonists in mice; a mutation that blocks such channels would be a logical underpinning for an inherited prediction toward depression and, perhaps, other disorders.
"[Single nucleotide polymorphisms] associated previously with different psychiatric disorders identified convergence of pathways in synaptogenesis, axonal guidance, and synaptic plasticity, and now calcium signaling, which is pivotal in the mechanisms of all these biological processes."
Not all patients with these genetic markers will develop one of the associated disorders. "We agree about the presence of some transdiagnostic risk factors, but many genes and polymorphisms are expected to confer a liability to individual psychiatric diseases." Prenatal and postnatal environments – both their negative and positive environmental conditions – modulate the expression of any genetic predilection.
But genetic predilections present an invaluable look into both the development and treatment of disease. "We therefore believe that genetics, possibly thanks to more comprehensive phenotype and endophenotype assessments, can contribute to prediction and prevention of psychiatric diseases, along with the identification of molecular targets for new generations of psychotropic drugs."
Dr. Serretti is a neuropsychiatrist at the University of Bologna, Italy. He had no financial disclosures.
Genetic variants at four chromosomal positions have now been linked to five diverse childhood- and adult-onset psychiatric illnesses: schizophrenia, autism spectrum disorders, attention-deficit/hyperactivity disorder, bipolar disorder, and major depressive disorder.
Two of the four loci encode for transmembranal calcium ion transport, a physiologic finding that could become a treatment target, Dr. Jordan W. Smoller and his colleagues wrote in the Feb. 28 online issue of the Lancet (2013 Feb. 28 [http://dx.doi.org/10.1016/S0140-6736(12)62129-1]).
"The finding that genetic variants have cross-disorder effects is an empirical step toward helping clinicians understand the common co-occurrence of clinical phenotypes in individual patients," wrote Dr. Smoller of the Massachusetts General Hospital, Boston, and his coauthors. "Our results implicate a specific biological pathway – voltage-gated calcium-channel signaling – as a contributor to the pathogenesis of several psychiatric disorders, and support the potential of this pathway as a therapeutic target for psychiatric disease."
The findings also could further the goal of "moving beyond descriptive syndromes in psychiatry and towards a nosology informed by disease cause," the investigators noted.
The genome-wide association study – the largest yet of its kind – analyzed single nucleotide polymorphisms (SNPs) for the five disorders among 33,332 cases and 27,888 controls, all of European ancestry. Four independent regions contained SNPs that were significantly related to the disorders. Three were related to all five disorders, and one to only bipolar disorder and schizophrenia.
The analysis also identified additional cross-disorder associations with a number of loci previously identified only with schizophrenia, although these associations were not as strong as those seen with the four primary regions.
Two of the four loci are related to voltage-gated transmembranal calcium channels, the authors noted. One of these has been previously identified as a risk gene for schizophrenia, bipolar disorder, and major depressive disorder. The gene facilitates the passage of calcium ions into the plasma membrane.
"Thus, our results suggest that voltage-gated calcium signaling, and, more broadly, calcium channel activity, could be an important biological process in psychiatric disorders," Dr. Smoller and his colleagues wrote. "Alterations in calcium channel signaling could represent a fundamental mechanism contributing to a broad vulnerability to psychopathology."
The authors cited several possible limitations. For example, diagnostic misclassification in cases of schizophrenia and bipolar disorder could produce "spurious evidence of genetic overlap between disorders." In addition, because their analyses were restricted to people of European ancestry, it is unclear whether their findings would apply to other populations.
Nevertheless, they said these result could provide "insights into the shared causation of psychiatric disorders."
Dr. Smoller and his coauthors are members of the Cross-Disorder Group of the Psychiatric Genomics Consortium. Their work was sponsored by the National Institutes of Health; none of the authors had any financial disclosures.
Genetic variants at four chromosomal positions have now been linked to five diverse childhood- and adult-onset psychiatric illnesses: schizophrenia, autism spectrum disorders, attention-deficit/hyperactivity disorder, bipolar disorder, and major depressive disorder.
Two of the four loci encode for transmembranal calcium ion transport, a physiologic finding that could become a treatment target, Dr. Jordan W. Smoller and his colleagues wrote in the Feb. 28 online issue of the Lancet (2013 Feb. 28 [http://dx.doi.org/10.1016/S0140-6736(12)62129-1]).
"The finding that genetic variants have cross-disorder effects is an empirical step toward helping clinicians understand the common co-occurrence of clinical phenotypes in individual patients," wrote Dr. Smoller of the Massachusetts General Hospital, Boston, and his coauthors. "Our results implicate a specific biological pathway – voltage-gated calcium-channel signaling – as a contributor to the pathogenesis of several psychiatric disorders, and support the potential of this pathway as a therapeutic target for psychiatric disease."
The findings also could further the goal of "moving beyond descriptive syndromes in psychiatry and towards a nosology informed by disease cause," the investigators noted.
The genome-wide association study – the largest yet of its kind – analyzed single nucleotide polymorphisms (SNPs) for the five disorders among 33,332 cases and 27,888 controls, all of European ancestry. Four independent regions contained SNPs that were significantly related to the disorders. Three were related to all five disorders, and one to only bipolar disorder and schizophrenia.
The analysis also identified additional cross-disorder associations with a number of loci previously identified only with schizophrenia, although these associations were not as strong as those seen with the four primary regions.
Two of the four loci are related to voltage-gated transmembranal calcium channels, the authors noted. One of these has been previously identified as a risk gene for schizophrenia, bipolar disorder, and major depressive disorder. The gene facilitates the passage of calcium ions into the plasma membrane.
"Thus, our results suggest that voltage-gated calcium signaling, and, more broadly, calcium channel activity, could be an important biological process in psychiatric disorders," Dr. Smoller and his colleagues wrote. "Alterations in calcium channel signaling could represent a fundamental mechanism contributing to a broad vulnerability to psychopathology."
The authors cited several possible limitations. For example, diagnostic misclassification in cases of schizophrenia and bipolar disorder could produce "spurious evidence of genetic overlap between disorders." In addition, because their analyses were restricted to people of European ancestry, it is unclear whether their findings would apply to other populations.
Nevertheless, they said these result could provide "insights into the shared causation of psychiatric disorders."
Dr. Smoller and his coauthors are members of the Cross-Disorder Group of the Psychiatric Genomics Consortium. Their work was sponsored by the National Institutes of Health; none of the authors had any financial disclosures.
FROM THE LANCET
Major Finding: Polymorphisms on four separate chromosomal locations are significantly associated with five separate psychiatric illnesses: schizophrenia, attention-deficit/hyperactivity disorder, major depressive disorder, autism spectrum disorders, and bipolar disorder.
Data Source: The genome-wide association study comprised 33,332 cases and 27,888 controls, all of European ancestry.
Disclosures: The work was sponsored by the National Institutes of Health; none of the authors had any financial disclosures.
Bipolar disorder and schizophrenia probably not the same disease
LAS VEGAS – Differences in drug responses probably offer the strongest evidence that bipolar disorder and schizophrenia may be separate entities, according to Dr. Charles B. Nemeroff.
The notion that the two are different manifestations of the same disease has gained traction in some quarters because, at least superficially, the two illnesses have quite a bit in common, including age of onset, response to atypical antipsychotics, ventricular enlargement, and shared genetic characteristics.
Early childhood abuse and neglect can increase the risk of both in vulnerable people, as well, and bipolar disorder patients can have auditory hallucinations and paranoid delusions, just as those with schizophrenia do, Dr. Nemeroff said at the Nevada Psychiatric Association’s annual psychopharmacology update.
Indeed, the possibility that they are even on the same spectrum has "implications in terms of current treatment and treatment development," but Dr. Nemeroff said he has his doubts. "The last chapter to this debate has not yet been written," he said.
That both respond to atypicals could be a red herring, for instance. "Imipramine is effective for treating both depression and enuresis, but I don’t think they’re the same thing," said Dr. Nemeroff, Leonard M. Miller Professor and chairman of the department of psychiatry and behavioral sciences at the University of Miami.
Regarding genes, some indeed are associated with both schizophrenia and bipolar disorder, but others appear to increase risk for one but not the other. Overall, genes account for about two-thirds of the risk for bipolar disorder, but only about half the risk for schizophrenia, he said (Neurosci. Biobehav. Rev. 2012;36:556-71).
Bipolar patients also do not lose brain volume over time, unlike patients with schizophrenia, and they often report an early onset of depression, sometimes before puberty. "We often don’t hear that in patients with schizophrenia, even when you talk to family members. There may have been something [odd] going on in those early years, but it doesn’t look like early-onset depression," he said.
Atypicals aside, differences in drug responses probably offer the strongest evidence that the two illnesses are separate entities.
Although of great help in manic patients, anticonvulsants "have no efficacy whatsoever in schizophrenia." Likewise, antidepressants can "rocket" bipolar patients into mania, but "have you ever seen a schizophrenic patient given an antidepressant become acutely manic? I haven’t," Dr. Nemeroff said.
Perhaps the response to lithium is the most telling of all. It remains one of the most effective treatments for bipolar disorder but "just does not work" for schizophrenia, according to numerous trials as both monotherapy and add-on therapy, he said.
"I’ve seen dozens of [bipolar] patients" struggle for years despite treatment with newer agents, but who have never had a lithium trial. When it’s finally tried, they often have "phenomenal response[s]. They became euthymic and remain euthymic for years, and never suffer an episode. I’ve rarely ever seen that with any other treatment for bipolar disorder. [Lithium is] a great drug; it’s also cheap," he said. "Cheap is good for our patients."
Lithium is underused because "nobody’s marketing it to you," he said.
Ongoing monitoring for kidney, thyroid, and other side effects probably puts some psychiatrists off, as well, but it’s really not any more complicated than the ongoing monitoring needed with atypicals and anticonvulsants, he said.
Plus, "there’s pretty good data that if you check TSH [thyroid-stimulating hormone] every 6 months, you’ll be able to catch any incipient hyperthyroidism. Patients who have [thyroid] antibodies at baseline are generally the ones who go on to develop thyroid problems, so you can check that at baseline," he said.
For patients with refractory bipolar depression, "my favorite combination has been lamotrigine and MAO inhibitors," particularly tranylcypromine. "I’ve probably gotten more people [out of] bipolar depression with that – after they’ve been marinated in everything else – than any other strategy besides [electroconvulsive therapy]."
And "lamotrigine doesn’t work in patients with schizophrenia," he noted.
Dr. Nemeroff reported stock ownership, consultant fees, or other income from Allergan, Lilly, Shire, Roche, NovaDel Pharma, BioPharma, AstraZeneca, and other companies.
LAS VEGAS – Differences in drug responses probably offer the strongest evidence that bipolar disorder and schizophrenia may be separate entities, according to Dr. Charles B. Nemeroff.
The notion that the two are different manifestations of the same disease has gained traction in some quarters because, at least superficially, the two illnesses have quite a bit in common, including age of onset, response to atypical antipsychotics, ventricular enlargement, and shared genetic characteristics.
Early childhood abuse and neglect can increase the risk of both in vulnerable people, as well, and bipolar disorder patients can have auditory hallucinations and paranoid delusions, just as those with schizophrenia do, Dr. Nemeroff said at the Nevada Psychiatric Association’s annual psychopharmacology update.
Indeed, the possibility that they are even on the same spectrum has "implications in terms of current treatment and treatment development," but Dr. Nemeroff said he has his doubts. "The last chapter to this debate has not yet been written," he said.
That both respond to atypicals could be a red herring, for instance. "Imipramine is effective for treating both depression and enuresis, but I don’t think they’re the same thing," said Dr. Nemeroff, Leonard M. Miller Professor and chairman of the department of psychiatry and behavioral sciences at the University of Miami.
Regarding genes, some indeed are associated with both schizophrenia and bipolar disorder, but others appear to increase risk for one but not the other. Overall, genes account for about two-thirds of the risk for bipolar disorder, but only about half the risk for schizophrenia, he said (Neurosci. Biobehav. Rev. 2012;36:556-71).
Bipolar patients also do not lose brain volume over time, unlike patients with schizophrenia, and they often report an early onset of depression, sometimes before puberty. "We often don’t hear that in patients with schizophrenia, even when you talk to family members. There may have been something [odd] going on in those early years, but it doesn’t look like early-onset depression," he said.
Atypicals aside, differences in drug responses probably offer the strongest evidence that the two illnesses are separate entities.
Although of great help in manic patients, anticonvulsants "have no efficacy whatsoever in schizophrenia." Likewise, antidepressants can "rocket" bipolar patients into mania, but "have you ever seen a schizophrenic patient given an antidepressant become acutely manic? I haven’t," Dr. Nemeroff said.
Perhaps the response to lithium is the most telling of all. It remains one of the most effective treatments for bipolar disorder but "just does not work" for schizophrenia, according to numerous trials as both monotherapy and add-on therapy, he said.
"I’ve seen dozens of [bipolar] patients" struggle for years despite treatment with newer agents, but who have never had a lithium trial. When it’s finally tried, they often have "phenomenal response[s]. They became euthymic and remain euthymic for years, and never suffer an episode. I’ve rarely ever seen that with any other treatment for bipolar disorder. [Lithium is] a great drug; it’s also cheap," he said. "Cheap is good for our patients."
Lithium is underused because "nobody’s marketing it to you," he said.
Ongoing monitoring for kidney, thyroid, and other side effects probably puts some psychiatrists off, as well, but it’s really not any more complicated than the ongoing monitoring needed with atypicals and anticonvulsants, he said.
Plus, "there’s pretty good data that if you check TSH [thyroid-stimulating hormone] every 6 months, you’ll be able to catch any incipient hyperthyroidism. Patients who have [thyroid] antibodies at baseline are generally the ones who go on to develop thyroid problems, so you can check that at baseline," he said.
For patients with refractory bipolar depression, "my favorite combination has been lamotrigine and MAO inhibitors," particularly tranylcypromine. "I’ve probably gotten more people [out of] bipolar depression with that – after they’ve been marinated in everything else – than any other strategy besides [electroconvulsive therapy]."
And "lamotrigine doesn’t work in patients with schizophrenia," he noted.
Dr. Nemeroff reported stock ownership, consultant fees, or other income from Allergan, Lilly, Shire, Roche, NovaDel Pharma, BioPharma, AstraZeneca, and other companies.
LAS VEGAS – Differences in drug responses probably offer the strongest evidence that bipolar disorder and schizophrenia may be separate entities, according to Dr. Charles B. Nemeroff.
The notion that the two are different manifestations of the same disease has gained traction in some quarters because, at least superficially, the two illnesses have quite a bit in common, including age of onset, response to atypical antipsychotics, ventricular enlargement, and shared genetic characteristics.
Early childhood abuse and neglect can increase the risk of both in vulnerable people, as well, and bipolar disorder patients can have auditory hallucinations and paranoid delusions, just as those with schizophrenia do, Dr. Nemeroff said at the Nevada Psychiatric Association’s annual psychopharmacology update.
Indeed, the possibility that they are even on the same spectrum has "implications in terms of current treatment and treatment development," but Dr. Nemeroff said he has his doubts. "The last chapter to this debate has not yet been written," he said.
That both respond to atypicals could be a red herring, for instance. "Imipramine is effective for treating both depression and enuresis, but I don’t think they’re the same thing," said Dr. Nemeroff, Leonard M. Miller Professor and chairman of the department of psychiatry and behavioral sciences at the University of Miami.
Regarding genes, some indeed are associated with both schizophrenia and bipolar disorder, but others appear to increase risk for one but not the other. Overall, genes account for about two-thirds of the risk for bipolar disorder, but only about half the risk for schizophrenia, he said (Neurosci. Biobehav. Rev. 2012;36:556-71).
Bipolar patients also do not lose brain volume over time, unlike patients with schizophrenia, and they often report an early onset of depression, sometimes before puberty. "We often don’t hear that in patients with schizophrenia, even when you talk to family members. There may have been something [odd] going on in those early years, but it doesn’t look like early-onset depression," he said.
Atypicals aside, differences in drug responses probably offer the strongest evidence that the two illnesses are separate entities.
Although of great help in manic patients, anticonvulsants "have no efficacy whatsoever in schizophrenia." Likewise, antidepressants can "rocket" bipolar patients into mania, but "have you ever seen a schizophrenic patient given an antidepressant become acutely manic? I haven’t," Dr. Nemeroff said.
Perhaps the response to lithium is the most telling of all. It remains one of the most effective treatments for bipolar disorder but "just does not work" for schizophrenia, according to numerous trials as both monotherapy and add-on therapy, he said.
"I’ve seen dozens of [bipolar] patients" struggle for years despite treatment with newer agents, but who have never had a lithium trial. When it’s finally tried, they often have "phenomenal response[s]. They became euthymic and remain euthymic for years, and never suffer an episode. I’ve rarely ever seen that with any other treatment for bipolar disorder. [Lithium is] a great drug; it’s also cheap," he said. "Cheap is good for our patients."
Lithium is underused because "nobody’s marketing it to you," he said.
Ongoing monitoring for kidney, thyroid, and other side effects probably puts some psychiatrists off, as well, but it’s really not any more complicated than the ongoing monitoring needed with atypicals and anticonvulsants, he said.
Plus, "there’s pretty good data that if you check TSH [thyroid-stimulating hormone] every 6 months, you’ll be able to catch any incipient hyperthyroidism. Patients who have [thyroid] antibodies at baseline are generally the ones who go on to develop thyroid problems, so you can check that at baseline," he said.
For patients with refractory bipolar depression, "my favorite combination has been lamotrigine and MAO inhibitors," particularly tranylcypromine. "I’ve probably gotten more people [out of] bipolar depression with that – after they’ve been marinated in everything else – than any other strategy besides [electroconvulsive therapy]."
And "lamotrigine doesn’t work in patients with schizophrenia," he noted.
Dr. Nemeroff reported stock ownership, consultant fees, or other income from Allergan, Lilly, Shire, Roche, NovaDel Pharma, BioPharma, AstraZeneca, and other companies.
EXPERT ANALYSIS FROM THE NPA ANNUAL PSYCHOPHARMACOLOGY UPDATE
Off-label use of antipsychotics
Inhaled loxapine for agitation
Discuss this article at www.facebook.com/CurrentPsychiatry
Approved by the FDA on December 21, 2012, loxapine inhalation powder is the newest agent commercialized for the acute treatment of agitation associated with schizophrenia or bipolar I disorder in adults (Table 1).1,2 Loxapine is a first-generation antipsychotic that garnered newfound interest because of its potential atypical properties.3 Loxapine’s reformulation allows for direct administration to the lungs, resulting in rapid absorption into systemic circulation. This formulation offers a different method to manage agitation, for which IM formulations of other antipsychotics have been approved.4
Inhaled loxapine is delivered using a handheld device that produces a thermally-generated condensation aerosol.5,6 A single inhalation is sufficient to activate the controlled rapid heating (300 to 500°C in approximately 100 ms) of a thin layer of excipient-free loxapine on a metal substrate. Once vaporized, the medication cools down rapidly and aggregates into particles. The 1- to 3.5-micron aerosol particles of loxapine enter the respiratory track in 7
Table 1
Inhaled loxapine: Fast facts
Brand name: Adasuve |
Class: Dibenzoxazepine antipsychotic |
Indication: Acute treatment of agitation associated with schizophrenia or bipolar I disorder in adults |
FDA approval date: December 21, 2012 |
Availability date: Third quarter of 2013 |
Manufacturer: Alexza Pharmaceuticals |
Dosing forms: Single-dose inhaler, 10 mg |
Recommended dose: 10 mg; only a single dose within a 24-hour period is recommended |
Source: References 1,2 |
How it works
As with all antipsychotics, loxapine is an antagonist at the dopamine D2 receptor. However, loxapine also has clinically relevant serotonin-2A antagonism.3 Pharmacologic effects for loxapine and its metabolites include biogenic amine transporter inhibitor activity, alpha adrenergic blocking effects, and histaminergic and muscarinic receptor affinity.3,8
Clinical pharmacokinetics
In a phase I study of healthy volunteers, inhaled loxapine produced IV administration-type kinetics, with maximum plasma concentration achieved in approximately 2 minutes.6 Plasma exposure to loxapine was dose-proportional. Half-life for the 5- and 10-mg doses was approximately 6 hours. In these patients, exposure to loxapine’s metabolites as a percentage of exposure to the parent compound were 8.79% for 7-OH loxapine, 52.6% for 8-OH loxapine, and 3.96% for amoxapine (all produced as a result of metabolism via liver cytochrome P450 [CYP] enzymes CYP1A2, CYP2D6, and/or CYP3A46). 7-OH loxapine has a 5-fold higher affinity for the dopamine D2 receptor compared with loxapine, and may contribute to the drug’s clinical effect.6
Based on loxapine levels observed in the pharmacokinetic study,6 loxapine is not extensively metabolized in the lungs. Peak plasma concentrations immediately after inhalation are higher than for oral loxapine, but concentration of loxapine and its metabolites after the initial distribution phase is similar to that of oral loxapine.6 Loxapine and its metabolites are excreted through the kidneys.
Efficacy
Three efficacy studies were completed (Table 2)9-11; all were double-blind randomized controlled trials that compared inhaled loxapine, 5 or 10 mg, with placebo. Patients were required to be clinically agitated at baseline, with a score of ≥14 on the Positive and Negative Syndrome Scale Excited Component (PANSS-EC)—which consists of the PANSS items of tension, excitement, hostility, uncooperativeness, and poor impulse control; each item is rated from 1 (absent) to 7 (extreme)—and a score of ≥4 (moderate) on ≥1 item. Patients who were intoxicated or had a positive drug screen for psychostimulants were excluded. Lorazepam was allowed ≥2 hours after the study drug was administered. Change in the PANSS-EC was measured 10 minutes to 24 hours post-dose. The primary endpoint used to statistically test loxapine vs placebo was 2 hours post-dose.
In the initial phase II trial, loxapine 10 mg, but not 5 mg, was superior to placebo on the PANSS-EC at 2 hours.9 The authors described the 5-mg dose effect size as intermediate between placebo and the 10-mg dose, suggesting a possible dose response relationship. The 10-mg dose did separate from placebo as early as 20 minutes post-dose. The small number of patients enrolled is a limitation of this trial, but this was addressed in studies in the phase III program, which were considerably larger. For each of the 2 phase III trials—1 for patients with schizophrenia10 and the other for those with bipolar disorder (BD)11—both doses of loxapine were superior to placebo starting at 10 minutes post-dose. The number needed to treat (NNT) for response—as defined by a Clinical Global Impressions-Improvement score of much improved or very much improved—for loxapine vs placebo is included in Table 2.9-11 NNT for other outcomes, such as reduction on the PANSS-EC by at least 40% from baseline, demonstrated similar results.
12 The lower the NNT, the stronger the effect size.13 See the Box for an explanation of NNT. NNTs in the range of 3 to 5 are comparable to other agents used to treat agitation.4
When examining each individual item on the PANSS-EC in each of the phase III trials, every item improved with treatment, starting 10 to 20 minutes after dosing.14 Each item improved an average of 1 to 2 units from baseline over the first 2 hours post-dose. Moreover, inhaled loxapine appears to reduce agitation equally well in patients with higher or lower levels of agitation at baseline.
Another clinically relevant outcome is whether or not a patient required an additional dose or rescue medication within 24 hours. In the phase III schizophrenia trial,10 60.9% of patients randomized to loxapine, 10 mg, did not require an additional dose or rescue medication, compared with 54.4% and 46.1% for loxapine, 5 mg, and placebo, respectively. This yielded an NNT of 7 when comparing loxapine, 10 mg, with placebo.12 In the BD study,10 61.5%, 41.3%, and 26.7% did not require an additional dose or rescue medication within 24 hours for loxapine, 10 mg, 5 mg, and placebo, respectively. In this study, the NNT for loxapine, 10 mg, vs placebo was 3.12
In general, there appears to be a dose response for efficacy with inhaled loxapine, and therefore the FDA approved the 10-mg dose.2
Table 2
Summary of double-blind RCTs for inhaled loxapine vs inhaled placebo
Study | Diagnosis | Loxapine | Placebo | Outcomes | Loxapine vs placebo NNT for response at 2 hoursa | ||
---|---|---|---|---|---|---|---|
5 mg | 10 mg | 5 mg | 10 mg | ||||
Allen et al, 20119 (Phase II) | Agitation associated with schizophrenia | n=45 | n=41 | n=43 | On the PANSS-EC score at 2 hours, loxapine, 10 mg, but not 5 mg, was superior to placebo. Loxapine, 10 mg, separated from placebo at 20 minutes, and control was sustained. On the CGI-I at 2 hours, both doses of loxapine were superior to placebo. Using the BARS, loxapine, 10 mg, was superior to placebo starting at 30 minutes and this effect was sustained. Dysgeusia was observed in 4% and 17% for loxapine, 5 mg and 10 mg, respectively, and 9% for placebo | 4 | 3 |
Lesem et al, 201110 (Phase III) | Agitation associated with schizophrenia | n=116 | n=113 | n=115 | On the PANSS-EC score and CGI-I at 2 hours, both doses of loxapine were superior to placebo. Loxapine separated from placebo at 10 minutes. Sustained control was observed over 24 hours. Dysgeusia was observed in 9% and 11% for loxapine 5 mg and 10 mg, respectively, and 3% for placebo | 5 | 4 |
Kwentus et al, 201211 (Phase III) | Agitation associated with bipolar I disorder (manic or mixed episode) | n=104 | n=105 | n=105 | On the PANSS-EC score and CGI-I at 2 hours, both doses of loxapine were superior to placebo. Loxapine separated from placebo at 10 minutes. Sustained control was observed over 24 hours. Dysgeusia was observed in 17% for either loxapine 5 mg or 10 mg, respectively, and 6% for placebo | 3 | 3 |
aas measured by a CGI-I score of 1 or 2 BARS: Behavioral Activity Rating Scale; CGI-I: Clinical Global Impression Improvement Scale; NNT: number needed to treat; PANSS-EC: Positive and Negative Syndrome Scale Excited Component; RCTs: randomized controlled trials |
Clinical trials produce a mountain of data that can be difficult to interpret and apply to clinical practice. When reading about studies you may wonder:
- How large is the effect being measured?
- Is it clinically important?
- Are we dealing with a result that may be statistically significant but irrelevant for day-to-day patient care?
Number needed to treat (NNT) and number needed to harm (NNH)—2 tools of evidence-based medicine—can help answer these questions. NNT helps us gauge effect size—or clinical significance. It is different from knowing if a clinical trial result is statistically significant. NNT allows us to place a number on how often we can expect to encounter a difference between 2 interventions. If we see a therapeutic difference once every 100 patients (an NNT of 100), the difference between 2 treatments is not of great concern under most circumstances. But if a difference in outcome is seen once in every 5 patients being treated with 1 intervention vs another (an NNT of 5), the result likely will influence day-to-day practice.
How to calculate NNT (or NNH)
What is the NNT for an outcome for drug A vs drug B?
fA= frequency of outcome for drug A
fB= frequency of outcome for drug B
NNT = 1/[ fA - fB]
By convention, we round up the NNT to the next higher whole number.
For example, let’s say drugs A and B are used to treat depression, and they result in 6-week response rates of 55% and 75%, respectively. The NNT to encounter a difference between drug B and drug A in terms of responders at 6 weeks can be calculated as follows:
- Difference in response rates = 0.75 - 0.55 = 0.20
- NNT = 1 / 0.20 = 5.
Source: Adapted from Citrome L. Dissecting clinical trials with ‘number needed to treat.’ Current Psychiatry. 2007;6(3): 66-71 and Citrome L. Can you interpret confidence intervals? It’s not that difficult. Current Psychiatry. 2007;6(8):77-82
Tolerability and safety
Combined safety results from phase III trials10,11 as well as information about a phase I ECG QT interval study were presented in a poster.15 Among 524 patients receiving loxapine vs 263 receiving placebo, there were no significant differences in the likelihood of experiencing any adverse event, a nervous system adverse event, sedation, sedation or somnolence, or sedation, somnolence or dizziness, when stratified by lorazepam rescue.16 Adverse events that were more frequently encountered with both doses of loxapine (ie, 5 and 10 mg) than placebo are listed in Table 3,15 along with the number needed to harm (NNH). The most commonly encountered adverse event was dysgeusia. The NNH of 10 for dysgeusia for loxapine, 10 mg, vs placebo means that for every 10 patients receiving inhaled loxapine, 10 mg, instead of inhaled placebo, you would encounter 1 additional case of dysgeusia. This contrasts with the NNT for response of 4 and 3 for agitation associated with schizophrenia and BD, respectively. Therefore, one would encounter response more often than dysgeusia when comparing loxapine with placebo.
No important changes in the ECG QT interval after inhaled loxapine, 10 mg, were observed in a phase I study with healthy volunteers.15 Difference from placebo in change from baseline for QTc was
Additional details regarding overall safety and tolerability can be found in a previously published review.17
Table 3
Inhaled loxapine: Incidence of adverse events
Adverse event | Placebo (n=220) | Loxapine | |||
---|---|---|---|---|---|
5 mg (n=220) | 10 mg (n=218) | ||||
Rate | Rate | NNH vs placebo | Rate | NNH vs placebo | |
Dysgeusia | 4% | 13% | 12 | 14% | 10 |
Sedation or somnolence | 8% | 11% | 34 | 10% | 50 |
Oral hypoesthesia | 0% | 200 | 2% | 50 | |
NNH: number needed to harm Source: Reference 15 |
Pulmonary safety
Because this product is inhaled, additional information on pulmonary safety was gathered.18,19 Among 1,095 patients without active airways disease, 1 (0.09%) required treatment for post-treatment airway-related symptoms (bronchospasm). In the agitated patient population, the rate of airway adverse events was 0.4% of loxapine exposures among 524 patients, in which 6.7% had a history of asthma or chronic obstructive pulmonary disease (COPD). Others were likely to have some respiratory impairment because of a history of cigarette smoking, but they did not have active respiratory symptoms that required treatment because such patients were excluded from the trials.12 Phase I spirometry-based studies also were completed in healthy nonsmoking volunteers, in patients with asthma, and in patients with COPD. No clinically relevant effects were observed in healthy volunteers, but in patients with asthma or COPD a reduction in forced expiratory volume was observed. In patients with asthma, rates of bronchospasm as an adverse event were 26.9% for loxapine vs 3.8% for placebo, for a NNH of 5.12 Bronchospasm was not reported for patients with COPD receiving loxapine but was observed in 1 patient who received placebo. All airway adverse events in patients with asthma or COPD were mild or moderate. All respiratory signs or symptoms requiring treatment in the phase I asthma and COPD studies were managed with an inhaled bronchodilator.
Product labeling notes in a warning that inhaled loxapine can cause bronchospasm that has the potential to lead to respiratory distress and respiratory arrest.2 Therefore, inhaled loxapine is available only through a restricted program under a Risk Evaluation and Mitigation Strategy (REMS) called the “ADASUVE REMS.” Enrolled health care facilities are required to have immediate, on-site access to equipment and personnel trained to manage acute bronchospasm, including advanced airway management (intubation and mechanical ventilation). Inhaled loxapine is contraindicated in patients with a current diagnosis or history of asthma, COPD, or other lung diseases associated with bronchospasm; acute respiratory signs or symptoms such as wheezing; current use of medications to treat airway diseases such as asthma or COPD; history of bronchospasm following inhaled loxapine treatment; or known hypersensitivity to loxapine and amoxapine.
Only a single dose within a 24-hour period is recommended. Before administration, patients should be screened for a history of pulmonary disease and examined (including chest auscultation) for respiratory abnormalities (eg, wheezing). After administration, patients require monitoring for signs and symptoms of bronchospasm at least every 15 minutes for ≥1 hour.
Related Resource
- Dinh K, Myers DJ, Glazer M, et al. In vitro aerosol characterization of Staccato(®) Loxapine. Int J Pharm. 2011; 403(1-2):101-108.
Drug Brand Names
- Haloperidol • Haldol
- Lorazepam • Ativan
- Loxapine • Loxitane
- Loxapine inhalation powder • Adasuve
Disclosure
In the past 36 months, Dr. Citrome has engaged in collaborative research with or received consulting or speaking fees from Alexza Pharmaceuticals, Alkermes, AstraZeneca, Avanir Pharmaceuticals, Bristol-Myers Squibb, Eli Lilly and Company, EnVivo Pharmaceuticals, Forest Pharmaceuticals, Genentech, Janssen, L.P., Lundbeck, Merck, Mylan, Novartis, Noven, Otsuka, Pfizer Inc., Shire, Sunovion, and Valeant.
1. Alexza Pharmaceuticals U.S. FDA Approves Alexza’s ADASUVE (loxapine) inhalation powder for the acute treatment of agitation associated with schizophrenia or bipolar I disorder in adults. http://nocache-phx.corporate-ir.net/phoenix.zhtml?c=196151
&p=RssLanding&cat=news&id=1769476. Published December 21, 2012. Accessed January 2, 2013.
2. ADASUVE [package insert]. Mountain View, CA: Alexza Pharmaceuticals; 2012.
3. Ereshefsky L. Pharmacologic and pharmacokinetic considerations in choosing an antipsychotic. J Clin Psychiatry. 1999;60(suppl 10):20-30.
4. Citrome L. Comparison of intramuscular ziprasidone olanzapine, or aripiprazole for agitation: a quantitative review of efficacy and safety. J Clin Psychiatry. 2007;68(12):1876-1885.
5. Noymer P, Myers D, Glazer M, et al. The staccato system: inhaler design characteristics for rapid treatment of CNS disorders. Respiratory Drug Delivery. 2010;1(1):11-20.
6. Spyker DA, Munzar P, Cassella JV. Pharmacokinetics of loxapine following inhalation of a thermally generated aerosol in healthy volunteers. J Clin Pharmacol. 2010;50(2):169-179.
7. Dinh KV, Myers DJ, Noymer PD, et al. In vitro aerosol deposition in the oropharyngeal region for Staccato Loxapine. J Aerosol Med Pulm Drug Deliv. 2010;23(4):253-260.
8. Brunton LL, Lazo JS, Parker KL. eds. Goodman & Gilman’s: the pharmacological basis of therapeutics. 11th ed. New York, NY: McGraw-Hill; 2005:472.
9. Allen MH, Feifel DA, Lesem MD, et al. Efficacy and safety of loxapine for inhalation in the treatment of agitation in patients with schizophrenia: a randomized, double-blind, placebo controlled trial. J Clin Psychiatry. 2011;72(10):1313-1321.
10. Lesem MD, Tran-Johnson TK, Riesenberg RA, et al. Rapid acute treatment of agitation in individuals with schizophrenia: multicentre, randomised, placebo-controlled study of inhaled loxapine. Br J Psychiatry. 2011;198(1):51-58.
11. Kwentus J, Riesenberg RA, Marandi M, et al. Rapid acute treatment of agitation in patients with bipolar I disorder: a multicenter, randomized, placebo-controlled clinical trial with inhaled loxapine. Bipolar Disord. 2012;14(1):31-40.
12. Citrome L. Inhaled loxapine for agitation revisited: focus on effect sizes from 2 Phase III randomised controlled trials in persons with schizophrenia or bipolar disorder. Int J Clin Pract. 2012;66(3):318-325.
13. Citrome L. Compelling or irrelevant? Using number needed to treat can help decide. Acta Psychiatr Scand. 2008;117(6):412-419.
14. Cassella J, Spyker D, Kwentus J, et al. Rapid improvement in the five-item Positive and Negative Syndrome-Excited Component (PANSS-EC) scale for agitation with inhaled loxapine. Poster presented at: 50th meeting of New Research Approaches for Mental Health Interventions; June 14-17, 2010; Boca Raton, FL.
15. Fishman R, Gottwald M, Cassella J. Inhaled loxapine (AZ-004) rapidly and effectively reduces agitation in patients with schizophrenia and bipolar disorder. Poster presented at: 13th annual meeting of the College of Psychiatric and Neurologic Pharmacists; April 18-21 2010; San Antonio, TX.
16. Fishman R, Spyker D, Cassella J. The safety of concomitant use of lorazepam rescue in treating agitation with inhaled loxapine (AZ-004). Poster presented at: 50th meeting of New Research Approaches for Mental Health Interventions; June 14-17, 2010; Boca Raton, FL.
17. Citrome L. Aerosolised antipsychotic assuages agitation: inhaled loxapine for agitation associated with schizophrenia or bipolar disorder. Int J Clin Pract. 2011;65(3):330-340.
18. Alexza Pharmaceuticals. Adasuve (loxapine) inhalation powder NDA 022549. Psychopharmacologic drug advisory committee briefing document. http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/Psychopharma
cologicDrugsAdvisoryCommittee/UCM282900.pdf. Published December 12, 2011. Accessed January 2, 2013.
19. Food and Drug Administration Briefing document for NDA 022549. Psychopharmacologic Drug Advisory Committee Briefing Document. http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/Psychopharma
cologicDrugsAdvisoryCommittee/UCM282897.pdf. Accessed January 2, 2013.
Discuss this article at www.facebook.com/CurrentPsychiatry
Approved by the FDA on December 21, 2012, loxapine inhalation powder is the newest agent commercialized for the acute treatment of agitation associated with schizophrenia or bipolar I disorder in adults (Table 1).1,2 Loxapine is a first-generation antipsychotic that garnered newfound interest because of its potential atypical properties.3 Loxapine’s reformulation allows for direct administration to the lungs, resulting in rapid absorption into systemic circulation. This formulation offers a different method to manage agitation, for which IM formulations of other antipsychotics have been approved.4
Inhaled loxapine is delivered using a handheld device that produces a thermally-generated condensation aerosol.5,6 A single inhalation is sufficient to activate the controlled rapid heating (300 to 500°C in approximately 100 ms) of a thin layer of excipient-free loxapine on a metal substrate. Once vaporized, the medication cools down rapidly and aggregates into particles. The 1- to 3.5-micron aerosol particles of loxapine enter the respiratory track in 7
Table 1
Inhaled loxapine: Fast facts
Brand name: Adasuve |
Class: Dibenzoxazepine antipsychotic |
Indication: Acute treatment of agitation associated with schizophrenia or bipolar I disorder in adults |
FDA approval date: December 21, 2012 |
Availability date: Third quarter of 2013 |
Manufacturer: Alexza Pharmaceuticals |
Dosing forms: Single-dose inhaler, 10 mg |
Recommended dose: 10 mg; only a single dose within a 24-hour period is recommended |
Source: References 1,2 |
How it works
As with all antipsychotics, loxapine is an antagonist at the dopamine D2 receptor. However, loxapine also has clinically relevant serotonin-2A antagonism.3 Pharmacologic effects for loxapine and its metabolites include biogenic amine transporter inhibitor activity, alpha adrenergic blocking effects, and histaminergic and muscarinic receptor affinity.3,8
Clinical pharmacokinetics
In a phase I study of healthy volunteers, inhaled loxapine produced IV administration-type kinetics, with maximum plasma concentration achieved in approximately 2 minutes.6 Plasma exposure to loxapine was dose-proportional. Half-life for the 5- and 10-mg doses was approximately 6 hours. In these patients, exposure to loxapine’s metabolites as a percentage of exposure to the parent compound were 8.79% for 7-OH loxapine, 52.6% for 8-OH loxapine, and 3.96% for amoxapine (all produced as a result of metabolism via liver cytochrome P450 [CYP] enzymes CYP1A2, CYP2D6, and/or CYP3A46). 7-OH loxapine has a 5-fold higher affinity for the dopamine D2 receptor compared with loxapine, and may contribute to the drug’s clinical effect.6
Based on loxapine levels observed in the pharmacokinetic study,6 loxapine is not extensively metabolized in the lungs. Peak plasma concentrations immediately after inhalation are higher than for oral loxapine, but concentration of loxapine and its metabolites after the initial distribution phase is similar to that of oral loxapine.6 Loxapine and its metabolites are excreted through the kidneys.
Efficacy
Three efficacy studies were completed (Table 2)9-11; all were double-blind randomized controlled trials that compared inhaled loxapine, 5 or 10 mg, with placebo. Patients were required to be clinically agitated at baseline, with a score of ≥14 on the Positive and Negative Syndrome Scale Excited Component (PANSS-EC)—which consists of the PANSS items of tension, excitement, hostility, uncooperativeness, and poor impulse control; each item is rated from 1 (absent) to 7 (extreme)—and a score of ≥4 (moderate) on ≥1 item. Patients who were intoxicated or had a positive drug screen for psychostimulants were excluded. Lorazepam was allowed ≥2 hours after the study drug was administered. Change in the PANSS-EC was measured 10 minutes to 24 hours post-dose. The primary endpoint used to statistically test loxapine vs placebo was 2 hours post-dose.
In the initial phase II trial, loxapine 10 mg, but not 5 mg, was superior to placebo on the PANSS-EC at 2 hours.9 The authors described the 5-mg dose effect size as intermediate between placebo and the 10-mg dose, suggesting a possible dose response relationship. The 10-mg dose did separate from placebo as early as 20 minutes post-dose. The small number of patients enrolled is a limitation of this trial, but this was addressed in studies in the phase III program, which were considerably larger. For each of the 2 phase III trials—1 for patients with schizophrenia10 and the other for those with bipolar disorder (BD)11—both doses of loxapine were superior to placebo starting at 10 minutes post-dose. The number needed to treat (NNT) for response—as defined by a Clinical Global Impressions-Improvement score of much improved or very much improved—for loxapine vs placebo is included in Table 2.9-11 NNT for other outcomes, such as reduction on the PANSS-EC by at least 40% from baseline, demonstrated similar results.
12 The lower the NNT, the stronger the effect size.13 See the Box for an explanation of NNT. NNTs in the range of 3 to 5 are comparable to other agents used to treat agitation.4
When examining each individual item on the PANSS-EC in each of the phase III trials, every item improved with treatment, starting 10 to 20 minutes after dosing.14 Each item improved an average of 1 to 2 units from baseline over the first 2 hours post-dose. Moreover, inhaled loxapine appears to reduce agitation equally well in patients with higher or lower levels of agitation at baseline.
Another clinically relevant outcome is whether or not a patient required an additional dose or rescue medication within 24 hours. In the phase III schizophrenia trial,10 60.9% of patients randomized to loxapine, 10 mg, did not require an additional dose or rescue medication, compared with 54.4% and 46.1% for loxapine, 5 mg, and placebo, respectively. This yielded an NNT of 7 when comparing loxapine, 10 mg, with placebo.12 In the BD study,10 61.5%, 41.3%, and 26.7% did not require an additional dose or rescue medication within 24 hours for loxapine, 10 mg, 5 mg, and placebo, respectively. In this study, the NNT for loxapine, 10 mg, vs placebo was 3.12
In general, there appears to be a dose response for efficacy with inhaled loxapine, and therefore the FDA approved the 10-mg dose.2
Table 2
Summary of double-blind RCTs for inhaled loxapine vs inhaled placebo
Study | Diagnosis | Loxapine | Placebo | Outcomes | Loxapine vs placebo NNT for response at 2 hoursa | ||
---|---|---|---|---|---|---|---|
5 mg | 10 mg | 5 mg | 10 mg | ||||
Allen et al, 20119 (Phase II) | Agitation associated with schizophrenia | n=45 | n=41 | n=43 | On the PANSS-EC score at 2 hours, loxapine, 10 mg, but not 5 mg, was superior to placebo. Loxapine, 10 mg, separated from placebo at 20 minutes, and control was sustained. On the CGI-I at 2 hours, both doses of loxapine were superior to placebo. Using the BARS, loxapine, 10 mg, was superior to placebo starting at 30 minutes and this effect was sustained. Dysgeusia was observed in 4% and 17% for loxapine, 5 mg and 10 mg, respectively, and 9% for placebo | 4 | 3 |
Lesem et al, 201110 (Phase III) | Agitation associated with schizophrenia | n=116 | n=113 | n=115 | On the PANSS-EC score and CGI-I at 2 hours, both doses of loxapine were superior to placebo. Loxapine separated from placebo at 10 minutes. Sustained control was observed over 24 hours. Dysgeusia was observed in 9% and 11% for loxapine 5 mg and 10 mg, respectively, and 3% for placebo | 5 | 4 |
Kwentus et al, 201211 (Phase III) | Agitation associated with bipolar I disorder (manic or mixed episode) | n=104 | n=105 | n=105 | On the PANSS-EC score and CGI-I at 2 hours, both doses of loxapine were superior to placebo. Loxapine separated from placebo at 10 minutes. Sustained control was observed over 24 hours. Dysgeusia was observed in 17% for either loxapine 5 mg or 10 mg, respectively, and 6% for placebo | 3 | 3 |
aas measured by a CGI-I score of 1 or 2 BARS: Behavioral Activity Rating Scale; CGI-I: Clinical Global Impression Improvement Scale; NNT: number needed to treat; PANSS-EC: Positive and Negative Syndrome Scale Excited Component; RCTs: randomized controlled trials |
Clinical trials produce a mountain of data that can be difficult to interpret and apply to clinical practice. When reading about studies you may wonder:
- How large is the effect being measured?
- Is it clinically important?
- Are we dealing with a result that may be statistically significant but irrelevant for day-to-day patient care?
Number needed to treat (NNT) and number needed to harm (NNH)—2 tools of evidence-based medicine—can help answer these questions. NNT helps us gauge effect size—or clinical significance. It is different from knowing if a clinical trial result is statistically significant. NNT allows us to place a number on how often we can expect to encounter a difference between 2 interventions. If we see a therapeutic difference once every 100 patients (an NNT of 100), the difference between 2 treatments is not of great concern under most circumstances. But if a difference in outcome is seen once in every 5 patients being treated with 1 intervention vs another (an NNT of 5), the result likely will influence day-to-day practice.
How to calculate NNT (or NNH)
What is the NNT for an outcome for drug A vs drug B?
fA= frequency of outcome for drug A
fB= frequency of outcome for drug B
NNT = 1/[ fA - fB]
By convention, we round up the NNT to the next higher whole number.
For example, let’s say drugs A and B are used to treat depression, and they result in 6-week response rates of 55% and 75%, respectively. The NNT to encounter a difference between drug B and drug A in terms of responders at 6 weeks can be calculated as follows:
- Difference in response rates = 0.75 - 0.55 = 0.20
- NNT = 1 / 0.20 = 5.
Source: Adapted from Citrome L. Dissecting clinical trials with ‘number needed to treat.’ Current Psychiatry. 2007;6(3): 66-71 and Citrome L. Can you interpret confidence intervals? It’s not that difficult. Current Psychiatry. 2007;6(8):77-82
Tolerability and safety
Combined safety results from phase III trials10,11 as well as information about a phase I ECG QT interval study were presented in a poster.15 Among 524 patients receiving loxapine vs 263 receiving placebo, there were no significant differences in the likelihood of experiencing any adverse event, a nervous system adverse event, sedation, sedation or somnolence, or sedation, somnolence or dizziness, when stratified by lorazepam rescue.16 Adverse events that were more frequently encountered with both doses of loxapine (ie, 5 and 10 mg) than placebo are listed in Table 3,15 along with the number needed to harm (NNH). The most commonly encountered adverse event was dysgeusia. The NNH of 10 for dysgeusia for loxapine, 10 mg, vs placebo means that for every 10 patients receiving inhaled loxapine, 10 mg, instead of inhaled placebo, you would encounter 1 additional case of dysgeusia. This contrasts with the NNT for response of 4 and 3 for agitation associated with schizophrenia and BD, respectively. Therefore, one would encounter response more often than dysgeusia when comparing loxapine with placebo.
No important changes in the ECG QT interval after inhaled loxapine, 10 mg, were observed in a phase I study with healthy volunteers.15 Difference from placebo in change from baseline for QTc was
Additional details regarding overall safety and tolerability can be found in a previously published review.17
Table 3
Inhaled loxapine: Incidence of adverse events
Adverse event | Placebo (n=220) | Loxapine | |||
---|---|---|---|---|---|
5 mg (n=220) | 10 mg (n=218) | ||||
Rate | Rate | NNH vs placebo | Rate | NNH vs placebo | |
Dysgeusia | 4% | 13% | 12 | 14% | 10 |
Sedation or somnolence | 8% | 11% | 34 | 10% | 50 |
Oral hypoesthesia | 0% | 200 | 2% | 50 | |
NNH: number needed to harm Source: Reference 15 |
Pulmonary safety
Because this product is inhaled, additional information on pulmonary safety was gathered.18,19 Among 1,095 patients without active airways disease, 1 (0.09%) required treatment for post-treatment airway-related symptoms (bronchospasm). In the agitated patient population, the rate of airway adverse events was 0.4% of loxapine exposures among 524 patients, in which 6.7% had a history of asthma or chronic obstructive pulmonary disease (COPD). Others were likely to have some respiratory impairment because of a history of cigarette smoking, but they did not have active respiratory symptoms that required treatment because such patients were excluded from the trials.12 Phase I spirometry-based studies also were completed in healthy nonsmoking volunteers, in patients with asthma, and in patients with COPD. No clinically relevant effects were observed in healthy volunteers, but in patients with asthma or COPD a reduction in forced expiratory volume was observed. In patients with asthma, rates of bronchospasm as an adverse event were 26.9% for loxapine vs 3.8% for placebo, for a NNH of 5.12 Bronchospasm was not reported for patients with COPD receiving loxapine but was observed in 1 patient who received placebo. All airway adverse events in patients with asthma or COPD were mild or moderate. All respiratory signs or symptoms requiring treatment in the phase I asthma and COPD studies were managed with an inhaled bronchodilator.
Product labeling notes in a warning that inhaled loxapine can cause bronchospasm that has the potential to lead to respiratory distress and respiratory arrest.2 Therefore, inhaled loxapine is available only through a restricted program under a Risk Evaluation and Mitigation Strategy (REMS) called the “ADASUVE REMS.” Enrolled health care facilities are required to have immediate, on-site access to equipment and personnel trained to manage acute bronchospasm, including advanced airway management (intubation and mechanical ventilation). Inhaled loxapine is contraindicated in patients with a current diagnosis or history of asthma, COPD, or other lung diseases associated with bronchospasm; acute respiratory signs or symptoms such as wheezing; current use of medications to treat airway diseases such as asthma or COPD; history of bronchospasm following inhaled loxapine treatment; or known hypersensitivity to loxapine and amoxapine.
Only a single dose within a 24-hour period is recommended. Before administration, patients should be screened for a history of pulmonary disease and examined (including chest auscultation) for respiratory abnormalities (eg, wheezing). After administration, patients require monitoring for signs and symptoms of bronchospasm at least every 15 minutes for ≥1 hour.
Related Resource
- Dinh K, Myers DJ, Glazer M, et al. In vitro aerosol characterization of Staccato(®) Loxapine. Int J Pharm. 2011; 403(1-2):101-108.
Drug Brand Names
- Haloperidol • Haldol
- Lorazepam • Ativan
- Loxapine • Loxitane
- Loxapine inhalation powder • Adasuve
Disclosure
In the past 36 months, Dr. Citrome has engaged in collaborative research with or received consulting or speaking fees from Alexza Pharmaceuticals, Alkermes, AstraZeneca, Avanir Pharmaceuticals, Bristol-Myers Squibb, Eli Lilly and Company, EnVivo Pharmaceuticals, Forest Pharmaceuticals, Genentech, Janssen, L.P., Lundbeck, Merck, Mylan, Novartis, Noven, Otsuka, Pfizer Inc., Shire, Sunovion, and Valeant.
Discuss this article at www.facebook.com/CurrentPsychiatry
Approved by the FDA on December 21, 2012, loxapine inhalation powder is the newest agent commercialized for the acute treatment of agitation associated with schizophrenia or bipolar I disorder in adults (Table 1).1,2 Loxapine is a first-generation antipsychotic that garnered newfound interest because of its potential atypical properties.3 Loxapine’s reformulation allows for direct administration to the lungs, resulting in rapid absorption into systemic circulation. This formulation offers a different method to manage agitation, for which IM formulations of other antipsychotics have been approved.4
Inhaled loxapine is delivered using a handheld device that produces a thermally-generated condensation aerosol.5,6 A single inhalation is sufficient to activate the controlled rapid heating (300 to 500°C in approximately 100 ms) of a thin layer of excipient-free loxapine on a metal substrate. Once vaporized, the medication cools down rapidly and aggregates into particles. The 1- to 3.5-micron aerosol particles of loxapine enter the respiratory track in 7
Table 1
Inhaled loxapine: Fast facts
Brand name: Adasuve |
Class: Dibenzoxazepine antipsychotic |
Indication: Acute treatment of agitation associated with schizophrenia or bipolar I disorder in adults |
FDA approval date: December 21, 2012 |
Availability date: Third quarter of 2013 |
Manufacturer: Alexza Pharmaceuticals |
Dosing forms: Single-dose inhaler, 10 mg |
Recommended dose: 10 mg; only a single dose within a 24-hour period is recommended |
Source: References 1,2 |
How it works
As with all antipsychotics, loxapine is an antagonist at the dopamine D2 receptor. However, loxapine also has clinically relevant serotonin-2A antagonism.3 Pharmacologic effects for loxapine and its metabolites include biogenic amine transporter inhibitor activity, alpha adrenergic blocking effects, and histaminergic and muscarinic receptor affinity.3,8
Clinical pharmacokinetics
In a phase I study of healthy volunteers, inhaled loxapine produced IV administration-type kinetics, with maximum plasma concentration achieved in approximately 2 minutes.6 Plasma exposure to loxapine was dose-proportional. Half-life for the 5- and 10-mg doses was approximately 6 hours. In these patients, exposure to loxapine’s metabolites as a percentage of exposure to the parent compound were 8.79% for 7-OH loxapine, 52.6% for 8-OH loxapine, and 3.96% for amoxapine (all produced as a result of metabolism via liver cytochrome P450 [CYP] enzymes CYP1A2, CYP2D6, and/or CYP3A46). 7-OH loxapine has a 5-fold higher affinity for the dopamine D2 receptor compared with loxapine, and may contribute to the drug’s clinical effect.6
Based on loxapine levels observed in the pharmacokinetic study,6 loxapine is not extensively metabolized in the lungs. Peak plasma concentrations immediately after inhalation are higher than for oral loxapine, but concentration of loxapine and its metabolites after the initial distribution phase is similar to that of oral loxapine.6 Loxapine and its metabolites are excreted through the kidneys.
Efficacy
Three efficacy studies were completed (Table 2)9-11; all were double-blind randomized controlled trials that compared inhaled loxapine, 5 or 10 mg, with placebo. Patients were required to be clinically agitated at baseline, with a score of ≥14 on the Positive and Negative Syndrome Scale Excited Component (PANSS-EC)—which consists of the PANSS items of tension, excitement, hostility, uncooperativeness, and poor impulse control; each item is rated from 1 (absent) to 7 (extreme)—and a score of ≥4 (moderate) on ≥1 item. Patients who were intoxicated or had a positive drug screen for psychostimulants were excluded. Lorazepam was allowed ≥2 hours after the study drug was administered. Change in the PANSS-EC was measured 10 minutes to 24 hours post-dose. The primary endpoint used to statistically test loxapine vs placebo was 2 hours post-dose.
In the initial phase II trial, loxapine 10 mg, but not 5 mg, was superior to placebo on the PANSS-EC at 2 hours.9 The authors described the 5-mg dose effect size as intermediate between placebo and the 10-mg dose, suggesting a possible dose response relationship. The 10-mg dose did separate from placebo as early as 20 minutes post-dose. The small number of patients enrolled is a limitation of this trial, but this was addressed in studies in the phase III program, which were considerably larger. For each of the 2 phase III trials—1 for patients with schizophrenia10 and the other for those with bipolar disorder (BD)11—both doses of loxapine were superior to placebo starting at 10 minutes post-dose. The number needed to treat (NNT) for response—as defined by a Clinical Global Impressions-Improvement score of much improved or very much improved—for loxapine vs placebo is included in Table 2.9-11 NNT for other outcomes, such as reduction on the PANSS-EC by at least 40% from baseline, demonstrated similar results.
12 The lower the NNT, the stronger the effect size.13 See the Box for an explanation of NNT. NNTs in the range of 3 to 5 are comparable to other agents used to treat agitation.4
When examining each individual item on the PANSS-EC in each of the phase III trials, every item improved with treatment, starting 10 to 20 minutes after dosing.14 Each item improved an average of 1 to 2 units from baseline over the first 2 hours post-dose. Moreover, inhaled loxapine appears to reduce agitation equally well in patients with higher or lower levels of agitation at baseline.
Another clinically relevant outcome is whether or not a patient required an additional dose or rescue medication within 24 hours. In the phase III schizophrenia trial,10 60.9% of patients randomized to loxapine, 10 mg, did not require an additional dose or rescue medication, compared with 54.4% and 46.1% for loxapine, 5 mg, and placebo, respectively. This yielded an NNT of 7 when comparing loxapine, 10 mg, with placebo.12 In the BD study,10 61.5%, 41.3%, and 26.7% did not require an additional dose or rescue medication within 24 hours for loxapine, 10 mg, 5 mg, and placebo, respectively. In this study, the NNT for loxapine, 10 mg, vs placebo was 3.12
In general, there appears to be a dose response for efficacy with inhaled loxapine, and therefore the FDA approved the 10-mg dose.2
Table 2
Summary of double-blind RCTs for inhaled loxapine vs inhaled placebo
Study | Diagnosis | Loxapine | Placebo | Outcomes | Loxapine vs placebo NNT for response at 2 hoursa | ||
---|---|---|---|---|---|---|---|
5 mg | 10 mg | 5 mg | 10 mg | ||||
Allen et al, 20119 (Phase II) | Agitation associated with schizophrenia | n=45 | n=41 | n=43 | On the PANSS-EC score at 2 hours, loxapine, 10 mg, but not 5 mg, was superior to placebo. Loxapine, 10 mg, separated from placebo at 20 minutes, and control was sustained. On the CGI-I at 2 hours, both doses of loxapine were superior to placebo. Using the BARS, loxapine, 10 mg, was superior to placebo starting at 30 minutes and this effect was sustained. Dysgeusia was observed in 4% and 17% for loxapine, 5 mg and 10 mg, respectively, and 9% for placebo | 4 | 3 |
Lesem et al, 201110 (Phase III) | Agitation associated with schizophrenia | n=116 | n=113 | n=115 | On the PANSS-EC score and CGI-I at 2 hours, both doses of loxapine were superior to placebo. Loxapine separated from placebo at 10 minutes. Sustained control was observed over 24 hours. Dysgeusia was observed in 9% and 11% for loxapine 5 mg and 10 mg, respectively, and 3% for placebo | 5 | 4 |
Kwentus et al, 201211 (Phase III) | Agitation associated with bipolar I disorder (manic or mixed episode) | n=104 | n=105 | n=105 | On the PANSS-EC score and CGI-I at 2 hours, both doses of loxapine were superior to placebo. Loxapine separated from placebo at 10 minutes. Sustained control was observed over 24 hours. Dysgeusia was observed in 17% for either loxapine 5 mg or 10 mg, respectively, and 6% for placebo | 3 | 3 |
aas measured by a CGI-I score of 1 or 2 BARS: Behavioral Activity Rating Scale; CGI-I: Clinical Global Impression Improvement Scale; NNT: number needed to treat; PANSS-EC: Positive and Negative Syndrome Scale Excited Component; RCTs: randomized controlled trials |
Clinical trials produce a mountain of data that can be difficult to interpret and apply to clinical practice. When reading about studies you may wonder:
- How large is the effect being measured?
- Is it clinically important?
- Are we dealing with a result that may be statistically significant but irrelevant for day-to-day patient care?
Number needed to treat (NNT) and number needed to harm (NNH)—2 tools of evidence-based medicine—can help answer these questions. NNT helps us gauge effect size—or clinical significance. It is different from knowing if a clinical trial result is statistically significant. NNT allows us to place a number on how often we can expect to encounter a difference between 2 interventions. If we see a therapeutic difference once every 100 patients (an NNT of 100), the difference between 2 treatments is not of great concern under most circumstances. But if a difference in outcome is seen once in every 5 patients being treated with 1 intervention vs another (an NNT of 5), the result likely will influence day-to-day practice.
How to calculate NNT (or NNH)
What is the NNT for an outcome for drug A vs drug B?
fA= frequency of outcome for drug A
fB= frequency of outcome for drug B
NNT = 1/[ fA - fB]
By convention, we round up the NNT to the next higher whole number.
For example, let’s say drugs A and B are used to treat depression, and they result in 6-week response rates of 55% and 75%, respectively. The NNT to encounter a difference between drug B and drug A in terms of responders at 6 weeks can be calculated as follows:
- Difference in response rates = 0.75 - 0.55 = 0.20
- NNT = 1 / 0.20 = 5.
Source: Adapted from Citrome L. Dissecting clinical trials with ‘number needed to treat.’ Current Psychiatry. 2007;6(3): 66-71 and Citrome L. Can you interpret confidence intervals? It’s not that difficult. Current Psychiatry. 2007;6(8):77-82
Tolerability and safety
Combined safety results from phase III trials10,11 as well as information about a phase I ECG QT interval study were presented in a poster.15 Among 524 patients receiving loxapine vs 263 receiving placebo, there were no significant differences in the likelihood of experiencing any adverse event, a nervous system adverse event, sedation, sedation or somnolence, or sedation, somnolence or dizziness, when stratified by lorazepam rescue.16 Adverse events that were more frequently encountered with both doses of loxapine (ie, 5 and 10 mg) than placebo are listed in Table 3,15 along with the number needed to harm (NNH). The most commonly encountered adverse event was dysgeusia. The NNH of 10 for dysgeusia for loxapine, 10 mg, vs placebo means that for every 10 patients receiving inhaled loxapine, 10 mg, instead of inhaled placebo, you would encounter 1 additional case of dysgeusia. This contrasts with the NNT for response of 4 and 3 for agitation associated with schizophrenia and BD, respectively. Therefore, one would encounter response more often than dysgeusia when comparing loxapine with placebo.
No important changes in the ECG QT interval after inhaled loxapine, 10 mg, were observed in a phase I study with healthy volunteers.15 Difference from placebo in change from baseline for QTc was
Additional details regarding overall safety and tolerability can be found in a previously published review.17
Table 3
Inhaled loxapine: Incidence of adverse events
Adverse event | Placebo (n=220) | Loxapine | |||
---|---|---|---|---|---|
5 mg (n=220) | 10 mg (n=218) | ||||
Rate | Rate | NNH vs placebo | Rate | NNH vs placebo | |
Dysgeusia | 4% | 13% | 12 | 14% | 10 |
Sedation or somnolence | 8% | 11% | 34 | 10% | 50 |
Oral hypoesthesia | 0% | 200 | 2% | 50 | |
NNH: number needed to harm Source: Reference 15 |
Pulmonary safety
Because this product is inhaled, additional information on pulmonary safety was gathered.18,19 Among 1,095 patients without active airways disease, 1 (0.09%) required treatment for post-treatment airway-related symptoms (bronchospasm). In the agitated patient population, the rate of airway adverse events was 0.4% of loxapine exposures among 524 patients, in which 6.7% had a history of asthma or chronic obstructive pulmonary disease (COPD). Others were likely to have some respiratory impairment because of a history of cigarette smoking, but they did not have active respiratory symptoms that required treatment because such patients were excluded from the trials.12 Phase I spirometry-based studies also were completed in healthy nonsmoking volunteers, in patients with asthma, and in patients with COPD. No clinically relevant effects were observed in healthy volunteers, but in patients with asthma or COPD a reduction in forced expiratory volume was observed. In patients with asthma, rates of bronchospasm as an adverse event were 26.9% for loxapine vs 3.8% for placebo, for a NNH of 5.12 Bronchospasm was not reported for patients with COPD receiving loxapine but was observed in 1 patient who received placebo. All airway adverse events in patients with asthma or COPD were mild or moderate. All respiratory signs or symptoms requiring treatment in the phase I asthma and COPD studies were managed with an inhaled bronchodilator.
Product labeling notes in a warning that inhaled loxapine can cause bronchospasm that has the potential to lead to respiratory distress and respiratory arrest.2 Therefore, inhaled loxapine is available only through a restricted program under a Risk Evaluation and Mitigation Strategy (REMS) called the “ADASUVE REMS.” Enrolled health care facilities are required to have immediate, on-site access to equipment and personnel trained to manage acute bronchospasm, including advanced airway management (intubation and mechanical ventilation). Inhaled loxapine is contraindicated in patients with a current diagnosis or history of asthma, COPD, or other lung diseases associated with bronchospasm; acute respiratory signs or symptoms such as wheezing; current use of medications to treat airway diseases such as asthma or COPD; history of bronchospasm following inhaled loxapine treatment; or known hypersensitivity to loxapine and amoxapine.
Only a single dose within a 24-hour period is recommended. Before administration, patients should be screened for a history of pulmonary disease and examined (including chest auscultation) for respiratory abnormalities (eg, wheezing). After administration, patients require monitoring for signs and symptoms of bronchospasm at least every 15 minutes for ≥1 hour.
Related Resource
- Dinh K, Myers DJ, Glazer M, et al. In vitro aerosol characterization of Staccato(®) Loxapine. Int J Pharm. 2011; 403(1-2):101-108.
Drug Brand Names
- Haloperidol • Haldol
- Lorazepam • Ativan
- Loxapine • Loxitane
- Loxapine inhalation powder • Adasuve
Disclosure
In the past 36 months, Dr. Citrome has engaged in collaborative research with or received consulting or speaking fees from Alexza Pharmaceuticals, Alkermes, AstraZeneca, Avanir Pharmaceuticals, Bristol-Myers Squibb, Eli Lilly and Company, EnVivo Pharmaceuticals, Forest Pharmaceuticals, Genentech, Janssen, L.P., Lundbeck, Merck, Mylan, Novartis, Noven, Otsuka, Pfizer Inc., Shire, Sunovion, and Valeant.
1. Alexza Pharmaceuticals U.S. FDA Approves Alexza’s ADASUVE (loxapine) inhalation powder for the acute treatment of agitation associated with schizophrenia or bipolar I disorder in adults. http://nocache-phx.corporate-ir.net/phoenix.zhtml?c=196151
&p=RssLanding&cat=news&id=1769476. Published December 21, 2012. Accessed January 2, 2013.
2. ADASUVE [package insert]. Mountain View, CA: Alexza Pharmaceuticals; 2012.
3. Ereshefsky L. Pharmacologic and pharmacokinetic considerations in choosing an antipsychotic. J Clin Psychiatry. 1999;60(suppl 10):20-30.
4. Citrome L. Comparison of intramuscular ziprasidone olanzapine, or aripiprazole for agitation: a quantitative review of efficacy and safety. J Clin Psychiatry. 2007;68(12):1876-1885.
5. Noymer P, Myers D, Glazer M, et al. The staccato system: inhaler design characteristics for rapid treatment of CNS disorders. Respiratory Drug Delivery. 2010;1(1):11-20.
6. Spyker DA, Munzar P, Cassella JV. Pharmacokinetics of loxapine following inhalation of a thermally generated aerosol in healthy volunteers. J Clin Pharmacol. 2010;50(2):169-179.
7. Dinh KV, Myers DJ, Noymer PD, et al. In vitro aerosol deposition in the oropharyngeal region for Staccato Loxapine. J Aerosol Med Pulm Drug Deliv. 2010;23(4):253-260.
8. Brunton LL, Lazo JS, Parker KL. eds. Goodman & Gilman’s: the pharmacological basis of therapeutics. 11th ed. New York, NY: McGraw-Hill; 2005:472.
9. Allen MH, Feifel DA, Lesem MD, et al. Efficacy and safety of loxapine for inhalation in the treatment of agitation in patients with schizophrenia: a randomized, double-blind, placebo controlled trial. J Clin Psychiatry. 2011;72(10):1313-1321.
10. Lesem MD, Tran-Johnson TK, Riesenberg RA, et al. Rapid acute treatment of agitation in individuals with schizophrenia: multicentre, randomised, placebo-controlled study of inhaled loxapine. Br J Psychiatry. 2011;198(1):51-58.
11. Kwentus J, Riesenberg RA, Marandi M, et al. Rapid acute treatment of agitation in patients with bipolar I disorder: a multicenter, randomized, placebo-controlled clinical trial with inhaled loxapine. Bipolar Disord. 2012;14(1):31-40.
12. Citrome L. Inhaled loxapine for agitation revisited: focus on effect sizes from 2 Phase III randomised controlled trials in persons with schizophrenia or bipolar disorder. Int J Clin Pract. 2012;66(3):318-325.
13. Citrome L. Compelling or irrelevant? Using number needed to treat can help decide. Acta Psychiatr Scand. 2008;117(6):412-419.
14. Cassella J, Spyker D, Kwentus J, et al. Rapid improvement in the five-item Positive and Negative Syndrome-Excited Component (PANSS-EC) scale for agitation with inhaled loxapine. Poster presented at: 50th meeting of New Research Approaches for Mental Health Interventions; June 14-17, 2010; Boca Raton, FL.
15. Fishman R, Gottwald M, Cassella J. Inhaled loxapine (AZ-004) rapidly and effectively reduces agitation in patients with schizophrenia and bipolar disorder. Poster presented at: 13th annual meeting of the College of Psychiatric and Neurologic Pharmacists; April 18-21 2010; San Antonio, TX.
16. Fishman R, Spyker D, Cassella J. The safety of concomitant use of lorazepam rescue in treating agitation with inhaled loxapine (AZ-004). Poster presented at: 50th meeting of New Research Approaches for Mental Health Interventions; June 14-17, 2010; Boca Raton, FL.
17. Citrome L. Aerosolised antipsychotic assuages agitation: inhaled loxapine for agitation associated with schizophrenia or bipolar disorder. Int J Clin Pract. 2011;65(3):330-340.
18. Alexza Pharmaceuticals. Adasuve (loxapine) inhalation powder NDA 022549. Psychopharmacologic drug advisory committee briefing document. http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/Psychopharma
cologicDrugsAdvisoryCommittee/UCM282900.pdf. Published December 12, 2011. Accessed January 2, 2013.
19. Food and Drug Administration Briefing document for NDA 022549. Psychopharmacologic Drug Advisory Committee Briefing Document. http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/Psychopharma
cologicDrugsAdvisoryCommittee/UCM282897.pdf. Accessed January 2, 2013.
1. Alexza Pharmaceuticals U.S. FDA Approves Alexza’s ADASUVE (loxapine) inhalation powder for the acute treatment of agitation associated with schizophrenia or bipolar I disorder in adults. http://nocache-phx.corporate-ir.net/phoenix.zhtml?c=196151
&p=RssLanding&cat=news&id=1769476. Published December 21, 2012. Accessed January 2, 2013.
2. ADASUVE [package insert]. Mountain View, CA: Alexza Pharmaceuticals; 2012.
3. Ereshefsky L. Pharmacologic and pharmacokinetic considerations in choosing an antipsychotic. J Clin Psychiatry. 1999;60(suppl 10):20-30.
4. Citrome L. Comparison of intramuscular ziprasidone olanzapine, or aripiprazole for agitation: a quantitative review of efficacy and safety. J Clin Psychiatry. 2007;68(12):1876-1885.
5. Noymer P, Myers D, Glazer M, et al. The staccato system: inhaler design characteristics for rapid treatment of CNS disorders. Respiratory Drug Delivery. 2010;1(1):11-20.
6. Spyker DA, Munzar P, Cassella JV. Pharmacokinetics of loxapine following inhalation of a thermally generated aerosol in healthy volunteers. J Clin Pharmacol. 2010;50(2):169-179.
7. Dinh KV, Myers DJ, Noymer PD, et al. In vitro aerosol deposition in the oropharyngeal region for Staccato Loxapine. J Aerosol Med Pulm Drug Deliv. 2010;23(4):253-260.
8. Brunton LL, Lazo JS, Parker KL. eds. Goodman & Gilman’s: the pharmacological basis of therapeutics. 11th ed. New York, NY: McGraw-Hill; 2005:472.
9. Allen MH, Feifel DA, Lesem MD, et al. Efficacy and safety of loxapine for inhalation in the treatment of agitation in patients with schizophrenia: a randomized, double-blind, placebo controlled trial. J Clin Psychiatry. 2011;72(10):1313-1321.
10. Lesem MD, Tran-Johnson TK, Riesenberg RA, et al. Rapid acute treatment of agitation in individuals with schizophrenia: multicentre, randomised, placebo-controlled study of inhaled loxapine. Br J Psychiatry. 2011;198(1):51-58.
11. Kwentus J, Riesenberg RA, Marandi M, et al. Rapid acute treatment of agitation in patients with bipolar I disorder: a multicenter, randomized, placebo-controlled clinical trial with inhaled loxapine. Bipolar Disord. 2012;14(1):31-40.
12. Citrome L. Inhaled loxapine for agitation revisited: focus on effect sizes from 2 Phase III randomised controlled trials in persons with schizophrenia or bipolar disorder. Int J Clin Pract. 2012;66(3):318-325.
13. Citrome L. Compelling or irrelevant? Using number needed to treat can help decide. Acta Psychiatr Scand. 2008;117(6):412-419.
14. Cassella J, Spyker D, Kwentus J, et al. Rapid improvement in the five-item Positive and Negative Syndrome-Excited Component (PANSS-EC) scale for agitation with inhaled loxapine. Poster presented at: 50th meeting of New Research Approaches for Mental Health Interventions; June 14-17, 2010; Boca Raton, FL.
15. Fishman R, Gottwald M, Cassella J. Inhaled loxapine (AZ-004) rapidly and effectively reduces agitation in patients with schizophrenia and bipolar disorder. Poster presented at: 13th annual meeting of the College of Psychiatric and Neurologic Pharmacists; April 18-21 2010; San Antonio, TX.
16. Fishman R, Spyker D, Cassella J. The safety of concomitant use of lorazepam rescue in treating agitation with inhaled loxapine (AZ-004). Poster presented at: 50th meeting of New Research Approaches for Mental Health Interventions; June 14-17, 2010; Boca Raton, FL.
17. Citrome L. Aerosolised antipsychotic assuages agitation: inhaled loxapine for agitation associated with schizophrenia or bipolar disorder. Int J Clin Pract. 2011;65(3):330-340.
18. Alexza Pharmaceuticals. Adasuve (loxapine) inhalation powder NDA 022549. Psychopharmacologic drug advisory committee briefing document. http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/Psychopharma
cologicDrugsAdvisoryCommittee/UCM282900.pdf. Published December 12, 2011. Accessed January 2, 2013.
19. Food and Drug Administration Briefing document for NDA 022549. Psychopharmacologic Drug Advisory Committee Briefing Document. http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/Psychopharma
cologicDrugsAdvisoryCommittee/UCM282897.pdf. Accessed January 2, 2013.
Bipolar disorder or something else?
CASE: Unclear diagnosis
Police find Ms. S, age 31, extremely intoxicated and drinking alcohol in her car in a city park parking lot. In the emergency room, she becomes increasingly somnolent and clinicians intubate her trachea to protect her airway. Lab testing shows she has elevated acetaminophen and lithium serum levels, and she is transferred to our hospital for further management after being started on N-acetylcysteine to treat acetaminophen toxicity. Her “ex-fiancé,” the father of her 2 children, saw her earlier the day of the episode and says she was distraught, intoxicated, and had several empty pill bottles in her purse.
In our hospital, Ms. S’ lithium level increases from 2.3 mEq/L to a peak of 5.32 mEq/L, and she undergoes hemodialysis. On hospital day 2, her serum lithium level is trending downward. After Ms. S is able to breathe spontaneously, her trachea is extubated and her hemodialysis line is removed. A psychiatric consultation is obtained, but she is unable to provide a coherent history and the treating clinicians believe she has delirium caused by multiple factors.
On hospital day 3, Ms. S’ delirium clears enough for her to engage in an interview, and she is transferred to our inpatient psychiatry ward for further monitoring and stabilization.
She reports that she was diagnosed with bipolar disorder (BD) at age 12, when she faced multiple psychosocial stressors, including physical abuse by her mother’s boyfriend. She took several psychotropics—although she cannot remember which ones—until age 14, when she stopped all medications until the year before her current hospitalization. Although throughout adolescence and adulthood Ms. S experienced chronic irritability, anxiety, impulsive behavior, poor self-esteem, abusive relationships, self-cutting, and depressed mood, she maintains that she felt worse when she was taking psychotropics and doubts the BD diagnosis. She attributes her longstanding mood issues to low self-worth, a “codependent nature,” and a tendency to gravitate toward abusive relationships. Although she admits to experimenting with several illicit drugs during adolescence, she denies more recent substance use and states she drinks alcohol only once every few months.
The authors’ observations
BD is underdiagnosed in several patient populations, such as individuals previously diagnosed with MDD.1-3 Misdiagnosis can have severe implications, including delay in receiving treatment with effective medications (eg, mood stabilizers) or use of agents that can induce mania or rapid-cycling, such as antidepressants. Perhaps in response to this concern, in recent years clinicians increasingly have diagnosed BD in adolescents and adults. An analysis of a national database of physician practices found a 40-fold increase in office visits for BD among youth and a near doubling among adults from 1994 to 2003.4
Although underdiagnosis of BD remains important, some researchers have suggested that overdiagnosis may be more prevalent and equally harmful. In a study of 180 patients being treated for depression in a family care clinic, there was a 21.6% initial underdiagnosis rate among those eventually found to have BD.1 However, among 43 patients with a prior BD diagnosis, the diagnosis was not confirmed in 33%.1 In a study of 700 psychiatric outpatients in Rhode Island, only 43% of 145 patients who reported a prior BD diagnosis had that diagnosis confirmed.5 Three times as many patients were overdiagnosed with BD as underdiagnosed.
Are there characteristics common to individuals incorrectly diagnosed with BD? In a study that compared patients who had been mistakenly diagnosed with BD with those who had not been diagnosed with BD, the overdiagnosis group was significantly more likely to be diagnosed with a personality disorder, in particular borderline or antisocial personality disorder.6 Only lifetime and current BPD, current posttraumatic stress disorder (PTSD), and lifetime impulse control disorders were independently associated with BD overdiagnosis. The odds ratio for overdiagnosis of BD in patients found to have BPD was 3.7.
EVALUATION: Rethink the diagnosis
In the last few months, Ms. S had complained to her primary care provider (PCP) of worsening anxiety and depressed mood. She was the victim of ongoing physical and emotional abuse by her ex-fiancé and was concerned that she may lose custody of her 2 sons. Approximately 8 months before admission, Ms. S’ PCP prescribed lithium, 450 mg, 3 times a day, for “mood stabilization” and depression because she’d already been diagnosed with BD. This was the first mood stabilizer she’d taken since she was 14. She also was taking unknown doses of hydrocodone/acetaminophen, cyclobenzaprine, and tramadol for pain and temazepam for insomnia. Ms. S continued to suffer from labile and depressed mood, and fought with her ex-fiancé and legal authorities to maintain custody of her 2 children until she was found in the park.
Throughout her hospitalization she denies that she attempted suicide that day, and maintains that this incident was caused by unintentional mismanagement of her medications. Although she continues to have a sense of low self-worth, she denies feeling depressed; in contrast, she says she feels like she has a “new lease on life.” During several interviews she cannot provide a history of any prolonged (ie, several days) episodes of elevated mood, increased goal-directed behavior, decreased need for sleep, tangential thought, pressured speech, or other symptoms that suggest hypomania or mania. She does not endorse prolonged periods of neurovegetative symptoms that would indicate a major depressive episode.
We feel that Ms. S’ symptoms of affective dysregulation, impulsivity, and interpersonal dysfunction are consistent with BPD, and we determine that she meets 6 of the 9 DSM-IV-TR diagnostic features of BPD (≥5 are required for a BPD diagnosis) (Table 1).7 Ms. S describes efforts to avoid abandonment, unstable and intense interpersonal relationships, marked and persistent unstable self-image, recurrent suicidal and self-mutilating behavior, affective instability, and chronic feelings of emptiness. She is discharged to follow up with a psychotherapist and family practitioner. She is not continued on any psychotropic medications.
The authors’ observations
Although it can be difficult to accurately diagnose psychiatric illness during a brief inpatient hospitalization, several clinicians who cared for Ms. S felt that her presentation was more consistent with BPD than BD. Her case is an example of the potential harm of incorrectly diagnosing personality-disordered patients with BD. Ms. S is impulsive and used lithium—a medication that is the standard of care for BD—in an overdose, which lead to a costly and dangerous hospitalization marked by a difficult tracheal intubation and hemodialysis.
Table 1
DSM-IV-TR diagnostic criteria for borderline personality disorder
A pervasive pattern of instability of interpersonal relationships, self-image, and affects, and marked impulsivity, as indicated by ≥5 of the following: |
|
Source: Reference 7 |
Distinguishing BD and BPD
There is considerable overlap in symptoms of BD and BPD. Although the episodic nature of BD is well differentiated from the more chronic course of BPD, many hypomania and mania symptoms are similar to those of BPD (Table 2).7 For example, patients with BD or BPD may exhibit impulsive behavior and labile moods. Substance use, risky and self-destructive behaviors, and inflammatory interpersonal relationships can occur in both disorders. Some researchers have suggested that pathophysiologically, BPD may fall on a spectrum of bipolar illness, and have proposed a clinical entity they call bipolar type IV or ultra-rapid cycling BD.2,8,9 There may be more co-occurrence of BD with BPD than would be expected by chance10; 1 review of BPD studies found the rate of comorbid BD ranged from 5.6% to 19%.11 However, because of differences in several factors—including phenomenology, family prevalence, longitudinal course, and medication response—some researchers have concluded that evidence does not support categorizing BPD as part of a bipolar spectrum.10-14 Nonetheless, BPD and other personality disorders often co-occur with axis I disorders, including MDD, BD, or PTSD.
Some research has suggested that the increasing availability and marketing campaigns of medications to treat BD may promote diagnosis of the disorder.15 Zimmerman15 hypothesizes that physicians may be more likely to diagnose a condition that responds to medication (ie, BD) than one that is less responsive (ie, BPD). Financial compensation for treating axis I disorders is significantly better than for treating personality disorders.16 The inpatient setting confers barriers to accurately diagnosing personality disorders, including limits on the amount of time that clinicians can spend with patients or ability to communicate with sources of collateral information. A patient’s observed personality and behaviors while hospitalized may not accurately reflect his or her personality and behaviors in that patient’s “natural” environment.
Several diagnostic strategies can help distinguish BPD from BD. For BD to be the primary diagnosis, a patient must have had a hypomanic or manic episode. Sustained episodes of elation or extreme irritability without evident stressors suggest BD rather than BPD.10 According to Gunderson et al,10 “repeated angry outbursts, suicide attempts, or acts of deliberate self harm that are reactive to interpersonal stress and reflect extreme rejection sensitivity are axiomatic of borderline personality disorder.” In a review of clinical practice, Gunderson17 found that hypersensitivity to rejection and fearful preoccupation with expected abandonment are the most distinctive characteristics of BPD patients. He suggested that clinicians can establish the diagnosis by asking patients directly if they believe the criteria for BPD characterize them, which also can help a patient to accept the diagnosis.
Finally, during a short hospitalization, it can be helpful to obtain collateral information from the patient’s friends and family or further characterize the time course of symptoms and diagnostic features in the patient’s natural environment. Clinicians who are reluctant to diagnose BPD in an inpatient setting could suggest the presence of borderline traits or discuss the possibility of the BPD diagnosis in documentation (eg, in the assessment or formulation). Doing so would avoid a premature BPD diagnosis and allow outpatient providers to confirm or rule out personality disorder diagnoses over time. It is important to screen patients with BPD for co-occurring axis I disorders, including BD, MDD, PTSD, and substance abuse.
A false-positive BD diagnosis in patients with BPD has serious treatment implications. Antipsychotics, antidepressants, and anticonvulsants have been used to target BPD symptoms such as affective dysregulation, impulsivity, and cognitive/perceptual abnormalities, but no medications are FDA-approved for treating BPD. American Psychiatric Association guidelines recommend symptom-based pharmacologic strategies for BPD,18 although some researchers believe that these recommendations are out-of-date and not evidence-based.17,19 Some evidence suggests pharmacotherapy can have modest short-term benefits on specific BPD symptoms, but no data suggest that medication can reduce the severity of BPD or lead to remission.19-23 Just 1 randomized controlled trial (N = 17) has examined lithium for BPD and found no effect on mood.11,24
Misdiagnosis of BD in the context of BPD may create unrealistic expectations regarding the potential efficacy of medications for relieving symptoms. Patients may be diverted from potentially helpful psychotherapeutic treatments—such as DBT or mentalization therapy—which evidence suggests can effectively reduce symptoms, the need for additional treatments, and self-harm or suicidal behaviors.10,17,19 Evidence from long-term longitudinal studies suggests that psychosocial or psychotherapeutic treatment may protect against suicide in BPD patients.25
Table 2
DSM-IV-TR diagnostic criteria for a manic episode
|
The DSM-IV-TR diagnostic criteria for a hypomanic episode are similar to criteria for a manic episode, except:
|
Source: Reference 7 |
Related Resources
- National Education Alliance Borderline Personality Disorder. www.borderlinepersonalitydisorder.com.
- Hoffman PD, Steiner-Grossman P. Borderline personality disorder: meeting the challenges to successful treatment. Philadelphia, PA: Haworth Press; 2008.
Drug Brand Names
- Cyclobenzaprine • Flexeril
- Hydrocodone/acetaminophen • Lorcet, Vicodin, others
- Lithium • Eskalith, Lithobid
- Temazepam • Restoril
- Tramadol • Ultram
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Hirschfeld RM, Cass AR, Holt DC, et al. Screening for bipolar disorder in patients treated for depression in a family medicine clinic. J Am Board Fam Pract. 2005;18(4):233-239.
2. Ghaemi SN, Ko JY, Goodwin FK. “Cade’s disease” and beyond: Misdiagnosis antidepressant use, and a proposed definition for bipolar spectrum disorder. Can J Psychiatry. 2002;47(2):125-134.
3. Bowden CL. Strategies to reduce misdiagnosis of bipolar depression. Psychiatr Serv. 2001;52(1):51-55.
4. Moreno C, Laje G, Blanco C, et al. National trends in the outpatient diagnosis and treatment of bipolar disorder in youth. Arch Gen Psychiatry. 2007;64(9):1032-1039.
5. Zimmerman M, Ruggero CJ, Chelminski I, et al. Is bipolar disorder overdiagnosed? J Clin Psychiatry. 2008;69(6):935-940.
6. Zimmerman M, Ruggero CJ, Chelminski I, et al. Psychiatric diagnoses in patients previously overdiagnosed with bipolar disorder. J Clin Psychiatry. 2010;71(1):26-31.
7. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.
8. Akiskal HS. The bipolar spectrum-the shaping of a new paradigm in psychiatry. Curr Psychiatry Rep. 2002;4(1):1-3.
9. Akiskal HS, Pinto O. The evolving bipolar spectrum. Prototypes I II, III, and IV. Psychiatr Clin North Am. 1999;22(3):517-534, vii.
10. Gunderson JG, Weinberg I, Daversa MT, et al. Descriptive and longitudinal observations on the relationship of borderline personality disorder and bipolar disorder. Am J Psychiatry. 2006;163(7):1173-1178.
11. Paris J, Gunderson J, Weinberg I. The interface between borderline personality disorder and bipolar spectrum disorders. Compr Psychiatry. 2007;48(2):145-154.
12. Paris J. Why psychiatrists are reluctant to diagnose: borderline personality disorder. Psychiatry (Edgmont). 2007;4(1):35-39.
13. Paris J. Borderline or bipolar? Distinguishing borderline personality disorder from bipolar spectrum disorders. Harv Rev Psychiatry. 2004;12(3):140-145.
14. Ruggero CJ, Zimmerman M, Chelminski I, et al. Borderline personality disorder and the misdiagnosis of bipolar disorder. J Psychiatr Res. 2010;44(6):405-408.
15. Zimmerman M. Problems diagnosing bipolar disorder in clinical practice. Expert Rev Neurother. 2010;10(7):1019-1021.
16. Stone MH. Relationship of borderline personality disorder and bipolar disorder. Am J Psychiatry. 2006;163(7):1126-1128.
17. Gunderson JG. Clinical practice. Borderline personality disorder. N Engl J Med. 2011;364(21):2037-2042.
18. American Psychiatric Association. Practice guideline for the treatment of patients with borderline personality disorder. Washington D.C.: American Psychiatric Association; 2001.
19. Paris J. The treatment of borderline personality disorder: implications of research on diagnosis etiology, and outcome. Annu Rev Clin Psychol. 2009;5:277-290.
20. Stoffers J, Völlm BA, Rücker G, et al. Pharmacological interventions for borderline personality disorder. Cochrane Database Syst Rev. 2010;(6):CD005653.-
21. Ripoll LH, Triebwasser J, Siever LJ. Evidence-based pharmacotherapy for personality disorders. Int J Neuropsychopharmacol. 2011;14(9):1257-1288.
22. Mercer D, Douglass AB, Links PS. Meta-analyses of mood stabilizers antidepressants and antipsychotics in the treatment of borderline personality disorder: effectiveness for depression and anger symptoms. J Pers Disord. 2009;23(2):156-174.
23. Lieb K, Völlm B, Rücker G, et al. Pharmacotherapy for borderline personality disorder: Cochrane systematic review of randomised trials. Br J Psychiatry. 2010;196(1):4-12.
24. Links PS, Steiner M, Boiago I, et al. Lithium therapy for borderline patients: preliminary findings. J Pers Disord. 1990;4(2):173-181.
25. Goodman M, Roiff T, Oakes AH, et al. Suicidal risk and management in borderline personality disorder. Curr Psychiatry Rep. 2012;14(1):79-85.
CASE: Unclear diagnosis
Police find Ms. S, age 31, extremely intoxicated and drinking alcohol in her car in a city park parking lot. In the emergency room, she becomes increasingly somnolent and clinicians intubate her trachea to protect her airway. Lab testing shows she has elevated acetaminophen and lithium serum levels, and she is transferred to our hospital for further management after being started on N-acetylcysteine to treat acetaminophen toxicity. Her “ex-fiancé,” the father of her 2 children, saw her earlier the day of the episode and says she was distraught, intoxicated, and had several empty pill bottles in her purse.
In our hospital, Ms. S’ lithium level increases from 2.3 mEq/L to a peak of 5.32 mEq/L, and she undergoes hemodialysis. On hospital day 2, her serum lithium level is trending downward. After Ms. S is able to breathe spontaneously, her trachea is extubated and her hemodialysis line is removed. A psychiatric consultation is obtained, but she is unable to provide a coherent history and the treating clinicians believe she has delirium caused by multiple factors.
On hospital day 3, Ms. S’ delirium clears enough for her to engage in an interview, and she is transferred to our inpatient psychiatry ward for further monitoring and stabilization.
She reports that she was diagnosed with bipolar disorder (BD) at age 12, when she faced multiple psychosocial stressors, including physical abuse by her mother’s boyfriend. She took several psychotropics—although she cannot remember which ones—until age 14, when she stopped all medications until the year before her current hospitalization. Although throughout adolescence and adulthood Ms. S experienced chronic irritability, anxiety, impulsive behavior, poor self-esteem, abusive relationships, self-cutting, and depressed mood, she maintains that she felt worse when she was taking psychotropics and doubts the BD diagnosis. She attributes her longstanding mood issues to low self-worth, a “codependent nature,” and a tendency to gravitate toward abusive relationships. Although she admits to experimenting with several illicit drugs during adolescence, she denies more recent substance use and states she drinks alcohol only once every few months.
The authors’ observations
BD is underdiagnosed in several patient populations, such as individuals previously diagnosed with MDD.1-3 Misdiagnosis can have severe implications, including delay in receiving treatment with effective medications (eg, mood stabilizers) or use of agents that can induce mania or rapid-cycling, such as antidepressants. Perhaps in response to this concern, in recent years clinicians increasingly have diagnosed BD in adolescents and adults. An analysis of a national database of physician practices found a 40-fold increase in office visits for BD among youth and a near doubling among adults from 1994 to 2003.4
Although underdiagnosis of BD remains important, some researchers have suggested that overdiagnosis may be more prevalent and equally harmful. In a study of 180 patients being treated for depression in a family care clinic, there was a 21.6% initial underdiagnosis rate among those eventually found to have BD.1 However, among 43 patients with a prior BD diagnosis, the diagnosis was not confirmed in 33%.1 In a study of 700 psychiatric outpatients in Rhode Island, only 43% of 145 patients who reported a prior BD diagnosis had that diagnosis confirmed.5 Three times as many patients were overdiagnosed with BD as underdiagnosed.
Are there characteristics common to individuals incorrectly diagnosed with BD? In a study that compared patients who had been mistakenly diagnosed with BD with those who had not been diagnosed with BD, the overdiagnosis group was significantly more likely to be diagnosed with a personality disorder, in particular borderline or antisocial personality disorder.6 Only lifetime and current BPD, current posttraumatic stress disorder (PTSD), and lifetime impulse control disorders were independently associated with BD overdiagnosis. The odds ratio for overdiagnosis of BD in patients found to have BPD was 3.7.
EVALUATION: Rethink the diagnosis
In the last few months, Ms. S had complained to her primary care provider (PCP) of worsening anxiety and depressed mood. She was the victim of ongoing physical and emotional abuse by her ex-fiancé and was concerned that she may lose custody of her 2 sons. Approximately 8 months before admission, Ms. S’ PCP prescribed lithium, 450 mg, 3 times a day, for “mood stabilization” and depression because she’d already been diagnosed with BD. This was the first mood stabilizer she’d taken since she was 14. She also was taking unknown doses of hydrocodone/acetaminophen, cyclobenzaprine, and tramadol for pain and temazepam for insomnia. Ms. S continued to suffer from labile and depressed mood, and fought with her ex-fiancé and legal authorities to maintain custody of her 2 children until she was found in the park.
Throughout her hospitalization she denies that she attempted suicide that day, and maintains that this incident was caused by unintentional mismanagement of her medications. Although she continues to have a sense of low self-worth, she denies feeling depressed; in contrast, she says she feels like she has a “new lease on life.” During several interviews she cannot provide a history of any prolonged (ie, several days) episodes of elevated mood, increased goal-directed behavior, decreased need for sleep, tangential thought, pressured speech, or other symptoms that suggest hypomania or mania. She does not endorse prolonged periods of neurovegetative symptoms that would indicate a major depressive episode.
We feel that Ms. S’ symptoms of affective dysregulation, impulsivity, and interpersonal dysfunction are consistent with BPD, and we determine that she meets 6 of the 9 DSM-IV-TR diagnostic features of BPD (≥5 are required for a BPD diagnosis) (Table 1).7 Ms. S describes efforts to avoid abandonment, unstable and intense interpersonal relationships, marked and persistent unstable self-image, recurrent suicidal and self-mutilating behavior, affective instability, and chronic feelings of emptiness. She is discharged to follow up with a psychotherapist and family practitioner. She is not continued on any psychotropic medications.
The authors’ observations
Although it can be difficult to accurately diagnose psychiatric illness during a brief inpatient hospitalization, several clinicians who cared for Ms. S felt that her presentation was more consistent with BPD than BD. Her case is an example of the potential harm of incorrectly diagnosing personality-disordered patients with BD. Ms. S is impulsive and used lithium—a medication that is the standard of care for BD—in an overdose, which lead to a costly and dangerous hospitalization marked by a difficult tracheal intubation and hemodialysis.
Table 1
DSM-IV-TR diagnostic criteria for borderline personality disorder
A pervasive pattern of instability of interpersonal relationships, self-image, and affects, and marked impulsivity, as indicated by ≥5 of the following: |
|
Source: Reference 7 |
Distinguishing BD and BPD
There is considerable overlap in symptoms of BD and BPD. Although the episodic nature of BD is well differentiated from the more chronic course of BPD, many hypomania and mania symptoms are similar to those of BPD (Table 2).7 For example, patients with BD or BPD may exhibit impulsive behavior and labile moods. Substance use, risky and self-destructive behaviors, and inflammatory interpersonal relationships can occur in both disorders. Some researchers have suggested that pathophysiologically, BPD may fall on a spectrum of bipolar illness, and have proposed a clinical entity they call bipolar type IV or ultra-rapid cycling BD.2,8,9 There may be more co-occurrence of BD with BPD than would be expected by chance10; 1 review of BPD studies found the rate of comorbid BD ranged from 5.6% to 19%.11 However, because of differences in several factors—including phenomenology, family prevalence, longitudinal course, and medication response—some researchers have concluded that evidence does not support categorizing BPD as part of a bipolar spectrum.10-14 Nonetheless, BPD and other personality disorders often co-occur with axis I disorders, including MDD, BD, or PTSD.
Some research has suggested that the increasing availability and marketing campaigns of medications to treat BD may promote diagnosis of the disorder.15 Zimmerman15 hypothesizes that physicians may be more likely to diagnose a condition that responds to medication (ie, BD) than one that is less responsive (ie, BPD). Financial compensation for treating axis I disorders is significantly better than for treating personality disorders.16 The inpatient setting confers barriers to accurately diagnosing personality disorders, including limits on the amount of time that clinicians can spend with patients or ability to communicate with sources of collateral information. A patient’s observed personality and behaviors while hospitalized may not accurately reflect his or her personality and behaviors in that patient’s “natural” environment.
Several diagnostic strategies can help distinguish BPD from BD. For BD to be the primary diagnosis, a patient must have had a hypomanic or manic episode. Sustained episodes of elation or extreme irritability without evident stressors suggest BD rather than BPD.10 According to Gunderson et al,10 “repeated angry outbursts, suicide attempts, or acts of deliberate self harm that are reactive to interpersonal stress and reflect extreme rejection sensitivity are axiomatic of borderline personality disorder.” In a review of clinical practice, Gunderson17 found that hypersensitivity to rejection and fearful preoccupation with expected abandonment are the most distinctive characteristics of BPD patients. He suggested that clinicians can establish the diagnosis by asking patients directly if they believe the criteria for BPD characterize them, which also can help a patient to accept the diagnosis.
Finally, during a short hospitalization, it can be helpful to obtain collateral information from the patient’s friends and family or further characterize the time course of symptoms and diagnostic features in the patient’s natural environment. Clinicians who are reluctant to diagnose BPD in an inpatient setting could suggest the presence of borderline traits or discuss the possibility of the BPD diagnosis in documentation (eg, in the assessment or formulation). Doing so would avoid a premature BPD diagnosis and allow outpatient providers to confirm or rule out personality disorder diagnoses over time. It is important to screen patients with BPD for co-occurring axis I disorders, including BD, MDD, PTSD, and substance abuse.
A false-positive BD diagnosis in patients with BPD has serious treatment implications. Antipsychotics, antidepressants, and anticonvulsants have been used to target BPD symptoms such as affective dysregulation, impulsivity, and cognitive/perceptual abnormalities, but no medications are FDA-approved for treating BPD. American Psychiatric Association guidelines recommend symptom-based pharmacologic strategies for BPD,18 although some researchers believe that these recommendations are out-of-date and not evidence-based.17,19 Some evidence suggests pharmacotherapy can have modest short-term benefits on specific BPD symptoms, but no data suggest that medication can reduce the severity of BPD or lead to remission.19-23 Just 1 randomized controlled trial (N = 17) has examined lithium for BPD and found no effect on mood.11,24
Misdiagnosis of BD in the context of BPD may create unrealistic expectations regarding the potential efficacy of medications for relieving symptoms. Patients may be diverted from potentially helpful psychotherapeutic treatments—such as DBT or mentalization therapy—which evidence suggests can effectively reduce symptoms, the need for additional treatments, and self-harm or suicidal behaviors.10,17,19 Evidence from long-term longitudinal studies suggests that psychosocial or psychotherapeutic treatment may protect against suicide in BPD patients.25
Table 2
DSM-IV-TR diagnostic criteria for a manic episode
|
The DSM-IV-TR diagnostic criteria for a hypomanic episode are similar to criteria for a manic episode, except:
|
Source: Reference 7 |
Related Resources
- National Education Alliance Borderline Personality Disorder. www.borderlinepersonalitydisorder.com.
- Hoffman PD, Steiner-Grossman P. Borderline personality disorder: meeting the challenges to successful treatment. Philadelphia, PA: Haworth Press; 2008.
Drug Brand Names
- Cyclobenzaprine • Flexeril
- Hydrocodone/acetaminophen • Lorcet, Vicodin, others
- Lithium • Eskalith, Lithobid
- Temazepam • Restoril
- Tramadol • Ultram
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
CASE: Unclear diagnosis
Police find Ms. S, age 31, extremely intoxicated and drinking alcohol in her car in a city park parking lot. In the emergency room, she becomes increasingly somnolent and clinicians intubate her trachea to protect her airway. Lab testing shows she has elevated acetaminophen and lithium serum levels, and she is transferred to our hospital for further management after being started on N-acetylcysteine to treat acetaminophen toxicity. Her “ex-fiancé,” the father of her 2 children, saw her earlier the day of the episode and says she was distraught, intoxicated, and had several empty pill bottles in her purse.
In our hospital, Ms. S’ lithium level increases from 2.3 mEq/L to a peak of 5.32 mEq/L, and she undergoes hemodialysis. On hospital day 2, her serum lithium level is trending downward. After Ms. S is able to breathe spontaneously, her trachea is extubated and her hemodialysis line is removed. A psychiatric consultation is obtained, but she is unable to provide a coherent history and the treating clinicians believe she has delirium caused by multiple factors.
On hospital day 3, Ms. S’ delirium clears enough for her to engage in an interview, and she is transferred to our inpatient psychiatry ward for further monitoring and stabilization.
She reports that she was diagnosed with bipolar disorder (BD) at age 12, when she faced multiple psychosocial stressors, including physical abuse by her mother’s boyfriend. She took several psychotropics—although she cannot remember which ones—until age 14, when she stopped all medications until the year before her current hospitalization. Although throughout adolescence and adulthood Ms. S experienced chronic irritability, anxiety, impulsive behavior, poor self-esteem, abusive relationships, self-cutting, and depressed mood, she maintains that she felt worse when she was taking psychotropics and doubts the BD diagnosis. She attributes her longstanding mood issues to low self-worth, a “codependent nature,” and a tendency to gravitate toward abusive relationships. Although she admits to experimenting with several illicit drugs during adolescence, she denies more recent substance use and states she drinks alcohol only once every few months.
The authors’ observations
BD is underdiagnosed in several patient populations, such as individuals previously diagnosed with MDD.1-3 Misdiagnosis can have severe implications, including delay in receiving treatment with effective medications (eg, mood stabilizers) or use of agents that can induce mania or rapid-cycling, such as antidepressants. Perhaps in response to this concern, in recent years clinicians increasingly have diagnosed BD in adolescents and adults. An analysis of a national database of physician practices found a 40-fold increase in office visits for BD among youth and a near doubling among adults from 1994 to 2003.4
Although underdiagnosis of BD remains important, some researchers have suggested that overdiagnosis may be more prevalent and equally harmful. In a study of 180 patients being treated for depression in a family care clinic, there was a 21.6% initial underdiagnosis rate among those eventually found to have BD.1 However, among 43 patients with a prior BD diagnosis, the diagnosis was not confirmed in 33%.1 In a study of 700 psychiatric outpatients in Rhode Island, only 43% of 145 patients who reported a prior BD diagnosis had that diagnosis confirmed.5 Three times as many patients were overdiagnosed with BD as underdiagnosed.
Are there characteristics common to individuals incorrectly diagnosed with BD? In a study that compared patients who had been mistakenly diagnosed with BD with those who had not been diagnosed with BD, the overdiagnosis group was significantly more likely to be diagnosed with a personality disorder, in particular borderline or antisocial personality disorder.6 Only lifetime and current BPD, current posttraumatic stress disorder (PTSD), and lifetime impulse control disorders were independently associated with BD overdiagnosis. The odds ratio for overdiagnosis of BD in patients found to have BPD was 3.7.
EVALUATION: Rethink the diagnosis
In the last few months, Ms. S had complained to her primary care provider (PCP) of worsening anxiety and depressed mood. She was the victim of ongoing physical and emotional abuse by her ex-fiancé and was concerned that she may lose custody of her 2 sons. Approximately 8 months before admission, Ms. S’ PCP prescribed lithium, 450 mg, 3 times a day, for “mood stabilization” and depression because she’d already been diagnosed with BD. This was the first mood stabilizer she’d taken since she was 14. She also was taking unknown doses of hydrocodone/acetaminophen, cyclobenzaprine, and tramadol for pain and temazepam for insomnia. Ms. S continued to suffer from labile and depressed mood, and fought with her ex-fiancé and legal authorities to maintain custody of her 2 children until she was found in the park.
Throughout her hospitalization she denies that she attempted suicide that day, and maintains that this incident was caused by unintentional mismanagement of her medications. Although she continues to have a sense of low self-worth, she denies feeling depressed; in contrast, she says she feels like she has a “new lease on life.” During several interviews she cannot provide a history of any prolonged (ie, several days) episodes of elevated mood, increased goal-directed behavior, decreased need for sleep, tangential thought, pressured speech, or other symptoms that suggest hypomania or mania. She does not endorse prolonged periods of neurovegetative symptoms that would indicate a major depressive episode.
We feel that Ms. S’ symptoms of affective dysregulation, impulsivity, and interpersonal dysfunction are consistent with BPD, and we determine that she meets 6 of the 9 DSM-IV-TR diagnostic features of BPD (≥5 are required for a BPD diagnosis) (Table 1).7 Ms. S describes efforts to avoid abandonment, unstable and intense interpersonal relationships, marked and persistent unstable self-image, recurrent suicidal and self-mutilating behavior, affective instability, and chronic feelings of emptiness. She is discharged to follow up with a psychotherapist and family practitioner. She is not continued on any psychotropic medications.
The authors’ observations
Although it can be difficult to accurately diagnose psychiatric illness during a brief inpatient hospitalization, several clinicians who cared for Ms. S felt that her presentation was more consistent with BPD than BD. Her case is an example of the potential harm of incorrectly diagnosing personality-disordered patients with BD. Ms. S is impulsive and used lithium—a medication that is the standard of care for BD—in an overdose, which lead to a costly and dangerous hospitalization marked by a difficult tracheal intubation and hemodialysis.
Table 1
DSM-IV-TR diagnostic criteria for borderline personality disorder
A pervasive pattern of instability of interpersonal relationships, self-image, and affects, and marked impulsivity, as indicated by ≥5 of the following: |
|
Source: Reference 7 |
Distinguishing BD and BPD
There is considerable overlap in symptoms of BD and BPD. Although the episodic nature of BD is well differentiated from the more chronic course of BPD, many hypomania and mania symptoms are similar to those of BPD (Table 2).7 For example, patients with BD or BPD may exhibit impulsive behavior and labile moods. Substance use, risky and self-destructive behaviors, and inflammatory interpersonal relationships can occur in both disorders. Some researchers have suggested that pathophysiologically, BPD may fall on a spectrum of bipolar illness, and have proposed a clinical entity they call bipolar type IV or ultra-rapid cycling BD.2,8,9 There may be more co-occurrence of BD with BPD than would be expected by chance10; 1 review of BPD studies found the rate of comorbid BD ranged from 5.6% to 19%.11 However, because of differences in several factors—including phenomenology, family prevalence, longitudinal course, and medication response—some researchers have concluded that evidence does not support categorizing BPD as part of a bipolar spectrum.10-14 Nonetheless, BPD and other personality disorders often co-occur with axis I disorders, including MDD, BD, or PTSD.
Some research has suggested that the increasing availability and marketing campaigns of medications to treat BD may promote diagnosis of the disorder.15 Zimmerman15 hypothesizes that physicians may be more likely to diagnose a condition that responds to medication (ie, BD) than one that is less responsive (ie, BPD). Financial compensation for treating axis I disorders is significantly better than for treating personality disorders.16 The inpatient setting confers barriers to accurately diagnosing personality disorders, including limits on the amount of time that clinicians can spend with patients or ability to communicate with sources of collateral information. A patient’s observed personality and behaviors while hospitalized may not accurately reflect his or her personality and behaviors in that patient’s “natural” environment.
Several diagnostic strategies can help distinguish BPD from BD. For BD to be the primary diagnosis, a patient must have had a hypomanic or manic episode. Sustained episodes of elation or extreme irritability without evident stressors suggest BD rather than BPD.10 According to Gunderson et al,10 “repeated angry outbursts, suicide attempts, or acts of deliberate self harm that are reactive to interpersonal stress and reflect extreme rejection sensitivity are axiomatic of borderline personality disorder.” In a review of clinical practice, Gunderson17 found that hypersensitivity to rejection and fearful preoccupation with expected abandonment are the most distinctive characteristics of BPD patients. He suggested that clinicians can establish the diagnosis by asking patients directly if they believe the criteria for BPD characterize them, which also can help a patient to accept the diagnosis.
Finally, during a short hospitalization, it can be helpful to obtain collateral information from the patient’s friends and family or further characterize the time course of symptoms and diagnostic features in the patient’s natural environment. Clinicians who are reluctant to diagnose BPD in an inpatient setting could suggest the presence of borderline traits or discuss the possibility of the BPD diagnosis in documentation (eg, in the assessment or formulation). Doing so would avoid a premature BPD diagnosis and allow outpatient providers to confirm or rule out personality disorder diagnoses over time. It is important to screen patients with BPD for co-occurring axis I disorders, including BD, MDD, PTSD, and substance abuse.
A false-positive BD diagnosis in patients with BPD has serious treatment implications. Antipsychotics, antidepressants, and anticonvulsants have been used to target BPD symptoms such as affective dysregulation, impulsivity, and cognitive/perceptual abnormalities, but no medications are FDA-approved for treating BPD. American Psychiatric Association guidelines recommend symptom-based pharmacologic strategies for BPD,18 although some researchers believe that these recommendations are out-of-date and not evidence-based.17,19 Some evidence suggests pharmacotherapy can have modest short-term benefits on specific BPD symptoms, but no data suggest that medication can reduce the severity of BPD or lead to remission.19-23 Just 1 randomized controlled trial (N = 17) has examined lithium for BPD and found no effect on mood.11,24
Misdiagnosis of BD in the context of BPD may create unrealistic expectations regarding the potential efficacy of medications for relieving symptoms. Patients may be diverted from potentially helpful psychotherapeutic treatments—such as DBT or mentalization therapy—which evidence suggests can effectively reduce symptoms, the need for additional treatments, and self-harm or suicidal behaviors.10,17,19 Evidence from long-term longitudinal studies suggests that psychosocial or psychotherapeutic treatment may protect against suicide in BPD patients.25
Table 2
DSM-IV-TR diagnostic criteria for a manic episode
|
The DSM-IV-TR diagnostic criteria for a hypomanic episode are similar to criteria for a manic episode, except:
|
Source: Reference 7 |
Related Resources
- National Education Alliance Borderline Personality Disorder. www.borderlinepersonalitydisorder.com.
- Hoffman PD, Steiner-Grossman P. Borderline personality disorder: meeting the challenges to successful treatment. Philadelphia, PA: Haworth Press; 2008.
Drug Brand Names
- Cyclobenzaprine • Flexeril
- Hydrocodone/acetaminophen • Lorcet, Vicodin, others
- Lithium • Eskalith, Lithobid
- Temazepam • Restoril
- Tramadol • Ultram
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Hirschfeld RM, Cass AR, Holt DC, et al. Screening for bipolar disorder in patients treated for depression in a family medicine clinic. J Am Board Fam Pract. 2005;18(4):233-239.
2. Ghaemi SN, Ko JY, Goodwin FK. “Cade’s disease” and beyond: Misdiagnosis antidepressant use, and a proposed definition for bipolar spectrum disorder. Can J Psychiatry. 2002;47(2):125-134.
3. Bowden CL. Strategies to reduce misdiagnosis of bipolar depression. Psychiatr Serv. 2001;52(1):51-55.
4. Moreno C, Laje G, Blanco C, et al. National trends in the outpatient diagnosis and treatment of bipolar disorder in youth. Arch Gen Psychiatry. 2007;64(9):1032-1039.
5. Zimmerman M, Ruggero CJ, Chelminski I, et al. Is bipolar disorder overdiagnosed? J Clin Psychiatry. 2008;69(6):935-940.
6. Zimmerman M, Ruggero CJ, Chelminski I, et al. Psychiatric diagnoses in patients previously overdiagnosed with bipolar disorder. J Clin Psychiatry. 2010;71(1):26-31.
7. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.
8. Akiskal HS. The bipolar spectrum-the shaping of a new paradigm in psychiatry. Curr Psychiatry Rep. 2002;4(1):1-3.
9. Akiskal HS, Pinto O. The evolving bipolar spectrum. Prototypes I II, III, and IV. Psychiatr Clin North Am. 1999;22(3):517-534, vii.
10. Gunderson JG, Weinberg I, Daversa MT, et al. Descriptive and longitudinal observations on the relationship of borderline personality disorder and bipolar disorder. Am J Psychiatry. 2006;163(7):1173-1178.
11. Paris J, Gunderson J, Weinberg I. The interface between borderline personality disorder and bipolar spectrum disorders. Compr Psychiatry. 2007;48(2):145-154.
12. Paris J. Why psychiatrists are reluctant to diagnose: borderline personality disorder. Psychiatry (Edgmont). 2007;4(1):35-39.
13. Paris J. Borderline or bipolar? Distinguishing borderline personality disorder from bipolar spectrum disorders. Harv Rev Psychiatry. 2004;12(3):140-145.
14. Ruggero CJ, Zimmerman M, Chelminski I, et al. Borderline personality disorder and the misdiagnosis of bipolar disorder. J Psychiatr Res. 2010;44(6):405-408.
15. Zimmerman M. Problems diagnosing bipolar disorder in clinical practice. Expert Rev Neurother. 2010;10(7):1019-1021.
16. Stone MH. Relationship of borderline personality disorder and bipolar disorder. Am J Psychiatry. 2006;163(7):1126-1128.
17. Gunderson JG. Clinical practice. Borderline personality disorder. N Engl J Med. 2011;364(21):2037-2042.
18. American Psychiatric Association. Practice guideline for the treatment of patients with borderline personality disorder. Washington D.C.: American Psychiatric Association; 2001.
19. Paris J. The treatment of borderline personality disorder: implications of research on diagnosis etiology, and outcome. Annu Rev Clin Psychol. 2009;5:277-290.
20. Stoffers J, Völlm BA, Rücker G, et al. Pharmacological interventions for borderline personality disorder. Cochrane Database Syst Rev. 2010;(6):CD005653.-
21. Ripoll LH, Triebwasser J, Siever LJ. Evidence-based pharmacotherapy for personality disorders. Int J Neuropsychopharmacol. 2011;14(9):1257-1288.
22. Mercer D, Douglass AB, Links PS. Meta-analyses of mood stabilizers antidepressants and antipsychotics in the treatment of borderline personality disorder: effectiveness for depression and anger symptoms. J Pers Disord. 2009;23(2):156-174.
23. Lieb K, Völlm B, Rücker G, et al. Pharmacotherapy for borderline personality disorder: Cochrane systematic review of randomised trials. Br J Psychiatry. 2010;196(1):4-12.
24. Links PS, Steiner M, Boiago I, et al. Lithium therapy for borderline patients: preliminary findings. J Pers Disord. 1990;4(2):173-181.
25. Goodman M, Roiff T, Oakes AH, et al. Suicidal risk and management in borderline personality disorder. Curr Psychiatry Rep. 2012;14(1):79-85.
1. Hirschfeld RM, Cass AR, Holt DC, et al. Screening for bipolar disorder in patients treated for depression in a family medicine clinic. J Am Board Fam Pract. 2005;18(4):233-239.
2. Ghaemi SN, Ko JY, Goodwin FK. “Cade’s disease” and beyond: Misdiagnosis antidepressant use, and a proposed definition for bipolar spectrum disorder. Can J Psychiatry. 2002;47(2):125-134.
3. Bowden CL. Strategies to reduce misdiagnosis of bipolar depression. Psychiatr Serv. 2001;52(1):51-55.
4. Moreno C, Laje G, Blanco C, et al. National trends in the outpatient diagnosis and treatment of bipolar disorder in youth. Arch Gen Psychiatry. 2007;64(9):1032-1039.
5. Zimmerman M, Ruggero CJ, Chelminski I, et al. Is bipolar disorder overdiagnosed? J Clin Psychiatry. 2008;69(6):935-940.
6. Zimmerman M, Ruggero CJ, Chelminski I, et al. Psychiatric diagnoses in patients previously overdiagnosed with bipolar disorder. J Clin Psychiatry. 2010;71(1):26-31.
7. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.
8. Akiskal HS. The bipolar spectrum-the shaping of a new paradigm in psychiatry. Curr Psychiatry Rep. 2002;4(1):1-3.
9. Akiskal HS, Pinto O. The evolving bipolar spectrum. Prototypes I II, III, and IV. Psychiatr Clin North Am. 1999;22(3):517-534, vii.
10. Gunderson JG, Weinberg I, Daversa MT, et al. Descriptive and longitudinal observations on the relationship of borderline personality disorder and bipolar disorder. Am J Psychiatry. 2006;163(7):1173-1178.
11. Paris J, Gunderson J, Weinberg I. The interface between borderline personality disorder and bipolar spectrum disorders. Compr Psychiatry. 2007;48(2):145-154.
12. Paris J. Why psychiatrists are reluctant to diagnose: borderline personality disorder. Psychiatry (Edgmont). 2007;4(1):35-39.
13. Paris J. Borderline or bipolar? Distinguishing borderline personality disorder from bipolar spectrum disorders. Harv Rev Psychiatry. 2004;12(3):140-145.
14. Ruggero CJ, Zimmerman M, Chelminski I, et al. Borderline personality disorder and the misdiagnosis of bipolar disorder. J Psychiatr Res. 2010;44(6):405-408.
15. Zimmerman M. Problems diagnosing bipolar disorder in clinical practice. Expert Rev Neurother. 2010;10(7):1019-1021.
16. Stone MH. Relationship of borderline personality disorder and bipolar disorder. Am J Psychiatry. 2006;163(7):1126-1128.
17. Gunderson JG. Clinical practice. Borderline personality disorder. N Engl J Med. 2011;364(21):2037-2042.
18. American Psychiatric Association. Practice guideline for the treatment of patients with borderline personality disorder. Washington D.C.: American Psychiatric Association; 2001.
19. Paris J. The treatment of borderline personality disorder: implications of research on diagnosis etiology, and outcome. Annu Rev Clin Psychol. 2009;5:277-290.
20. Stoffers J, Völlm BA, Rücker G, et al. Pharmacological interventions for borderline personality disorder. Cochrane Database Syst Rev. 2010;(6):CD005653.-
21. Ripoll LH, Triebwasser J, Siever LJ. Evidence-based pharmacotherapy for personality disorders. Int J Neuropsychopharmacol. 2011;14(9):1257-1288.
22. Mercer D, Douglass AB, Links PS. Meta-analyses of mood stabilizers antidepressants and antipsychotics in the treatment of borderline personality disorder: effectiveness for depression and anger symptoms. J Pers Disord. 2009;23(2):156-174.
23. Lieb K, Völlm B, Rücker G, et al. Pharmacotherapy for borderline personality disorder: Cochrane systematic review of randomised trials. Br J Psychiatry. 2010;196(1):4-12.
24. Links PS, Steiner M, Boiago I, et al. Lithium therapy for borderline patients: preliminary findings. J Pers Disord. 1990;4(2):173-181.
25. Goodman M, Roiff T, Oakes AH, et al. Suicidal risk and management in borderline personality disorder. Curr Psychiatry Rep. 2012;14(1):79-85.
Some psychiatric disorders hamper ability to get pregnant
Some psychiatric disorders appear to be associated with lower fecundity in both men and women, suggesting that natural selection attempts to discourage the perpetuation of genetic variants associated with them.
Instead, new mutations could be one reason that the disorders continue to exist, Robert A. Power and his colleagues wrote in the January issue of JAMA Psychiatry (formerly Archives of General Psychiatry).
The authors also found that psychiatric disorders affected men’s fecundity more than women’s. "This sex-specific effect suggests that psychiatric morbidity impairs interest or ability to find suitable mating partners or inhibits biological fertility to a greater extent in men," wrote Mr. Power of Kings College, London.
The study data were extracted from two of Sweden’s population registries – the Multi-Generation Register and the Swedish Hospital Discharge Register. More than 2.3 million people born from 1950-1970 were cross-linked by individual patient identification numbers, which allowed the researchers to trace not only patients but also their siblings. At the time of the analysis, no patient was younger than 40 years (JAMA Psychiatry 2013;70:22-30).
The authors tracked fecundity in about 177,000 patients who had schizophrenia, autism, bipolar disorder, anorexia nervosa, or substance abuse. Rates were compared with fecundity in 261,000 siblings. The researchers then compared these rates with those found in the general population.
About 19,000 patients had schizophrenia. They had significantly fewer children than the general population’s (fecundity rate [FR] 0.23 for men and 0.47 for women). In a univariate analysis, sisters of the patients had a significantly higher fecundity rate (FR, 1.02), but this difference disappeared once comorbidities were factored into the analysis. Brothers also had significantly decreased fecundity (FR, 0.97).
"Our results suggest a strong selection pressure to remove genetic variants associated with schizophrenia from the population," the authors said. "This is further evidence for the role of recent or de novo mutations in the genetic susceptibility to schizophrenia that has neither reached the frequency of nor existed long enough to be removed from the population."
Autism was present in 2,947 patients; these had 4,471 siblings. Fecundity was significantly lower in both men and women (FR, 0.25 and 0.48, respectively). Among the siblings, brothers also had fewer children (FR, 0.94). Among sisters, the rate was not significantly different than the general population.
"Individuals with autism showed the greatest reduction in fecundity among all examined disorders. This was not unexpected because previous investigations have shown that few individuals with autism ever married or had children.
"We propose that rare highly deleterious variants and sexually antagonistic polymorphisms may contribute to the genetic disposition to autism. The similarity to schizophrenia is notable because it has been proposed that the autistic and psychotic spectrums reflect two extremes of social cognition."
Bipolar disorder was present in 14,439 patients, among whom were 22,986 siblings. Fecundity was significantly lower than the general population in both men (FR, 0.75) and women (FR, 0.85). While brothers had similar rates to that of the general population, sisters had significantly more children (FR, 1.03). But incorporating comorbidities into the analysis changed the significance for both patients and sisters, with the patient rate increasing to just below that of the general population (FR, 0.94), and the sisters’ rate increased rate no longer being significant (FR, 0.95).
"It has been suggested that the introduction of lithium as a treatment for bipolar disorder has led to improved functioning and, as a result, greater fecundity in those populations where treatment is available."
There were 81,295 patients with depression, among who were 119,645 siblings. While men with depression had significantly lower rates (FR, 0.93), women with the disorder were not significantly different than the general population. Siblings had significantly more children than the general population (FR, brothers 1.01, sisters 1.04) – a difference that was unchanged by factoring in comorbidities. The addition of comorbidities to the analysis did not change the decreased fecundity rate for male patients, but actually increased the rate for female patients (FR, 1.03).
"Notably, depression was an exception to the five other studied disorders ... genes associated with depression seems to be maintained in the population by balancing selection because the cost to affected individuals is roughly equal to the benefit to their siblings. If this is the case, it would be the first strong evidence for balancing selection in a psychiatric disorder."
There were 3,275 patients with anorexia, who had a total of 5,172 siblings. Both men and women with the disorder had significantly reduced fecundity (0.54 and 0.58, respectively). In the sibling group, there were no significant differences for either brothers or sisters. None of the findings changed when comorbidities were factored in.
"Our calculations suggest that anorexia is under weaker negative selection relative to schizophrenia and autism," the authors said.
Substance abuse was present in 55,933 patients, who had a total of 81,592 siblings. The fecundity rate was significantly lower in both men and women (FR, 0.78 and 0.93, respectively). Siblings had significantly more children than the general population (FR, 1.03 for brothers and 1.05 for sisters).
"Our findings suggest that this increased fecundity in siblings almost entirely accounts for the cost to affected individuals, with only a slight decrease in the frequency of these individuals’ genes predicted each generation. Considering that most drugs are a new environmental exposure when seen from an evolutionary perspective, it is possible that there has been insufficient time for selection to act on risk alleles. ... It has also been suggested that substance abuse is associated with risk-taking behavior in both sexes, including sexual risk taking."
The study was funded by the Medical Research Council of the United Kingdom. One of the coauthors reported having received consulting fees and honoraria from GlaxoSmithKline and Lundbeck.
Some psychiatric disorders appear to be associated with lower fecundity in both men and women, suggesting that natural selection attempts to discourage the perpetuation of genetic variants associated with them.
Instead, new mutations could be one reason that the disorders continue to exist, Robert A. Power and his colleagues wrote in the January issue of JAMA Psychiatry (formerly Archives of General Psychiatry).
The authors also found that psychiatric disorders affected men’s fecundity more than women’s. "This sex-specific effect suggests that psychiatric morbidity impairs interest or ability to find suitable mating partners or inhibits biological fertility to a greater extent in men," wrote Mr. Power of Kings College, London.
The study data were extracted from two of Sweden’s population registries – the Multi-Generation Register and the Swedish Hospital Discharge Register. More than 2.3 million people born from 1950-1970 were cross-linked by individual patient identification numbers, which allowed the researchers to trace not only patients but also their siblings. At the time of the analysis, no patient was younger than 40 years (JAMA Psychiatry 2013;70:22-30).
The authors tracked fecundity in about 177,000 patients who had schizophrenia, autism, bipolar disorder, anorexia nervosa, or substance abuse. Rates were compared with fecundity in 261,000 siblings. The researchers then compared these rates with those found in the general population.
About 19,000 patients had schizophrenia. They had significantly fewer children than the general population’s (fecundity rate [FR] 0.23 for men and 0.47 for women). In a univariate analysis, sisters of the patients had a significantly higher fecundity rate (FR, 1.02), but this difference disappeared once comorbidities were factored into the analysis. Brothers also had significantly decreased fecundity (FR, 0.97).
"Our results suggest a strong selection pressure to remove genetic variants associated with schizophrenia from the population," the authors said. "This is further evidence for the role of recent or de novo mutations in the genetic susceptibility to schizophrenia that has neither reached the frequency of nor existed long enough to be removed from the population."
Autism was present in 2,947 patients; these had 4,471 siblings. Fecundity was significantly lower in both men and women (FR, 0.25 and 0.48, respectively). Among the siblings, brothers also had fewer children (FR, 0.94). Among sisters, the rate was not significantly different than the general population.
"Individuals with autism showed the greatest reduction in fecundity among all examined disorders. This was not unexpected because previous investigations have shown that few individuals with autism ever married or had children.
"We propose that rare highly deleterious variants and sexually antagonistic polymorphisms may contribute to the genetic disposition to autism. The similarity to schizophrenia is notable because it has been proposed that the autistic and psychotic spectrums reflect two extremes of social cognition."
Bipolar disorder was present in 14,439 patients, among whom were 22,986 siblings. Fecundity was significantly lower than the general population in both men (FR, 0.75) and women (FR, 0.85). While brothers had similar rates to that of the general population, sisters had significantly more children (FR, 1.03). But incorporating comorbidities into the analysis changed the significance for both patients and sisters, with the patient rate increasing to just below that of the general population (FR, 0.94), and the sisters’ rate increased rate no longer being significant (FR, 0.95).
"It has been suggested that the introduction of lithium as a treatment for bipolar disorder has led to improved functioning and, as a result, greater fecundity in those populations where treatment is available."
There were 81,295 patients with depression, among who were 119,645 siblings. While men with depression had significantly lower rates (FR, 0.93), women with the disorder were not significantly different than the general population. Siblings had significantly more children than the general population (FR, brothers 1.01, sisters 1.04) – a difference that was unchanged by factoring in comorbidities. The addition of comorbidities to the analysis did not change the decreased fecundity rate for male patients, but actually increased the rate for female patients (FR, 1.03).
"Notably, depression was an exception to the five other studied disorders ... genes associated with depression seems to be maintained in the population by balancing selection because the cost to affected individuals is roughly equal to the benefit to their siblings. If this is the case, it would be the first strong evidence for balancing selection in a psychiatric disorder."
There were 3,275 patients with anorexia, who had a total of 5,172 siblings. Both men and women with the disorder had significantly reduced fecundity (0.54 and 0.58, respectively). In the sibling group, there were no significant differences for either brothers or sisters. None of the findings changed when comorbidities were factored in.
"Our calculations suggest that anorexia is under weaker negative selection relative to schizophrenia and autism," the authors said.
Substance abuse was present in 55,933 patients, who had a total of 81,592 siblings. The fecundity rate was significantly lower in both men and women (FR, 0.78 and 0.93, respectively). Siblings had significantly more children than the general population (FR, 1.03 for brothers and 1.05 for sisters).
"Our findings suggest that this increased fecundity in siblings almost entirely accounts for the cost to affected individuals, with only a slight decrease in the frequency of these individuals’ genes predicted each generation. Considering that most drugs are a new environmental exposure when seen from an evolutionary perspective, it is possible that there has been insufficient time for selection to act on risk alleles. ... It has also been suggested that substance abuse is associated with risk-taking behavior in both sexes, including sexual risk taking."
The study was funded by the Medical Research Council of the United Kingdom. One of the coauthors reported having received consulting fees and honoraria from GlaxoSmithKline and Lundbeck.
Some psychiatric disorders appear to be associated with lower fecundity in both men and women, suggesting that natural selection attempts to discourage the perpetuation of genetic variants associated with them.
Instead, new mutations could be one reason that the disorders continue to exist, Robert A. Power and his colleagues wrote in the January issue of JAMA Psychiatry (formerly Archives of General Psychiatry).
The authors also found that psychiatric disorders affected men’s fecundity more than women’s. "This sex-specific effect suggests that psychiatric morbidity impairs interest or ability to find suitable mating partners or inhibits biological fertility to a greater extent in men," wrote Mr. Power of Kings College, London.
The study data were extracted from two of Sweden’s population registries – the Multi-Generation Register and the Swedish Hospital Discharge Register. More than 2.3 million people born from 1950-1970 were cross-linked by individual patient identification numbers, which allowed the researchers to trace not only patients but also their siblings. At the time of the analysis, no patient was younger than 40 years (JAMA Psychiatry 2013;70:22-30).
The authors tracked fecundity in about 177,000 patients who had schizophrenia, autism, bipolar disorder, anorexia nervosa, or substance abuse. Rates were compared with fecundity in 261,000 siblings. The researchers then compared these rates with those found in the general population.
About 19,000 patients had schizophrenia. They had significantly fewer children than the general population’s (fecundity rate [FR] 0.23 for men and 0.47 for women). In a univariate analysis, sisters of the patients had a significantly higher fecundity rate (FR, 1.02), but this difference disappeared once comorbidities were factored into the analysis. Brothers also had significantly decreased fecundity (FR, 0.97).
"Our results suggest a strong selection pressure to remove genetic variants associated with schizophrenia from the population," the authors said. "This is further evidence for the role of recent or de novo mutations in the genetic susceptibility to schizophrenia that has neither reached the frequency of nor existed long enough to be removed from the population."
Autism was present in 2,947 patients; these had 4,471 siblings. Fecundity was significantly lower in both men and women (FR, 0.25 and 0.48, respectively). Among the siblings, brothers also had fewer children (FR, 0.94). Among sisters, the rate was not significantly different than the general population.
"Individuals with autism showed the greatest reduction in fecundity among all examined disorders. This was not unexpected because previous investigations have shown that few individuals with autism ever married or had children.
"We propose that rare highly deleterious variants and sexually antagonistic polymorphisms may contribute to the genetic disposition to autism. The similarity to schizophrenia is notable because it has been proposed that the autistic and psychotic spectrums reflect two extremes of social cognition."
Bipolar disorder was present in 14,439 patients, among whom were 22,986 siblings. Fecundity was significantly lower than the general population in both men (FR, 0.75) and women (FR, 0.85). While brothers had similar rates to that of the general population, sisters had significantly more children (FR, 1.03). But incorporating comorbidities into the analysis changed the significance for both patients and sisters, with the patient rate increasing to just below that of the general population (FR, 0.94), and the sisters’ rate increased rate no longer being significant (FR, 0.95).
"It has been suggested that the introduction of lithium as a treatment for bipolar disorder has led to improved functioning and, as a result, greater fecundity in those populations where treatment is available."
There were 81,295 patients with depression, among who were 119,645 siblings. While men with depression had significantly lower rates (FR, 0.93), women with the disorder were not significantly different than the general population. Siblings had significantly more children than the general population (FR, brothers 1.01, sisters 1.04) – a difference that was unchanged by factoring in comorbidities. The addition of comorbidities to the analysis did not change the decreased fecundity rate for male patients, but actually increased the rate for female patients (FR, 1.03).
"Notably, depression was an exception to the five other studied disorders ... genes associated with depression seems to be maintained in the population by balancing selection because the cost to affected individuals is roughly equal to the benefit to their siblings. If this is the case, it would be the first strong evidence for balancing selection in a psychiatric disorder."
There were 3,275 patients with anorexia, who had a total of 5,172 siblings. Both men and women with the disorder had significantly reduced fecundity (0.54 and 0.58, respectively). In the sibling group, there were no significant differences for either brothers or sisters. None of the findings changed when comorbidities were factored in.
"Our calculations suggest that anorexia is under weaker negative selection relative to schizophrenia and autism," the authors said.
Substance abuse was present in 55,933 patients, who had a total of 81,592 siblings. The fecundity rate was significantly lower in both men and women (FR, 0.78 and 0.93, respectively). Siblings had significantly more children than the general population (FR, 1.03 for brothers and 1.05 for sisters).
"Our findings suggest that this increased fecundity in siblings almost entirely accounts for the cost to affected individuals, with only a slight decrease in the frequency of these individuals’ genes predicted each generation. Considering that most drugs are a new environmental exposure when seen from an evolutionary perspective, it is possible that there has been insufficient time for selection to act on risk alleles. ... It has also been suggested that substance abuse is associated with risk-taking behavior in both sexes, including sexual risk taking."
The study was funded by the Medical Research Council of the United Kingdom. One of the coauthors reported having received consulting fees and honoraria from GlaxoSmithKline and Lundbeck.
FROM JAMA PSYCHIATRY
Major Finding: Fecundity rates appear lower in patients with some psychiatric disorders, ranging from 25%-95% that of the general population.
Data Source: The population registry-based study included about 177,000 patients in Sweden with schizophrenia, autism, bipolar disorder, depression, anorexia, and substance abuse.
Disclosures: The study was funded by the Medical Research Council of the United Kingdom. One of the coauthors reported having received consulting fees and honoraria from GlaxoSmithKline and Lundbeck.
Paranoid, agitated, and manipulative
CASE: Agitation
Mrs. M, age 39, presents to the emergency department (ED) with altered mental status. She is escorted by her husband and the police. She has a history of severe alcohol dependence, bipolar disorder (BD), anxiety, borderline personality disorder (BPD), hypothyroidism, and bulimia, and had gastric bypass surgery 4 years ago. Her husband called 911 when he could no longer manage Mrs. M’s agitated state. The police found her to be extremely paranoid, restless, and disoriented. Her husband reports that she shouted “the world is going to end” before she escaped naked into her neighborhood streets.
On several occasions Mrs. M had been admitted to the same hospital for alcohol withdrawal and dependence with subsequent liver failure, leading to jaundice, coagulopathy, and ascites. During these hospitalizations, she exhibited poor behavioral tendencies, unhealthy psychological defenses, and chronic maladaptive coping and defense mechanisms congruent with her BPD diagnosis. Specifically, she engaged in splitting of hospital staff, ranging from extreme flattery to overt devaluation and hostility. Other defense mechanisms included denial, distortion, acting out, and passive-aggressive behavior. During these admissions, Mrs. M often displayed deficits in recall and attention on Mini-Mental State Examination (MMSE), but these deficits were associated with concurrent alcohol use and improved rapidly during her stay.
In her current presentation, Mrs. M’s mental status change is more pronounced and atypical compared with earlier admissions. Her outpatient medication regimen includes lamotrigine, 100 mg/d, levothyroxine, 88 mcg/d, venlafaxine extended release (XR), 75 mg/d, clonazepam, 3 mg/d, docusate as needed for constipation, and a daily multivitamin.
The authors’ observations
Delirium is a disturbance of consciousness manifested by a reduced clarity of awareness (impairment in attention) and change in cognition (impairment in orientation, memory, and language).1,2 The disturbance develops over a short time and tends to fluctuate during the day. Delirium is a direct physiological consequence of a general medical condition, substance use (intoxication or withdrawal), or both (Table).3
Delirium generally is a reversible mental disorder but can progress to irreversible brain damage. Prompt and accurate diagnosis of delirium is essential,4 although the condition often is underdiagnosed or misdiagnosed because of lack of recognition.
Table
DSM-IV-TR diagnostic criteria for delirium
|
Source: Reference 3 |
Patients who have convoluted histories, such as Mrs. M, are common and difficult to manage and treat. These patients become substantially more complex when they are admitted to inpatient medical or surgical services. The need to clarify between delirium (primarily medical) and depression (primarily psychiatric) becomes paramount when administering treatment and evaluating decision-making capacity.5 In Mrs. M’s case, internal medicine, neurology, and psychiatry teams each had a different approach to altered mental status. Each team’s different terminology, assessment, and objectives further complicated an already challenging case.6
EVALUATION: Confounding results
The ED physicians offer a working diagnosis of acute mental status change, administer IV lorazepam, 4 mg, and order restraints for Mrs. M’s severe agitation. Her initial vital signs reveal slightly elevated blood pressure (140/90 mm Hg) and tachycardia (115 beats per minute). Internal medicine clinicians note that Mrs. M is not in acute distress, although she refuses to speak and has a small amount of dried blood on her lips, presumably from a struggle with the police before coming to the hospital, but this is not certain. Her abdomen is not tender; she has normal bowel sounds, and no asterixis is noted on neurologic exam. Physical exam is otherwise normal. A noncontrast head CT scan shows no acute process. Initial lab values show elevations in ammonia (277 μg/dL) and γ-glutamyl transpeptidase (68 U/L). Thyroid-stimulating hormone is 1.45 mlU/L, prothrombin time is 19.5 s, partial thromboplastin time is 40.3 s, and international normalized ratio is 1.67. The internal medicine team admits Mrs. M to the intensive care unit (ICU) for further management of her mental status change with alcohol withdrawal or hepatic encephalopathy as the most likely etiologies.
Mrs. M’s husband says that his wife has not consumed alcohol in the last 4 months in preparation for a possible liver transplant; however, past interactions with Mrs. M’s family suggest they are unreliable. The Clinical Institute Withdrawal Assessment (CIWA) protocol is implemented in case her symptoms are caused by alcohol withdrawal. Her vital signs are stable and IV lorazepam, 4 mg, is administered once for agitation. Mrs. M’s husband also reports that 1 month ago his wife underwent a transjugular intrahepatic portosystemic shunt (TIPS) procedure for portal hypertension. Outpatient psychotropics (lamotrigine, 100 mg/d, and venlafaxine XR, 75 mg/d) are restarted because withdrawal from these drugs may exacerbate her symptoms. In the ICU Mrs. M experiences a tonic-clonic seizure with fecal incontinence and bitten tongue, which results in a consultation from neurology and the psychiatry consultation-liaison service.
Psychiatry recommends withholding psychotropics, stopping CIWA, and using vital sign parameters along with objective signs of diaphoresis and tremors as indicators of alcohol withdrawal for lorazepam administration. Mrs. M receives IV haloperidol, 1 mg, once during her second day in the hospital for severe agitation, but this medication is discontinued because of concern about lowering her seizure threshold.7 After treatment with lactulose, her ammonia levels trend down to 33 μg/dL, but her altered mental state persists with significant deficits in attention and orientation.
The neurology service performs an EEG that shows no slow-wave, triphasic waves, or epileptiform activity, which likely would be present in delirium or seizures. See Figure 1 for an example of triphasic waves on an EEG and Figure 2 for Mrs. M's EEG results. Subsequent lumbar puncture, MRI, and a second EEG are unremarkable. By the fifth hospital day, Mrs. M is calm and her paranoia has subsided, but she still is confused and disoriented. Psychiatry orders a third EEG while she is in this confused state; it shows no pathologic process. Based on these examinations, neurology posits that Mrs. M is not encephalopathic.
Figure 1: Representative sample of triphasic waves
This EEG tracing is from a 54-year-old woman who underwent prolonged abdominal surgery for lysis of adhesions during which she suffered an intraoperative left subinsular stroke followed by nonconvulsive status epilepticus. The tracing demonstrates typical morphology with the positive sharp transient preceded and followed by smaller amplitude negative deflections. Symmetric, frontal predominance of findings seen is this tracing is common
Figure 2: Mrs. M’s EEG results
This is a representative tracing of Mrs. M’s 3 EEGs revealing an 8.5 to 9 Hz dominant alpha rhythm. There is superimposed frontally dominant beta fast activity, which is consistent with known administration of benzodiazepines
The authors’ observations
Mrs. M had repeated admissions for alcohol dependence and subsequent liver failure. Her recent hospitalization was complicated by a TIPS procedure done 1 month ago. The incidence of hepatic encephalopathy in patients undergoing TIPS is >30%, especially in the first month post-procedure, which raised suspicion that hepatic encephalopathy played a significant role in Mrs. M’s delirium.8
Because of frequent hospitalization, Mrs. M was well known to the internal medicine, neurology, and psychiatry teams, and each used different terms to describe her mental state. Internal medicine used the phrase “acute mental status change,” which covers a broad differential. Neurology used “encephalopathy,” which also is a general term. Psychiatry used “delirium,” which has narrower and more specific diagnostic criteria. Engel et al9 described the delirious patient as having “cerebral insufficiency” with universally abnormal EEG. Regardless of terminology, based on Mrs. M’s acute confusion, one would expect an abnormal EEG, but repeat EEGs were unremarkable.
Interpreting EEG
EEG is one of the few tools available for measuring acute changes in cerebral function, and an EEG slowing remains a hallmark in encephalopathic processes.10,11 Initially, the 3 specialties agreed that Mrs. M’s presentation likely was caused by underlying medical issues or substances (alcohol or others). EEG can help recognize delirium, and, in some cases, elucidate the underlying cause.10,12 It was surprising that Mrs. M’s EEGs were normal despite a clinical presentation of delirium. Because of the normal EEG findings, neurology leaned toward a primary psychiatric (“functional”) etiology as the cause of her delirium vs a general medical condition or alcohol withdrawal (“organic”).
A literature search in regards to sensitivity of EEG in delirium revealed conflicting statements and data. A standard textbook in neurology and psychiatry states that “a normal EEG virtually excludes a toxic-metabolic encephalopathy.”13 The American Psychiatric Association’s (APA) practice guidelines for delirium states: “The presence of EEG abnormalities has fairly good sensitivities for delirium (in one study, the sensitivity was found to be 75%), but the absence does not rule out the diagnosis; thus the EEG is no substitute for careful clinical observation.”6
At the beginning of Mrs. M’s care, in discussion with the neurology and internal medicine teams, we argued that Mrs. M was experiencing delirium despite her initial normal EEG. We did not expect that 2 subsequent EEGs would be normal, especially because the teams witnessed the final EEG being performed while Mrs. M was clinically evaluated and observed to be in a state of delirium.
OUTCOME: Cause still unknown
By the 6th day of hospitalization, Mrs. M’s vitals are normal and she remains hemodynamically stable. Differential diagnosis remains wide and unclear. The psychiatry team feels she could have atypical catatonia due to an underlying mood disorder. One hour after a trial of IV lorazepam, 1 mg, Mrs. M is more lucid and fully oriented, with MMSE of 28/30 (recall was 1/3), indicating normal cognition. During the exam, a psychiatry resident notes Mrs. M winks and feigns a yawn at the medical students and nurses in the room, displaying her boredom with the interview and simplicity of the mental status exam questions. Later that evening, Mrs. M exhibits bizarre sexual gestures toward male hospital staff, including licking a male nursing staff member’s hand.
Although Mrs. M’s initial confusion resolved, the severity of her comorbid psychiatric history warrants inpatient psychiatric hospitalization. She agrees to transfer to the psychiatric ward after she confesses anxiety regarding death, intense demoralization, and guilt related to her condition and her relationship with her 12-year-old daughter. She tearfully reports that she discontinued her psychotropic medications shortly after stopping alcohol 4 months ago. Shortly before her transfer, psychiatry is called back to the medicine floor because of Mrs. M’s disruptive behavior.
The team finds Mrs. M in her hospital gown, pursuing her husband in the hallway as he is leaving, yelling profanities and blaming him for her horrible experience in the hospital. Based on her demeanor, the team determines that she is back to her baseline mental state despite her mood disorder, and that her upcoming inpatient psychiatric stay likely would be too short to address her comorbid personality disorder. The next day she signs out of the hospital against medical advice.
The authors’ observations
We never clearly identified the specific etiology responsible for Mrs. M’s delirium. We assume at the initial presentation she had toxic-metabolic encephalopathy that rapidly resolved with lactulose treatment and lowering her ammonia. She then had a single tonic-clonic seizure, perhaps related to stopping and then restarting her psychotropics. Her subsequent confusion, bizarre sexual behavior, and demeanor on her final hospital days were more indicative of her psychiatric diagnoses. We now suspect that Mrs. M’s delirium was briefer than presumed and she returned to her baseline borderline personality, resulting in some factitious staging of delirium to confuse her 3 treating teams (a psychoanalyst may say this was a form of projective identification).
We felt that if Mrs. M truly was delirious due to metabolic or hepatic dysfunction or alcohol withdrawal, she would have had abnormal EEG findings. We discovered that the notion of “75% sensitivity” of EEG abnormalities cited in the APA guidelines comes from studies that include patients with “psychogenic” and “organic” delirium. Acute manias and agitated psychoses were termed “psychogenic delirium” and acute confusion due to medical conditions or substance issues was termed “organic delirium.”9,12,14-16
This poses a circular reasoning in the diagnostic criteria and clinical approach to delirium. The fallacy is that, according to DSM-IV-TR, delirium is supposed to be the result of a direct physiological consequence of a general medical condition or substance use (criterion D), and cannot be due to psychosis (eg, schizophrenia) or mania (eg, BD). We question the presumptive 75% sensitivity of EEG abnormalities in patients with delirium because it is possible that when some of these studies were conducted the definition of delirium was not solidified or fully understood. We suspect the sensitivity would be much higher if the correct definition of delirium according to DSM-IV-TR is used in future studies. To improve interdisciplinary communication and future research, it would be constructive if all disciplines could agree on a single term, with the same diagnostic criteria, when evaluating a patient with acute confusion.
Related Resources
- Meagher D. Delirium: the role of psychiatry. Advances in Psychiatric Treatment. 2001;7:433-442.
- Casey DA, DeFazio JV Jr, Vansickle K, et al. Delirium. Quick recognition, careful evaluation, and appropriate treatment. Postgrad Med. 1996;100(1):121-4, 128, 133-134.
Drug Brand Names
- Clonazepam • Klonopin
- Docusate • Surfak
- Haloperidol • Haldol
- Lamotrigine • Lamictal
- Lorazepam • Ativan
- Levothyroxine • Levoxyl, Synthtoid
- Venlafaxine XR • Effexor XR
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
Acknowledgment
The views expressed in this article are those of the authors and do not necessarily reflect the official policy or position of the Department of the Navy, Department of Defense, or the U.S. Government. The authors are employees of the U.S. Government. This work was prepared as part of their official duties. Title 17 U.S.C. 105 provides that “Copyright protection under this title is not available for any work of the U.S. Government.” Title 17 U.S.C. 101 defines a U.S. Government work as a work prepared by a military service member or employee of the U.S. Government as part of that person’s official duties.
1. Katz IR, Mossey J, Sussman N, et al. Bedside clinical and electrophysiological assessment: assessment of change in vulnerable patients. Int Psychogeriatr. 1991;3(2):289-300.
2. Inouye SK. Delirium in older persons. N Engl J Med. 2006;354(11):1157-1165.
3. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.
4. McPhee SJ, Papadakis M, Rabow MW. CURRENT medical diagnosis and treatment. New York NY: McGraw Hill Medical; 2012.
5. Brody B. Who has capacity? N Engl J Med. 2009;361(3):232-233.
6. Practice guideline for the treatment of patients with delirium. American Psychiatric Association. Am J Psychiatry. 1999;156(5 suppl):1-20.
7. Fricchione GL, Nejad SH, Esses JA, et al. Postoperative delirium. Am J Psychiatry. 2008;165(7):803-812.
8. Sanyal AJ, Freedman AM, Shiffman ML, et al. Portosystemic encephalopathy after transjugular intrahepatic portosystemic shunt: results of a prospective controlled study. Hepatology. 1994;20(1 pt 1):46-55.
9. Engel GL, Romano J. Delirium a syndrome of cerebral insufficiency. 1959. J Neuropsychiatry Clin Neurosci. 2004;16(4):526-538.
10. Pro JD, Wells CE. The use of the electroencephalogram in the diagnosis of delirium. Dis Nerv Syst. 1977;38(10):804-808.
11. Sidhu KS, Balon R, Ajluni V, et al. Standard EEG and the difficult-to-assess mental status. Ann Clin Psychiatry. 2009;21(2):103-108.
12. Brenner RP. Utility of EEG in delirium: past views and current practice. Int Psychogeriatr. 1991;3(2):211-229.
13. Kaufman DM. Clinical neurology for psychiatrists. 5th ed. Philadelphia PA: Saunders; 2001: 230-232.
14. Bond TC. Recognition of acute delirious mania. Arch Gen Psychiatry. 1980;37(5):553-554.
15. Krauthammer C, Klerman GL. Secondary mania: manic syndromes associated with antecedent physical illness or drugs. Arch Gen Psychiatry. 1978;35(11):1333-1339.
16. Larson EW, Richelson E. Organic causes of mania. Mayo Clin Proc. 1988;63(9):906-912.
CASE: Agitation
Mrs. M, age 39, presents to the emergency department (ED) with altered mental status. She is escorted by her husband and the police. She has a history of severe alcohol dependence, bipolar disorder (BD), anxiety, borderline personality disorder (BPD), hypothyroidism, and bulimia, and had gastric bypass surgery 4 years ago. Her husband called 911 when he could no longer manage Mrs. M’s agitated state. The police found her to be extremely paranoid, restless, and disoriented. Her husband reports that she shouted “the world is going to end” before she escaped naked into her neighborhood streets.
On several occasions Mrs. M had been admitted to the same hospital for alcohol withdrawal and dependence with subsequent liver failure, leading to jaundice, coagulopathy, and ascites. During these hospitalizations, she exhibited poor behavioral tendencies, unhealthy psychological defenses, and chronic maladaptive coping and defense mechanisms congruent with her BPD diagnosis. Specifically, she engaged in splitting of hospital staff, ranging from extreme flattery to overt devaluation and hostility. Other defense mechanisms included denial, distortion, acting out, and passive-aggressive behavior. During these admissions, Mrs. M often displayed deficits in recall and attention on Mini-Mental State Examination (MMSE), but these deficits were associated with concurrent alcohol use and improved rapidly during her stay.
In her current presentation, Mrs. M’s mental status change is more pronounced and atypical compared with earlier admissions. Her outpatient medication regimen includes lamotrigine, 100 mg/d, levothyroxine, 88 mcg/d, venlafaxine extended release (XR), 75 mg/d, clonazepam, 3 mg/d, docusate as needed for constipation, and a daily multivitamin.
The authors’ observations
Delirium is a disturbance of consciousness manifested by a reduced clarity of awareness (impairment in attention) and change in cognition (impairment in orientation, memory, and language).1,2 The disturbance develops over a short time and tends to fluctuate during the day. Delirium is a direct physiological consequence of a general medical condition, substance use (intoxication or withdrawal), or both (Table).3
Delirium generally is a reversible mental disorder but can progress to irreversible brain damage. Prompt and accurate diagnosis of delirium is essential,4 although the condition often is underdiagnosed or misdiagnosed because of lack of recognition.
Table
DSM-IV-TR diagnostic criteria for delirium
|
Source: Reference 3 |
Patients who have convoluted histories, such as Mrs. M, are common and difficult to manage and treat. These patients become substantially more complex when they are admitted to inpatient medical or surgical services. The need to clarify between delirium (primarily medical) and depression (primarily psychiatric) becomes paramount when administering treatment and evaluating decision-making capacity.5 In Mrs. M’s case, internal medicine, neurology, and psychiatry teams each had a different approach to altered mental status. Each team’s different terminology, assessment, and objectives further complicated an already challenging case.6
EVALUATION: Confounding results
The ED physicians offer a working diagnosis of acute mental status change, administer IV lorazepam, 4 mg, and order restraints for Mrs. M’s severe agitation. Her initial vital signs reveal slightly elevated blood pressure (140/90 mm Hg) and tachycardia (115 beats per minute). Internal medicine clinicians note that Mrs. M is not in acute distress, although she refuses to speak and has a small amount of dried blood on her lips, presumably from a struggle with the police before coming to the hospital, but this is not certain. Her abdomen is not tender; she has normal bowel sounds, and no asterixis is noted on neurologic exam. Physical exam is otherwise normal. A noncontrast head CT scan shows no acute process. Initial lab values show elevations in ammonia (277 μg/dL) and γ-glutamyl transpeptidase (68 U/L). Thyroid-stimulating hormone is 1.45 mlU/L, prothrombin time is 19.5 s, partial thromboplastin time is 40.3 s, and international normalized ratio is 1.67. The internal medicine team admits Mrs. M to the intensive care unit (ICU) for further management of her mental status change with alcohol withdrawal or hepatic encephalopathy as the most likely etiologies.
Mrs. M’s husband says that his wife has not consumed alcohol in the last 4 months in preparation for a possible liver transplant; however, past interactions with Mrs. M’s family suggest they are unreliable. The Clinical Institute Withdrawal Assessment (CIWA) protocol is implemented in case her symptoms are caused by alcohol withdrawal. Her vital signs are stable and IV lorazepam, 4 mg, is administered once for agitation. Mrs. M’s husband also reports that 1 month ago his wife underwent a transjugular intrahepatic portosystemic shunt (TIPS) procedure for portal hypertension. Outpatient psychotropics (lamotrigine, 100 mg/d, and venlafaxine XR, 75 mg/d) are restarted because withdrawal from these drugs may exacerbate her symptoms. In the ICU Mrs. M experiences a tonic-clonic seizure with fecal incontinence and bitten tongue, which results in a consultation from neurology and the psychiatry consultation-liaison service.
Psychiatry recommends withholding psychotropics, stopping CIWA, and using vital sign parameters along with objective signs of diaphoresis and tremors as indicators of alcohol withdrawal for lorazepam administration. Mrs. M receives IV haloperidol, 1 mg, once during her second day in the hospital for severe agitation, but this medication is discontinued because of concern about lowering her seizure threshold.7 After treatment with lactulose, her ammonia levels trend down to 33 μg/dL, but her altered mental state persists with significant deficits in attention and orientation.
The neurology service performs an EEG that shows no slow-wave, triphasic waves, or epileptiform activity, which likely would be present in delirium or seizures. See Figure 1 for an example of triphasic waves on an EEG and Figure 2 for Mrs. M's EEG results. Subsequent lumbar puncture, MRI, and a second EEG are unremarkable. By the fifth hospital day, Mrs. M is calm and her paranoia has subsided, but she still is confused and disoriented. Psychiatry orders a third EEG while she is in this confused state; it shows no pathologic process. Based on these examinations, neurology posits that Mrs. M is not encephalopathic.
Figure 1: Representative sample of triphasic waves
This EEG tracing is from a 54-year-old woman who underwent prolonged abdominal surgery for lysis of adhesions during which she suffered an intraoperative left subinsular stroke followed by nonconvulsive status epilepticus. The tracing demonstrates typical morphology with the positive sharp transient preceded and followed by smaller amplitude negative deflections. Symmetric, frontal predominance of findings seen is this tracing is common
Figure 2: Mrs. M’s EEG results
This is a representative tracing of Mrs. M’s 3 EEGs revealing an 8.5 to 9 Hz dominant alpha rhythm. There is superimposed frontally dominant beta fast activity, which is consistent with known administration of benzodiazepines
The authors’ observations
Mrs. M had repeated admissions for alcohol dependence and subsequent liver failure. Her recent hospitalization was complicated by a TIPS procedure done 1 month ago. The incidence of hepatic encephalopathy in patients undergoing TIPS is >30%, especially in the first month post-procedure, which raised suspicion that hepatic encephalopathy played a significant role in Mrs. M’s delirium.8
Because of frequent hospitalization, Mrs. M was well known to the internal medicine, neurology, and psychiatry teams, and each used different terms to describe her mental state. Internal medicine used the phrase “acute mental status change,” which covers a broad differential. Neurology used “encephalopathy,” which also is a general term. Psychiatry used “delirium,” which has narrower and more specific diagnostic criteria. Engel et al9 described the delirious patient as having “cerebral insufficiency” with universally abnormal EEG. Regardless of terminology, based on Mrs. M’s acute confusion, one would expect an abnormal EEG, but repeat EEGs were unremarkable.
Interpreting EEG
EEG is one of the few tools available for measuring acute changes in cerebral function, and an EEG slowing remains a hallmark in encephalopathic processes.10,11 Initially, the 3 specialties agreed that Mrs. M’s presentation likely was caused by underlying medical issues or substances (alcohol or others). EEG can help recognize delirium, and, in some cases, elucidate the underlying cause.10,12 It was surprising that Mrs. M’s EEGs were normal despite a clinical presentation of delirium. Because of the normal EEG findings, neurology leaned toward a primary psychiatric (“functional”) etiology as the cause of her delirium vs a general medical condition or alcohol withdrawal (“organic”).
A literature search in regards to sensitivity of EEG in delirium revealed conflicting statements and data. A standard textbook in neurology and psychiatry states that “a normal EEG virtually excludes a toxic-metabolic encephalopathy.”13 The American Psychiatric Association’s (APA) practice guidelines for delirium states: “The presence of EEG abnormalities has fairly good sensitivities for delirium (in one study, the sensitivity was found to be 75%), but the absence does not rule out the diagnosis; thus the EEG is no substitute for careful clinical observation.”6
At the beginning of Mrs. M’s care, in discussion with the neurology and internal medicine teams, we argued that Mrs. M was experiencing delirium despite her initial normal EEG. We did not expect that 2 subsequent EEGs would be normal, especially because the teams witnessed the final EEG being performed while Mrs. M was clinically evaluated and observed to be in a state of delirium.
OUTCOME: Cause still unknown
By the 6th day of hospitalization, Mrs. M’s vitals are normal and she remains hemodynamically stable. Differential diagnosis remains wide and unclear. The psychiatry team feels she could have atypical catatonia due to an underlying mood disorder. One hour after a trial of IV lorazepam, 1 mg, Mrs. M is more lucid and fully oriented, with MMSE of 28/30 (recall was 1/3), indicating normal cognition. During the exam, a psychiatry resident notes Mrs. M winks and feigns a yawn at the medical students and nurses in the room, displaying her boredom with the interview and simplicity of the mental status exam questions. Later that evening, Mrs. M exhibits bizarre sexual gestures toward male hospital staff, including licking a male nursing staff member’s hand.
Although Mrs. M’s initial confusion resolved, the severity of her comorbid psychiatric history warrants inpatient psychiatric hospitalization. She agrees to transfer to the psychiatric ward after she confesses anxiety regarding death, intense demoralization, and guilt related to her condition and her relationship with her 12-year-old daughter. She tearfully reports that she discontinued her psychotropic medications shortly after stopping alcohol 4 months ago. Shortly before her transfer, psychiatry is called back to the medicine floor because of Mrs. M’s disruptive behavior.
The team finds Mrs. M in her hospital gown, pursuing her husband in the hallway as he is leaving, yelling profanities and blaming him for her horrible experience in the hospital. Based on her demeanor, the team determines that she is back to her baseline mental state despite her mood disorder, and that her upcoming inpatient psychiatric stay likely would be too short to address her comorbid personality disorder. The next day she signs out of the hospital against medical advice.
The authors’ observations
We never clearly identified the specific etiology responsible for Mrs. M’s delirium. We assume at the initial presentation she had toxic-metabolic encephalopathy that rapidly resolved with lactulose treatment and lowering her ammonia. She then had a single tonic-clonic seizure, perhaps related to stopping and then restarting her psychotropics. Her subsequent confusion, bizarre sexual behavior, and demeanor on her final hospital days were more indicative of her psychiatric diagnoses. We now suspect that Mrs. M’s delirium was briefer than presumed and she returned to her baseline borderline personality, resulting in some factitious staging of delirium to confuse her 3 treating teams (a psychoanalyst may say this was a form of projective identification).
We felt that if Mrs. M truly was delirious due to metabolic or hepatic dysfunction or alcohol withdrawal, she would have had abnormal EEG findings. We discovered that the notion of “75% sensitivity” of EEG abnormalities cited in the APA guidelines comes from studies that include patients with “psychogenic” and “organic” delirium. Acute manias and agitated psychoses were termed “psychogenic delirium” and acute confusion due to medical conditions or substance issues was termed “organic delirium.”9,12,14-16
This poses a circular reasoning in the diagnostic criteria and clinical approach to delirium. The fallacy is that, according to DSM-IV-TR, delirium is supposed to be the result of a direct physiological consequence of a general medical condition or substance use (criterion D), and cannot be due to psychosis (eg, schizophrenia) or mania (eg, BD). We question the presumptive 75% sensitivity of EEG abnormalities in patients with delirium because it is possible that when some of these studies were conducted the definition of delirium was not solidified or fully understood. We suspect the sensitivity would be much higher if the correct definition of delirium according to DSM-IV-TR is used in future studies. To improve interdisciplinary communication and future research, it would be constructive if all disciplines could agree on a single term, with the same diagnostic criteria, when evaluating a patient with acute confusion.
Related Resources
- Meagher D. Delirium: the role of psychiatry. Advances in Psychiatric Treatment. 2001;7:433-442.
- Casey DA, DeFazio JV Jr, Vansickle K, et al. Delirium. Quick recognition, careful evaluation, and appropriate treatment. Postgrad Med. 1996;100(1):121-4, 128, 133-134.
Drug Brand Names
- Clonazepam • Klonopin
- Docusate • Surfak
- Haloperidol • Haldol
- Lamotrigine • Lamictal
- Lorazepam • Ativan
- Levothyroxine • Levoxyl, Synthtoid
- Venlafaxine XR • Effexor XR
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
Acknowledgment
The views expressed in this article are those of the authors and do not necessarily reflect the official policy or position of the Department of the Navy, Department of Defense, or the U.S. Government. The authors are employees of the U.S. Government. This work was prepared as part of their official duties. Title 17 U.S.C. 105 provides that “Copyright protection under this title is not available for any work of the U.S. Government.” Title 17 U.S.C. 101 defines a U.S. Government work as a work prepared by a military service member or employee of the U.S. Government as part of that person’s official duties.
CASE: Agitation
Mrs. M, age 39, presents to the emergency department (ED) with altered mental status. She is escorted by her husband and the police. She has a history of severe alcohol dependence, bipolar disorder (BD), anxiety, borderline personality disorder (BPD), hypothyroidism, and bulimia, and had gastric bypass surgery 4 years ago. Her husband called 911 when he could no longer manage Mrs. M’s agitated state. The police found her to be extremely paranoid, restless, and disoriented. Her husband reports that she shouted “the world is going to end” before she escaped naked into her neighborhood streets.
On several occasions Mrs. M had been admitted to the same hospital for alcohol withdrawal and dependence with subsequent liver failure, leading to jaundice, coagulopathy, and ascites. During these hospitalizations, she exhibited poor behavioral tendencies, unhealthy psychological defenses, and chronic maladaptive coping and defense mechanisms congruent with her BPD diagnosis. Specifically, she engaged in splitting of hospital staff, ranging from extreme flattery to overt devaluation and hostility. Other defense mechanisms included denial, distortion, acting out, and passive-aggressive behavior. During these admissions, Mrs. M often displayed deficits in recall and attention on Mini-Mental State Examination (MMSE), but these deficits were associated with concurrent alcohol use and improved rapidly during her stay.
In her current presentation, Mrs. M’s mental status change is more pronounced and atypical compared with earlier admissions. Her outpatient medication regimen includes lamotrigine, 100 mg/d, levothyroxine, 88 mcg/d, venlafaxine extended release (XR), 75 mg/d, clonazepam, 3 mg/d, docusate as needed for constipation, and a daily multivitamin.
The authors’ observations
Delirium is a disturbance of consciousness manifested by a reduced clarity of awareness (impairment in attention) and change in cognition (impairment in orientation, memory, and language).1,2 The disturbance develops over a short time and tends to fluctuate during the day. Delirium is a direct physiological consequence of a general medical condition, substance use (intoxication or withdrawal), or both (Table).3
Delirium generally is a reversible mental disorder but can progress to irreversible brain damage. Prompt and accurate diagnosis of delirium is essential,4 although the condition often is underdiagnosed or misdiagnosed because of lack of recognition.
Table
DSM-IV-TR diagnostic criteria for delirium
|
Source: Reference 3 |
Patients who have convoluted histories, such as Mrs. M, are common and difficult to manage and treat. These patients become substantially more complex when they are admitted to inpatient medical or surgical services. The need to clarify between delirium (primarily medical) and depression (primarily psychiatric) becomes paramount when administering treatment and evaluating decision-making capacity.5 In Mrs. M’s case, internal medicine, neurology, and psychiatry teams each had a different approach to altered mental status. Each team’s different terminology, assessment, and objectives further complicated an already challenging case.6
EVALUATION: Confounding results
The ED physicians offer a working diagnosis of acute mental status change, administer IV lorazepam, 4 mg, and order restraints for Mrs. M’s severe agitation. Her initial vital signs reveal slightly elevated blood pressure (140/90 mm Hg) and tachycardia (115 beats per minute). Internal medicine clinicians note that Mrs. M is not in acute distress, although she refuses to speak and has a small amount of dried blood on her lips, presumably from a struggle with the police before coming to the hospital, but this is not certain. Her abdomen is not tender; she has normal bowel sounds, and no asterixis is noted on neurologic exam. Physical exam is otherwise normal. A noncontrast head CT scan shows no acute process. Initial lab values show elevations in ammonia (277 μg/dL) and γ-glutamyl transpeptidase (68 U/L). Thyroid-stimulating hormone is 1.45 mlU/L, prothrombin time is 19.5 s, partial thromboplastin time is 40.3 s, and international normalized ratio is 1.67. The internal medicine team admits Mrs. M to the intensive care unit (ICU) for further management of her mental status change with alcohol withdrawal or hepatic encephalopathy as the most likely etiologies.
Mrs. M’s husband says that his wife has not consumed alcohol in the last 4 months in preparation for a possible liver transplant; however, past interactions with Mrs. M’s family suggest they are unreliable. The Clinical Institute Withdrawal Assessment (CIWA) protocol is implemented in case her symptoms are caused by alcohol withdrawal. Her vital signs are stable and IV lorazepam, 4 mg, is administered once for agitation. Mrs. M’s husband also reports that 1 month ago his wife underwent a transjugular intrahepatic portosystemic shunt (TIPS) procedure for portal hypertension. Outpatient psychotropics (lamotrigine, 100 mg/d, and venlafaxine XR, 75 mg/d) are restarted because withdrawal from these drugs may exacerbate her symptoms. In the ICU Mrs. M experiences a tonic-clonic seizure with fecal incontinence and bitten tongue, which results in a consultation from neurology and the psychiatry consultation-liaison service.
Psychiatry recommends withholding psychotropics, stopping CIWA, and using vital sign parameters along with objective signs of diaphoresis and tremors as indicators of alcohol withdrawal for lorazepam administration. Mrs. M receives IV haloperidol, 1 mg, once during her second day in the hospital for severe agitation, but this medication is discontinued because of concern about lowering her seizure threshold.7 After treatment with lactulose, her ammonia levels trend down to 33 μg/dL, but her altered mental state persists with significant deficits in attention and orientation.
The neurology service performs an EEG that shows no slow-wave, triphasic waves, or epileptiform activity, which likely would be present in delirium or seizures. See Figure 1 for an example of triphasic waves on an EEG and Figure 2 for Mrs. M's EEG results. Subsequent lumbar puncture, MRI, and a second EEG are unremarkable. By the fifth hospital day, Mrs. M is calm and her paranoia has subsided, but she still is confused and disoriented. Psychiatry orders a third EEG while she is in this confused state; it shows no pathologic process. Based on these examinations, neurology posits that Mrs. M is not encephalopathic.
Figure 1: Representative sample of triphasic waves
This EEG tracing is from a 54-year-old woman who underwent prolonged abdominal surgery for lysis of adhesions during which she suffered an intraoperative left subinsular stroke followed by nonconvulsive status epilepticus. The tracing demonstrates typical morphology with the positive sharp transient preceded and followed by smaller amplitude negative deflections. Symmetric, frontal predominance of findings seen is this tracing is common
Figure 2: Mrs. M’s EEG results
This is a representative tracing of Mrs. M’s 3 EEGs revealing an 8.5 to 9 Hz dominant alpha rhythm. There is superimposed frontally dominant beta fast activity, which is consistent with known administration of benzodiazepines
The authors’ observations
Mrs. M had repeated admissions for alcohol dependence and subsequent liver failure. Her recent hospitalization was complicated by a TIPS procedure done 1 month ago. The incidence of hepatic encephalopathy in patients undergoing TIPS is >30%, especially in the first month post-procedure, which raised suspicion that hepatic encephalopathy played a significant role in Mrs. M’s delirium.8
Because of frequent hospitalization, Mrs. M was well known to the internal medicine, neurology, and psychiatry teams, and each used different terms to describe her mental state. Internal medicine used the phrase “acute mental status change,” which covers a broad differential. Neurology used “encephalopathy,” which also is a general term. Psychiatry used “delirium,” which has narrower and more specific diagnostic criteria. Engel et al9 described the delirious patient as having “cerebral insufficiency” with universally abnormal EEG. Regardless of terminology, based on Mrs. M’s acute confusion, one would expect an abnormal EEG, but repeat EEGs were unremarkable.
Interpreting EEG
EEG is one of the few tools available for measuring acute changes in cerebral function, and an EEG slowing remains a hallmark in encephalopathic processes.10,11 Initially, the 3 specialties agreed that Mrs. M’s presentation likely was caused by underlying medical issues or substances (alcohol or others). EEG can help recognize delirium, and, in some cases, elucidate the underlying cause.10,12 It was surprising that Mrs. M’s EEGs were normal despite a clinical presentation of delirium. Because of the normal EEG findings, neurology leaned toward a primary psychiatric (“functional”) etiology as the cause of her delirium vs a general medical condition or alcohol withdrawal (“organic”).
A literature search in regards to sensitivity of EEG in delirium revealed conflicting statements and data. A standard textbook in neurology and psychiatry states that “a normal EEG virtually excludes a toxic-metabolic encephalopathy.”13 The American Psychiatric Association’s (APA) practice guidelines for delirium states: “The presence of EEG abnormalities has fairly good sensitivities for delirium (in one study, the sensitivity was found to be 75%), but the absence does not rule out the diagnosis; thus the EEG is no substitute for careful clinical observation.”6
At the beginning of Mrs. M’s care, in discussion with the neurology and internal medicine teams, we argued that Mrs. M was experiencing delirium despite her initial normal EEG. We did not expect that 2 subsequent EEGs would be normal, especially because the teams witnessed the final EEG being performed while Mrs. M was clinically evaluated and observed to be in a state of delirium.
OUTCOME: Cause still unknown
By the 6th day of hospitalization, Mrs. M’s vitals are normal and she remains hemodynamically stable. Differential diagnosis remains wide and unclear. The psychiatry team feels she could have atypical catatonia due to an underlying mood disorder. One hour after a trial of IV lorazepam, 1 mg, Mrs. M is more lucid and fully oriented, with MMSE of 28/30 (recall was 1/3), indicating normal cognition. During the exam, a psychiatry resident notes Mrs. M winks and feigns a yawn at the medical students and nurses in the room, displaying her boredom with the interview and simplicity of the mental status exam questions. Later that evening, Mrs. M exhibits bizarre sexual gestures toward male hospital staff, including licking a male nursing staff member’s hand.
Although Mrs. M’s initial confusion resolved, the severity of her comorbid psychiatric history warrants inpatient psychiatric hospitalization. She agrees to transfer to the psychiatric ward after she confesses anxiety regarding death, intense demoralization, and guilt related to her condition and her relationship with her 12-year-old daughter. She tearfully reports that she discontinued her psychotropic medications shortly after stopping alcohol 4 months ago. Shortly before her transfer, psychiatry is called back to the medicine floor because of Mrs. M’s disruptive behavior.
The team finds Mrs. M in her hospital gown, pursuing her husband in the hallway as he is leaving, yelling profanities and blaming him for her horrible experience in the hospital. Based on her demeanor, the team determines that she is back to her baseline mental state despite her mood disorder, and that her upcoming inpatient psychiatric stay likely would be too short to address her comorbid personality disorder. The next day she signs out of the hospital against medical advice.
The authors’ observations
We never clearly identified the specific etiology responsible for Mrs. M’s delirium. We assume at the initial presentation she had toxic-metabolic encephalopathy that rapidly resolved with lactulose treatment and lowering her ammonia. She then had a single tonic-clonic seizure, perhaps related to stopping and then restarting her psychotropics. Her subsequent confusion, bizarre sexual behavior, and demeanor on her final hospital days were more indicative of her psychiatric diagnoses. We now suspect that Mrs. M’s delirium was briefer than presumed and she returned to her baseline borderline personality, resulting in some factitious staging of delirium to confuse her 3 treating teams (a psychoanalyst may say this was a form of projective identification).
We felt that if Mrs. M truly was delirious due to metabolic or hepatic dysfunction or alcohol withdrawal, she would have had abnormal EEG findings. We discovered that the notion of “75% sensitivity” of EEG abnormalities cited in the APA guidelines comes from studies that include patients with “psychogenic” and “organic” delirium. Acute manias and agitated psychoses were termed “psychogenic delirium” and acute confusion due to medical conditions or substance issues was termed “organic delirium.”9,12,14-16
This poses a circular reasoning in the diagnostic criteria and clinical approach to delirium. The fallacy is that, according to DSM-IV-TR, delirium is supposed to be the result of a direct physiological consequence of a general medical condition or substance use (criterion D), and cannot be due to psychosis (eg, schizophrenia) or mania (eg, BD). We question the presumptive 75% sensitivity of EEG abnormalities in patients with delirium because it is possible that when some of these studies were conducted the definition of delirium was not solidified or fully understood. We suspect the sensitivity would be much higher if the correct definition of delirium according to DSM-IV-TR is used in future studies. To improve interdisciplinary communication and future research, it would be constructive if all disciplines could agree on a single term, with the same diagnostic criteria, when evaluating a patient with acute confusion.
Related Resources
- Meagher D. Delirium: the role of psychiatry. Advances in Psychiatric Treatment. 2001;7:433-442.
- Casey DA, DeFazio JV Jr, Vansickle K, et al. Delirium. Quick recognition, careful evaluation, and appropriate treatment. Postgrad Med. 1996;100(1):121-4, 128, 133-134.
Drug Brand Names
- Clonazepam • Klonopin
- Docusate • Surfak
- Haloperidol • Haldol
- Lamotrigine • Lamictal
- Lorazepam • Ativan
- Levothyroxine • Levoxyl, Synthtoid
- Venlafaxine XR • Effexor XR
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
Acknowledgment
The views expressed in this article are those of the authors and do not necessarily reflect the official policy or position of the Department of the Navy, Department of Defense, or the U.S. Government. The authors are employees of the U.S. Government. This work was prepared as part of their official duties. Title 17 U.S.C. 105 provides that “Copyright protection under this title is not available for any work of the U.S. Government.” Title 17 U.S.C. 101 defines a U.S. Government work as a work prepared by a military service member or employee of the U.S. Government as part of that person’s official duties.
1. Katz IR, Mossey J, Sussman N, et al. Bedside clinical and electrophysiological assessment: assessment of change in vulnerable patients. Int Psychogeriatr. 1991;3(2):289-300.
2. Inouye SK. Delirium in older persons. N Engl J Med. 2006;354(11):1157-1165.
3. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.
4. McPhee SJ, Papadakis M, Rabow MW. CURRENT medical diagnosis and treatment. New York NY: McGraw Hill Medical; 2012.
5. Brody B. Who has capacity? N Engl J Med. 2009;361(3):232-233.
6. Practice guideline for the treatment of patients with delirium. American Psychiatric Association. Am J Psychiatry. 1999;156(5 suppl):1-20.
7. Fricchione GL, Nejad SH, Esses JA, et al. Postoperative delirium. Am J Psychiatry. 2008;165(7):803-812.
8. Sanyal AJ, Freedman AM, Shiffman ML, et al. Portosystemic encephalopathy after transjugular intrahepatic portosystemic shunt: results of a prospective controlled study. Hepatology. 1994;20(1 pt 1):46-55.
9. Engel GL, Romano J. Delirium a syndrome of cerebral insufficiency. 1959. J Neuropsychiatry Clin Neurosci. 2004;16(4):526-538.
10. Pro JD, Wells CE. The use of the electroencephalogram in the diagnosis of delirium. Dis Nerv Syst. 1977;38(10):804-808.
11. Sidhu KS, Balon R, Ajluni V, et al. Standard EEG and the difficult-to-assess mental status. Ann Clin Psychiatry. 2009;21(2):103-108.
12. Brenner RP. Utility of EEG in delirium: past views and current practice. Int Psychogeriatr. 1991;3(2):211-229.
13. Kaufman DM. Clinical neurology for psychiatrists. 5th ed. Philadelphia PA: Saunders; 2001: 230-232.
14. Bond TC. Recognition of acute delirious mania. Arch Gen Psychiatry. 1980;37(5):553-554.
15. Krauthammer C, Klerman GL. Secondary mania: manic syndromes associated with antecedent physical illness or drugs. Arch Gen Psychiatry. 1978;35(11):1333-1339.
16. Larson EW, Richelson E. Organic causes of mania. Mayo Clin Proc. 1988;63(9):906-912.
1. Katz IR, Mossey J, Sussman N, et al. Bedside clinical and electrophysiological assessment: assessment of change in vulnerable patients. Int Psychogeriatr. 1991;3(2):289-300.
2. Inouye SK. Delirium in older persons. N Engl J Med. 2006;354(11):1157-1165.
3. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.
4. McPhee SJ, Papadakis M, Rabow MW. CURRENT medical diagnosis and treatment. New York NY: McGraw Hill Medical; 2012.
5. Brody B. Who has capacity? N Engl J Med. 2009;361(3):232-233.
6. Practice guideline for the treatment of patients with delirium. American Psychiatric Association. Am J Psychiatry. 1999;156(5 suppl):1-20.
7. Fricchione GL, Nejad SH, Esses JA, et al. Postoperative delirium. Am J Psychiatry. 2008;165(7):803-812.
8. Sanyal AJ, Freedman AM, Shiffman ML, et al. Portosystemic encephalopathy after transjugular intrahepatic portosystemic shunt: results of a prospective controlled study. Hepatology. 1994;20(1 pt 1):46-55.
9. Engel GL, Romano J. Delirium a syndrome of cerebral insufficiency. 1959. J Neuropsychiatry Clin Neurosci. 2004;16(4):526-538.
10. Pro JD, Wells CE. The use of the electroencephalogram in the diagnosis of delirium. Dis Nerv Syst. 1977;38(10):804-808.
11. Sidhu KS, Balon R, Ajluni V, et al. Standard EEG and the difficult-to-assess mental status. Ann Clin Psychiatry. 2009;21(2):103-108.
12. Brenner RP. Utility of EEG in delirium: past views and current practice. Int Psychogeriatr. 1991;3(2):211-229.
13. Kaufman DM. Clinical neurology for psychiatrists. 5th ed. Philadelphia PA: Saunders; 2001: 230-232.
14. Bond TC. Recognition of acute delirious mania. Arch Gen Psychiatry. 1980;37(5):553-554.
15. Krauthammer C, Klerman GL. Secondary mania: manic syndromes associated with antecedent physical illness or drugs. Arch Gen Psychiatry. 1978;35(11):1333-1339.
16. Larson EW, Richelson E. Organic causes of mania. Mayo Clin Proc. 1988;63(9):906-912.
Drug interactions with tobacco smoke: Implications for patient care
- Tobacco smokers often are treated with medications that are metabolized by hepatic cytochrome (CYP) 1A2 enzymes. Starting or stopping tobacco smoking may cause drug interactions because polycyclic aromatic hydrocarbons in cigarette smoke induce CYP1A2 enzymes.
- Drugs that are significantly metabolized by CYP1A2 (major substrates) are more likely to be impacted by changes in tobacco smoking compared with minor substrates.
- Induction of hepatic CYP1A2 enzymes may be greater in heavy or moderate smokers compared with light smokers (eg, <10 cigarettes per day).
- Evidence-based approaches for treating tobacco use in health care settings should address the risk of CYP1A2 drug interactions in tobacco smokers and how this impacts their clinical care.
Mrs. C, age 51, experiences exacerbated asthma and difficulty breathing and is admitted to a non-smoking hospital. She also has chronic obstructive pulmonary disease, type 2 diabetes mellitus, hypertension, hypercholesterolemia, hypothyroidism, gastroesophageal reflux disease, overactive bladder, muscle spasms, fibromyalgia, bipolar disorder, insomnia, and nicotine and caffeine dependence. She takes 19 prescribed and over-the-counter medications, drinks up to 8 cups of coffee per day, and smokes 20 to 30 cigarettes per day. In the emergency room, she receives albuterol/ipratropium inhalation therapy to help her breathing and a 21-mg nicotine replacement patch to avoid nicotine withdrawal.
In the United States, 19% of adults smoke cigarettes.1 Heavy tobacco smoking and nicotine dependence are common among psychiatric patients and contribute to higher rates of tobacco-related morbidity and mortality.2 When smokers stop smoking or are admitted to smoke-free facilities and are forced to abstain, nicotine withdrawal symptoms and changes in drug metabolism can develop over several days.3-5
Smokers such as Mrs. C are at risk for cytochrome (CYP) P450 drug interactions when they are admitted to or discharged from a smoke-free facility. Nine of Mrs. C’s medications are substrates of CYP1A2 (acetaminophen, caffeine, cyclobenzaprine, diazepam, duloxetine, melatonin, olanzapine, ondansetron, and zolpidem). When Mrs. C stops smoking while in the hospital, she could experience higher serum concentrations and adverse effects of these medications. If Mrs. C resumes smoking after bring discharged, metabolism and clearance of any medications started while she was hospitalized that are substrates of CYP1A2 enzymes could be increased, which could lead to reduced efficacy and poor clinical outcomes.
Pharmacokinetic effects
Polycyclic aromatic hydrocarbons in tobacco smoke induce hepatic CYP1A1, 1A2, and possibly 2E1 isoenzymes.6-12 CYP1A2 is a hepatic enzyme responsible for metabolizing and eliminating several classes of substrates (eg, drugs, hormones, endogenous compounds, and procarcinogens).6,13 Genetic, epigenetic, and environmental factors such as smoking impact the expression and activity of CYP1A2 and result in large interpatient variability in pharmacokinetic drug interactions.6,12,13 CYP1A2 enzymes can be induced or inhibited by drugs and substances, which can result in decreased or increased serum concentrations of substrates, respectively. When individuals stop smoking and switch to other nicotine products or devices, CYP1A2 induction of hepatic enzymes will revert to normal metabolism over several weeks to a month.10 Besides tobacco smoke, other CYP1A2 inducers include charbroiled food, carbamazepine, omeprazole, phenobarbital, primidone, and rifampin.4,5 Nicotine replacement products—such as gum, inhalers, lozenges, patches, and nasal spray—and nicotine delivery devices such as electronic cigarettes do not induce hepatic CYP1A2 enzymes or cause the same drug interactions as cigarette smoking.
Table 13-11 and Table 23-11 list commonly prescribed CYP1A2 substrates that could be affected by tobacco smoke. There are no specific guidelines for how to assess, monitor, or manage pharmacokinetic drug interactions with tobacco smoke.6-13 Induction of hepatic CYP1A2 enzymes by cigarette smoke may require increased dosages of some psychotropics—such as tricyclic antidepressants, duloxetine, mirtazapine, and some first- and second-generation antipsychotics (SGAs)—to achieve serum concentrations adequate for clinical efficacy. Serum concentrations may increase to toxic levels and result in adverse effects when a person quits smoking cigarettes or if a CYP1A2 inhibitor, such as amlodipine, cimetidine, ciprofloxacin, diclofenac, fluoxetine, fluvoxamine, or nifedipine, is added.5
Table 1
Common major cytochrome P450 (CYP) 1A2 substrates
Drug | Class |
---|---|
Alosetron3,5,6 | Irritable bowel syndrome: serotonin 3 antagonist |
Aminophylline3,5 | Bronchodilator: theophylline derivative |
Betaxolol3,5 | β-1 selective adrenergic receptor blocking agent |
Caffeine3-9 | Stimulant |
Clomipramine3-11 | Tricyclic antidepressant |
Clozapine3-10 | Second-generation antipsychotic |
Cyclobenzaprine3-7 | Skeletal muscle relaxant |
Doxepin3,7,10,11 | Tricyclic antidepressant |
Duloxetine3-6 | Serotonin-norepinephrine reuptake inhibitor |
Estradiol3,5-8 | Estrogen (active) |
Estrogens: conjugated and estropipate3,5; estrone3,7 | Estrogen (derivatives) |
Fluvoxamine3,8,9 | Selective serotonin reuptake inhibitor |
Guanabenz3,5-7 | α-2 adrenergic agonist |
Mirtazapine3-7 | Antidepressant: α-2 antagonist/serotonin 2A, 2C antagonist |
Olanzapine3-11 | Second-generation antipsychotic |
Pimozide3,5,7 | First-generation antipsychotic |
Propranolol3-11 | β-adrenergic blocker |
Ramelteon3,5,10 | Melatonin receptor agonist |
Rasagiline3,5 | Antiparkinson: type B monoamine oxidase inhibitor |
Riluzole3-7,10 | Glutamate inhibitor |
Ropinirole3,5-7 | Antiparkinson: dopamine agonist |
Theophylline3-6,8-11 | Bronchodilator: methylxanthine |
Thiothixene3,5 | First-generation antipsychotic |
Trifluoperazine3,5,9 | First-generation antipsychotic |
Several classes of CYP1A2 substrates are not included and may cause toxicity with smoking cessation or require dosage increases in tobacco smokers (eg, antiarrhythmic, antifungal, antimalarial, antineoplastic, antiretroviral, and anthelmintic agents and the antibiotic quinolone). Clinicians should be most concerned about drugs with a narrow therapeutic index and those that may be toxic with smoking cessation (eg, bleeding from warfarin and clopidogrel; high serum concentrations of caffeine, clozapine, olanzapine, propranolol, and theophylline) |
Table 2
Common minor cytochrome P450 (CYP) 1A2 substrates
Drug | Class |
---|---|
Acetaminophen3-9 | Analgesic |
Almotriptan6 | Antimigraine: serotonin 1B, 1D receptor agonist |
Amitriptyline3-7,9-11 | Tricyclic antidepressant |
Asenapine9 | Second-generation antipsychotic |
Carvedilol5-7 | β and α adrenergic blocking activity |
Chlorpromazine3,4,7-9,11 | First-generation antipsychotic |
Chlorzoxazone4,7 | Skeletal muscle relaxant |
Clopidogrel5 | Antiplatelet |
Desipramine4,7,10,11 | Tricyclic antidepressant |
Diazepam4,7,9,10 | Benzodiazepine |
Diclofenac5,7 | Nonsteroidal anti-inflammatory drug |
Diphenhydramine6 | Antihistamine |
Febuxostat5 | Xanthine oxidase inhibitor |
Fluphenazine3,9 | First-generation antipsychotic |
Frovatriptan3 | Antimigraine: serotonin 1 agonist |
Haloperidol3,4,6,8,9 | First-generation antipsychotic |
Imipramine3,4,6-11 | Tricyclic antidepressant |
Maprotiline6 | Tetracyclic antidepressant |
Melatonin3,4,6,7 | Sleep-regulating hormone |
Metoclopramide3 | Antiemetic: prokinetic gastrointestinal agent |
Nabumetone6 | Nonsteroidal anti-inflammatory drug |
Naproxen3,4,6,7 | Nonsteroidal anti-inflammatory drug |
Naratriptan10 | Antimigraine: serotonin 1B, 1D receptor agonist |
Nicardipine3,7 | Calcium channel blocker |
Nortriptyline4,6,7,9-11 | Tricyclic antidepressant |
Ondansetron3,4,6,7 | Antiemetic: serotonin 3 antagonist |
Palonosetron5 | Antiemetic: serotonin 3 antagonist |
Perphenazine3,7 | First-generation antipsychotic |
Progesterone5,7 | Progestin |
Propofol4,6,7 | General anesthetic |
Ranitidine5,7 | H2 antagonist |
Rivastigmine10 | Acetylcholinesterase inhibitor |
Selegiline6,7 | Antiparkinson: type B monoamine oxidase inhibitor |
Thioridazine3,4,6 | First-generation antipsychotic |
Tizanidine3-6 | Skeletal muscle relaxant: α-2 adrenergic agonist |
Trazodone6,9 | Serotonin reuptake inhibitor and antagonist |
Triamterene6 | Diuretic: potassium sparing |
Verapamil3,4,6,7,10 | Calcium channel blocker |
Warfarin3,4,6-10 | Anticoagulant: coumarin derivative |
Zileuton3,4,6,7 | Asthma agent: 5-lipoxygenase inhibitor |
Ziprasidone3,4 | Second-generation antipsychotic |
Zolmitriptan3,6,7 | Antimigraine: serotonin 1B, 1D receptor agonist |
Zolpidem4,6,7 | Nonbenzodiazepine hypnotic |
Several classes of CYP1A2 substrates are not included and may cause toxicity with smoking cessation or require dosage increases in tobacco smokers (eg, antiarrhythmic, antifungal, antimalarial, antineoplastic, antiretroviral and anthelmintic agents and the antibiotic quinolone). Clinicians should be most concerned about drugs with a narrow therapeutic index and those that may be toxic with smoking cessation (eg, bleeding from warfarin and clopidogrel; high serum concentrations of caffeine, clozapine, olanzapine, propranolol, and theophylline) |
SGA such as clozapine and olanzapine are major substrates of CYP1A2 and dosages may need to be adjusted when smoking status changes, depending on clinical efficacy and adverse effects.10,14,15 Maximum induction of clozapine and olanzapine metabolism may occur with 7 to 12 cigarettes per day and smokers may have 40% to 50% lower serum concentrations compared with nonsmokers.14 When a patient stops smoking, clozapine and olanzapine dosages may need to be reduced by 30% to 40% (eg, a stepwise 10% reduction in daily dose until day 4) to avoid elevated serum concentrations and risk of toxicity symptoms.15
Tobacco smokers can tolerate high daily intake of caffeinated beverages because of increased metabolism and clearance of caffeine, a major substrate of CYP1A2.11 When patients stop smoking, increased caffeine serum concentrations may cause anxiety, irritability, restlessness, insomnia, tremors, palpitations, and tachycardia. Caffeine toxicity also can mimic symptoms of nicotine withdrawal; therefore, smokers should be advised to reduce their caffeine intake by half to avoid adverse effects when they stop smoking.10,11
Adjusting dosing
Factors such as the amount and frequency of tobacco smoking, how quickly CYP1A2 enzymes change when starting and stopping smoking, exposure to secondhand smoke, and other concomitant drugs contribute to variability in pharmacokinetic drug interactions. Heavy smokers (≥30 cigarettes per day) should be closely monitored because variations in drug serum concentrations may be affected significantly by changes in smoking status.4,9,11 Dosage reductions of potentially toxic drugs should be done immediately when a heavy tobacco user stops smoking.10 For CYP1A2 substrates with a narrow therapeutic range, a conservative approach is to reduce the daily dose by 10% per day for several days after smoking cessation.11,16 The impact on drug metabolism may continue for weeks to a month after the person stops smoking; therefore, there may be a delay until CYP1A2 enzymes return to normal hepatic metabolism.4,8,9,15 In most situations, smoking cessation reverses induction of hepatic CYP1A2 enzymes back to normal metabolism and causes serum drug concentrations to increase.10 Because secondhand smoke induces hepatic CYP1A2 enzymes, those exposed to smoke may have changes in drug metabolism due to environmental smoke exposure.11
Tobacco smokers who take medications and hormones that are metabolized by CYP1A2 enzymes should be closely monitored because dosage adjustments may be necessary when they start or stop smoking cigarettes. An assessment of CYP drug interactions and routine monitoring of efficacy and/or toxicity should be done to avoid potential adverse effects from medications and to determine if changes in dosages and disease state management are required.
Related Resources
- Rx for Change. Drug interactions with smoking. http://smokingcessationleadership.ucsf.edu/interactions.pdf.
- Fiore MC, Baker TB. Treating smokers in the health care setting. N Engl J Med. 2011;365(13):1222-1231.
Drug Brand Names
- Albuterol/ipratropium • Combivent
- Almotriptan • Axert
- Alosetron • Lotronex
- Aminophylline • Phyllocontin, Truphylline
- Amitriptyline • Elavil
- Amlodipine • Norvasc
- Asenapine • Saphris
- Betaxolol • Kerlone
- Carbamazepine • Carbatrol, Tegretol
- Carvedilol • Coreg
- Chlorpromazine • Thorazine
- Chlorzoxazone • Parafon Forte
- Cimetidine • Tagamet
- Ciprofloxacin • Cipro
- Clomipramine • Anafranil
- Clopidogrel • Plavix
- Clozapine • Clozaril
- Cyclobenzaprine • Flexeril
- Desipramine • Norpramin
- Diazepam • Valium
- Diclofenac • Voltaren
- Diphenhydramine • Benadryl
- Doxepin • Silenor, Sinequan
- Duloxetine • Cymbalta
- Estradiol • Estrace
- Estrogens (conjugated) • Cenestin, Premarin
- Estropipate • Ogen
- Febuxostat • Uloric
- Fluoxetine • Prozac
- Fluphenazine • Prolixin
- Fluvoxamine • Luvox
- Frovatriptan • Frova
- Guanabenz • Wytensin
- Haloperidol • Haldol
- Imipramine • Tofranil
- Maprotiline • Ludiomil
- Metoclopramide • Reglan
- Mirtazapine • Remeron
- Nabumetone • Relafen
- Naratriptan • Amerge
- Nicardipine • Cardene
- Nifedipine • Adalat, Procardia
- Nortriptyline • Aventyl, Pamelor
- Olanzapine • Zyprexa
- Omeprazole • Prilosec
- Ondansetron • Zofran
- Palonosetron • Aloxi
- Perphenazine • Trilafon
- Pimozide • Orap
- Primidone • Mysoline
- Progesterone • Prometrium
- Propofol • Diprivan
- Propranolol • Inderal
- Ramelteon • Rozerem
- Ranitidine • Zantac
- Rasagiline • Azilect
- Rifampin • Rifadin, Rimactane
- Riluzole • Rilutek
- Rivastigmine • Exelon
- Ropinirole • Requip
- Selegiline • Eldepryl, EMSAM, others
- Theophylline • Elixophyllin
- Thioridazine • Mellaril
- Thiothixene • Navane
- Tizanidine • Zanaflex
- Trazodone • Desyrel, Oleptro
- Triamterene • Dyrenium
- Trifluoperazine • Stelazine
- Verapamil • Calan, Verelan
- Warfarin • Coumadin, Jantoven
- Zileuton • Zyflo
- Ziprasidone • Geodon
- Zolmitriptan • Zomig
- Zolpidem • Ambien, Edluar
Disclosure
Ms. Fankhauser reports no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Centers for Disease Control and Prevention (CDC). Vital signs: current cigarette smoking among adults aged ≥18 years—United States 2005-2010. MMWR Morb Mortal Wkly Rep. 2011;60(35):1207-1212.
2. Ziedonis D, Hitsman B, Beckham JC, et al. Tobacco use and cessation in psychiatric disorders: National Institute of Mental Health report. Nicotine Tob Res. 2008;10(12):1691-1715.
3. Choe JY. Drug actions and interactions. New York NY: McGraw-Hill Medical; 2011.
4. Tatro DS. Drug interaction facts. St. Louis MO: Wolters Kluwer Health; 2011.
5. Lacy CF, Armstrong LL, Goldman MP, et al. eds. Drug information handbook, 20th ed. Hudson, OH: Lexicomp; 2011.
6. Zhou SF, Yang LP, Zhou ZW, et al. Insights into the substrate specificity, inhibitors, regulation, and polymorphisms and the clinical impact of human cytochrome P450 1A2. AAPS J. 2009;11(3):481-494.
7. Rendic S. Summary of information on human CYP enzymes: human P450 metabolism data. Drug Metab Rev. 2002;34(1-2):83-448.
8. Zevin S, Benowitz NL. Drug interactions with tobacco smoking. An update. Clin Pharmacokinet. 1999;36(6):425-438.
9. Desai HD, Seabolt J, Jann MW. Smoking in patients receiving psychotropic medications: a pharmacokinetic perspective. CNS Drugs. 2001;15(6):469-494.
10. Schaffer SD, Yoon S, Zadezensky I. A review of smoking cessation: potentially risky effects on prescribed medications. J Clin Nurs. 2009;18(11):1533-1540.
11. Kroon LA. Drug interactions with smoking. Am J Health Syst Pharm. 2007;64(18):1917-1921.
12. Plowchalk DR, Yeo KR. Prediction of drug clearance in a smoking population: modeling the impact of variable cigarette consumption on the induction of CYP1A2. Eur J Pharmacol. 2012;68(6):951-960.
13. Faber MS, Jetter A, Fuhr U. Assessment of CYP1A2 activity in clinical practice: why how, and when? Basic Clin Pharmacol Toxicol. 2005;97(3):125-134.
14. Haslemo T, Eikeseth PH, Tanum L, et al. The effect of variable cigarette consumption on the interaction with clozapine and olanzapine. Eur J Clin Pharmacol. 2006;62(12):1049-1053.
15. Lowe EJ, Ackman ML. Impact of tobacco smoking cessation on stable clozapine or olanzapine treatment. Ann Pharmacother. 2010;44(4):727-732.
16. Faber MS, Fuhr U. Time response of cytochrome P4501A2 activity on cessation of heavy smoking. Clin Pharmacol Ther. 2004;76(2):178-184.
- Tobacco smokers often are treated with medications that are metabolized by hepatic cytochrome (CYP) 1A2 enzymes. Starting or stopping tobacco smoking may cause drug interactions because polycyclic aromatic hydrocarbons in cigarette smoke induce CYP1A2 enzymes.
- Drugs that are significantly metabolized by CYP1A2 (major substrates) are more likely to be impacted by changes in tobacco smoking compared with minor substrates.
- Induction of hepatic CYP1A2 enzymes may be greater in heavy or moderate smokers compared with light smokers (eg, <10 cigarettes per day).
- Evidence-based approaches for treating tobacco use in health care settings should address the risk of CYP1A2 drug interactions in tobacco smokers and how this impacts their clinical care.
Mrs. C, age 51, experiences exacerbated asthma and difficulty breathing and is admitted to a non-smoking hospital. She also has chronic obstructive pulmonary disease, type 2 diabetes mellitus, hypertension, hypercholesterolemia, hypothyroidism, gastroesophageal reflux disease, overactive bladder, muscle spasms, fibromyalgia, bipolar disorder, insomnia, and nicotine and caffeine dependence. She takes 19 prescribed and over-the-counter medications, drinks up to 8 cups of coffee per day, and smokes 20 to 30 cigarettes per day. In the emergency room, she receives albuterol/ipratropium inhalation therapy to help her breathing and a 21-mg nicotine replacement patch to avoid nicotine withdrawal.
In the United States, 19% of adults smoke cigarettes.1 Heavy tobacco smoking and nicotine dependence are common among psychiatric patients and contribute to higher rates of tobacco-related morbidity and mortality.2 When smokers stop smoking or are admitted to smoke-free facilities and are forced to abstain, nicotine withdrawal symptoms and changes in drug metabolism can develop over several days.3-5
Smokers such as Mrs. C are at risk for cytochrome (CYP) P450 drug interactions when they are admitted to or discharged from a smoke-free facility. Nine of Mrs. C’s medications are substrates of CYP1A2 (acetaminophen, caffeine, cyclobenzaprine, diazepam, duloxetine, melatonin, olanzapine, ondansetron, and zolpidem). When Mrs. C stops smoking while in the hospital, she could experience higher serum concentrations and adverse effects of these medications. If Mrs. C resumes smoking after bring discharged, metabolism and clearance of any medications started while she was hospitalized that are substrates of CYP1A2 enzymes could be increased, which could lead to reduced efficacy and poor clinical outcomes.
Pharmacokinetic effects
Polycyclic aromatic hydrocarbons in tobacco smoke induce hepatic CYP1A1, 1A2, and possibly 2E1 isoenzymes.6-12 CYP1A2 is a hepatic enzyme responsible for metabolizing and eliminating several classes of substrates (eg, drugs, hormones, endogenous compounds, and procarcinogens).6,13 Genetic, epigenetic, and environmental factors such as smoking impact the expression and activity of CYP1A2 and result in large interpatient variability in pharmacokinetic drug interactions.6,12,13 CYP1A2 enzymes can be induced or inhibited by drugs and substances, which can result in decreased or increased serum concentrations of substrates, respectively. When individuals stop smoking and switch to other nicotine products or devices, CYP1A2 induction of hepatic enzymes will revert to normal metabolism over several weeks to a month.10 Besides tobacco smoke, other CYP1A2 inducers include charbroiled food, carbamazepine, omeprazole, phenobarbital, primidone, and rifampin.4,5 Nicotine replacement products—such as gum, inhalers, lozenges, patches, and nasal spray—and nicotine delivery devices such as electronic cigarettes do not induce hepatic CYP1A2 enzymes or cause the same drug interactions as cigarette smoking.
Table 13-11 and Table 23-11 list commonly prescribed CYP1A2 substrates that could be affected by tobacco smoke. There are no specific guidelines for how to assess, monitor, or manage pharmacokinetic drug interactions with tobacco smoke.6-13 Induction of hepatic CYP1A2 enzymes by cigarette smoke may require increased dosages of some psychotropics—such as tricyclic antidepressants, duloxetine, mirtazapine, and some first- and second-generation antipsychotics (SGAs)—to achieve serum concentrations adequate for clinical efficacy. Serum concentrations may increase to toxic levels and result in adverse effects when a person quits smoking cigarettes or if a CYP1A2 inhibitor, such as amlodipine, cimetidine, ciprofloxacin, diclofenac, fluoxetine, fluvoxamine, or nifedipine, is added.5
Table 1
Common major cytochrome P450 (CYP) 1A2 substrates
Drug | Class |
---|---|
Alosetron3,5,6 | Irritable bowel syndrome: serotonin 3 antagonist |
Aminophylline3,5 | Bronchodilator: theophylline derivative |
Betaxolol3,5 | β-1 selective adrenergic receptor blocking agent |
Caffeine3-9 | Stimulant |
Clomipramine3-11 | Tricyclic antidepressant |
Clozapine3-10 | Second-generation antipsychotic |
Cyclobenzaprine3-7 | Skeletal muscle relaxant |
Doxepin3,7,10,11 | Tricyclic antidepressant |
Duloxetine3-6 | Serotonin-norepinephrine reuptake inhibitor |
Estradiol3,5-8 | Estrogen (active) |
Estrogens: conjugated and estropipate3,5; estrone3,7 | Estrogen (derivatives) |
Fluvoxamine3,8,9 | Selective serotonin reuptake inhibitor |
Guanabenz3,5-7 | α-2 adrenergic agonist |
Mirtazapine3-7 | Antidepressant: α-2 antagonist/serotonin 2A, 2C antagonist |
Olanzapine3-11 | Second-generation antipsychotic |
Pimozide3,5,7 | First-generation antipsychotic |
Propranolol3-11 | β-adrenergic blocker |
Ramelteon3,5,10 | Melatonin receptor agonist |
Rasagiline3,5 | Antiparkinson: type B monoamine oxidase inhibitor |
Riluzole3-7,10 | Glutamate inhibitor |
Ropinirole3,5-7 | Antiparkinson: dopamine agonist |
Theophylline3-6,8-11 | Bronchodilator: methylxanthine |
Thiothixene3,5 | First-generation antipsychotic |
Trifluoperazine3,5,9 | First-generation antipsychotic |
Several classes of CYP1A2 substrates are not included and may cause toxicity with smoking cessation or require dosage increases in tobacco smokers (eg, antiarrhythmic, antifungal, antimalarial, antineoplastic, antiretroviral, and anthelmintic agents and the antibiotic quinolone). Clinicians should be most concerned about drugs with a narrow therapeutic index and those that may be toxic with smoking cessation (eg, bleeding from warfarin and clopidogrel; high serum concentrations of caffeine, clozapine, olanzapine, propranolol, and theophylline) |
Table 2
Common minor cytochrome P450 (CYP) 1A2 substrates
Drug | Class |
---|---|
Acetaminophen3-9 | Analgesic |
Almotriptan6 | Antimigraine: serotonin 1B, 1D receptor agonist |
Amitriptyline3-7,9-11 | Tricyclic antidepressant |
Asenapine9 | Second-generation antipsychotic |
Carvedilol5-7 | β and α adrenergic blocking activity |
Chlorpromazine3,4,7-9,11 | First-generation antipsychotic |
Chlorzoxazone4,7 | Skeletal muscle relaxant |
Clopidogrel5 | Antiplatelet |
Desipramine4,7,10,11 | Tricyclic antidepressant |
Diazepam4,7,9,10 | Benzodiazepine |
Diclofenac5,7 | Nonsteroidal anti-inflammatory drug |
Diphenhydramine6 | Antihistamine |
Febuxostat5 | Xanthine oxidase inhibitor |
Fluphenazine3,9 | First-generation antipsychotic |
Frovatriptan3 | Antimigraine: serotonin 1 agonist |
Haloperidol3,4,6,8,9 | First-generation antipsychotic |
Imipramine3,4,6-11 | Tricyclic antidepressant |
Maprotiline6 | Tetracyclic antidepressant |
Melatonin3,4,6,7 | Sleep-regulating hormone |
Metoclopramide3 | Antiemetic: prokinetic gastrointestinal agent |
Nabumetone6 | Nonsteroidal anti-inflammatory drug |
Naproxen3,4,6,7 | Nonsteroidal anti-inflammatory drug |
Naratriptan10 | Antimigraine: serotonin 1B, 1D receptor agonist |
Nicardipine3,7 | Calcium channel blocker |
Nortriptyline4,6,7,9-11 | Tricyclic antidepressant |
Ondansetron3,4,6,7 | Antiemetic: serotonin 3 antagonist |
Palonosetron5 | Antiemetic: serotonin 3 antagonist |
Perphenazine3,7 | First-generation antipsychotic |
Progesterone5,7 | Progestin |
Propofol4,6,7 | General anesthetic |
Ranitidine5,7 | H2 antagonist |
Rivastigmine10 | Acetylcholinesterase inhibitor |
Selegiline6,7 | Antiparkinson: type B monoamine oxidase inhibitor |
Thioridazine3,4,6 | First-generation antipsychotic |
Tizanidine3-6 | Skeletal muscle relaxant: α-2 adrenergic agonist |
Trazodone6,9 | Serotonin reuptake inhibitor and antagonist |
Triamterene6 | Diuretic: potassium sparing |
Verapamil3,4,6,7,10 | Calcium channel blocker |
Warfarin3,4,6-10 | Anticoagulant: coumarin derivative |
Zileuton3,4,6,7 | Asthma agent: 5-lipoxygenase inhibitor |
Ziprasidone3,4 | Second-generation antipsychotic |
Zolmitriptan3,6,7 | Antimigraine: serotonin 1B, 1D receptor agonist |
Zolpidem4,6,7 | Nonbenzodiazepine hypnotic |
Several classes of CYP1A2 substrates are not included and may cause toxicity with smoking cessation or require dosage increases in tobacco smokers (eg, antiarrhythmic, antifungal, antimalarial, antineoplastic, antiretroviral and anthelmintic agents and the antibiotic quinolone). Clinicians should be most concerned about drugs with a narrow therapeutic index and those that may be toxic with smoking cessation (eg, bleeding from warfarin and clopidogrel; high serum concentrations of caffeine, clozapine, olanzapine, propranolol, and theophylline) |
SGA such as clozapine and olanzapine are major substrates of CYP1A2 and dosages may need to be adjusted when smoking status changes, depending on clinical efficacy and adverse effects.10,14,15 Maximum induction of clozapine and olanzapine metabolism may occur with 7 to 12 cigarettes per day and smokers may have 40% to 50% lower serum concentrations compared with nonsmokers.14 When a patient stops smoking, clozapine and olanzapine dosages may need to be reduced by 30% to 40% (eg, a stepwise 10% reduction in daily dose until day 4) to avoid elevated serum concentrations and risk of toxicity symptoms.15
Tobacco smokers can tolerate high daily intake of caffeinated beverages because of increased metabolism and clearance of caffeine, a major substrate of CYP1A2.11 When patients stop smoking, increased caffeine serum concentrations may cause anxiety, irritability, restlessness, insomnia, tremors, palpitations, and tachycardia. Caffeine toxicity also can mimic symptoms of nicotine withdrawal; therefore, smokers should be advised to reduce their caffeine intake by half to avoid adverse effects when they stop smoking.10,11
Adjusting dosing
Factors such as the amount and frequency of tobacco smoking, how quickly CYP1A2 enzymes change when starting and stopping smoking, exposure to secondhand smoke, and other concomitant drugs contribute to variability in pharmacokinetic drug interactions. Heavy smokers (≥30 cigarettes per day) should be closely monitored because variations in drug serum concentrations may be affected significantly by changes in smoking status.4,9,11 Dosage reductions of potentially toxic drugs should be done immediately when a heavy tobacco user stops smoking.10 For CYP1A2 substrates with a narrow therapeutic range, a conservative approach is to reduce the daily dose by 10% per day for several days after smoking cessation.11,16 The impact on drug metabolism may continue for weeks to a month after the person stops smoking; therefore, there may be a delay until CYP1A2 enzymes return to normal hepatic metabolism.4,8,9,15 In most situations, smoking cessation reverses induction of hepatic CYP1A2 enzymes back to normal metabolism and causes serum drug concentrations to increase.10 Because secondhand smoke induces hepatic CYP1A2 enzymes, those exposed to smoke may have changes in drug metabolism due to environmental smoke exposure.11
Tobacco smokers who take medications and hormones that are metabolized by CYP1A2 enzymes should be closely monitored because dosage adjustments may be necessary when they start or stop smoking cigarettes. An assessment of CYP drug interactions and routine monitoring of efficacy and/or toxicity should be done to avoid potential adverse effects from medications and to determine if changes in dosages and disease state management are required.
Related Resources
- Rx for Change. Drug interactions with smoking. http://smokingcessationleadership.ucsf.edu/interactions.pdf.
- Fiore MC, Baker TB. Treating smokers in the health care setting. N Engl J Med. 2011;365(13):1222-1231.
Drug Brand Names
- Albuterol/ipratropium • Combivent
- Almotriptan • Axert
- Alosetron • Lotronex
- Aminophylline • Phyllocontin, Truphylline
- Amitriptyline • Elavil
- Amlodipine • Norvasc
- Asenapine • Saphris
- Betaxolol • Kerlone
- Carbamazepine • Carbatrol, Tegretol
- Carvedilol • Coreg
- Chlorpromazine • Thorazine
- Chlorzoxazone • Parafon Forte
- Cimetidine • Tagamet
- Ciprofloxacin • Cipro
- Clomipramine • Anafranil
- Clopidogrel • Plavix
- Clozapine • Clozaril
- Cyclobenzaprine • Flexeril
- Desipramine • Norpramin
- Diazepam • Valium
- Diclofenac • Voltaren
- Diphenhydramine • Benadryl
- Doxepin • Silenor, Sinequan
- Duloxetine • Cymbalta
- Estradiol • Estrace
- Estrogens (conjugated) • Cenestin, Premarin
- Estropipate • Ogen
- Febuxostat • Uloric
- Fluoxetine • Prozac
- Fluphenazine • Prolixin
- Fluvoxamine • Luvox
- Frovatriptan • Frova
- Guanabenz • Wytensin
- Haloperidol • Haldol
- Imipramine • Tofranil
- Maprotiline • Ludiomil
- Metoclopramide • Reglan
- Mirtazapine • Remeron
- Nabumetone • Relafen
- Naratriptan • Amerge
- Nicardipine • Cardene
- Nifedipine • Adalat, Procardia
- Nortriptyline • Aventyl, Pamelor
- Olanzapine • Zyprexa
- Omeprazole • Prilosec
- Ondansetron • Zofran
- Palonosetron • Aloxi
- Perphenazine • Trilafon
- Pimozide • Orap
- Primidone • Mysoline
- Progesterone • Prometrium
- Propofol • Diprivan
- Propranolol • Inderal
- Ramelteon • Rozerem
- Ranitidine • Zantac
- Rasagiline • Azilect
- Rifampin • Rifadin, Rimactane
- Riluzole • Rilutek
- Rivastigmine • Exelon
- Ropinirole • Requip
- Selegiline • Eldepryl, EMSAM, others
- Theophylline • Elixophyllin
- Thioridazine • Mellaril
- Thiothixene • Navane
- Tizanidine • Zanaflex
- Trazodone • Desyrel, Oleptro
- Triamterene • Dyrenium
- Trifluoperazine • Stelazine
- Verapamil • Calan, Verelan
- Warfarin • Coumadin, Jantoven
- Zileuton • Zyflo
- Ziprasidone • Geodon
- Zolmitriptan • Zomig
- Zolpidem • Ambien, Edluar
Disclosure
Ms. Fankhauser reports no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
- Tobacco smokers often are treated with medications that are metabolized by hepatic cytochrome (CYP) 1A2 enzymes. Starting or stopping tobacco smoking may cause drug interactions because polycyclic aromatic hydrocarbons in cigarette smoke induce CYP1A2 enzymes.
- Drugs that are significantly metabolized by CYP1A2 (major substrates) are more likely to be impacted by changes in tobacco smoking compared with minor substrates.
- Induction of hepatic CYP1A2 enzymes may be greater in heavy or moderate smokers compared with light smokers (eg, <10 cigarettes per day).
- Evidence-based approaches for treating tobacco use in health care settings should address the risk of CYP1A2 drug interactions in tobacco smokers and how this impacts their clinical care.
Mrs. C, age 51, experiences exacerbated asthma and difficulty breathing and is admitted to a non-smoking hospital. She also has chronic obstructive pulmonary disease, type 2 diabetes mellitus, hypertension, hypercholesterolemia, hypothyroidism, gastroesophageal reflux disease, overactive bladder, muscle spasms, fibromyalgia, bipolar disorder, insomnia, and nicotine and caffeine dependence. She takes 19 prescribed and over-the-counter medications, drinks up to 8 cups of coffee per day, and smokes 20 to 30 cigarettes per day. In the emergency room, she receives albuterol/ipratropium inhalation therapy to help her breathing and a 21-mg nicotine replacement patch to avoid nicotine withdrawal.
In the United States, 19% of adults smoke cigarettes.1 Heavy tobacco smoking and nicotine dependence are common among psychiatric patients and contribute to higher rates of tobacco-related morbidity and mortality.2 When smokers stop smoking or are admitted to smoke-free facilities and are forced to abstain, nicotine withdrawal symptoms and changes in drug metabolism can develop over several days.3-5
Smokers such as Mrs. C are at risk for cytochrome (CYP) P450 drug interactions when they are admitted to or discharged from a smoke-free facility. Nine of Mrs. C’s medications are substrates of CYP1A2 (acetaminophen, caffeine, cyclobenzaprine, diazepam, duloxetine, melatonin, olanzapine, ondansetron, and zolpidem). When Mrs. C stops smoking while in the hospital, she could experience higher serum concentrations and adverse effects of these medications. If Mrs. C resumes smoking after bring discharged, metabolism and clearance of any medications started while she was hospitalized that are substrates of CYP1A2 enzymes could be increased, which could lead to reduced efficacy and poor clinical outcomes.
Pharmacokinetic effects
Polycyclic aromatic hydrocarbons in tobacco smoke induce hepatic CYP1A1, 1A2, and possibly 2E1 isoenzymes.6-12 CYP1A2 is a hepatic enzyme responsible for metabolizing and eliminating several classes of substrates (eg, drugs, hormones, endogenous compounds, and procarcinogens).6,13 Genetic, epigenetic, and environmental factors such as smoking impact the expression and activity of CYP1A2 and result in large interpatient variability in pharmacokinetic drug interactions.6,12,13 CYP1A2 enzymes can be induced or inhibited by drugs and substances, which can result in decreased or increased serum concentrations of substrates, respectively. When individuals stop smoking and switch to other nicotine products or devices, CYP1A2 induction of hepatic enzymes will revert to normal metabolism over several weeks to a month.10 Besides tobacco smoke, other CYP1A2 inducers include charbroiled food, carbamazepine, omeprazole, phenobarbital, primidone, and rifampin.4,5 Nicotine replacement products—such as gum, inhalers, lozenges, patches, and nasal spray—and nicotine delivery devices such as electronic cigarettes do not induce hepatic CYP1A2 enzymes or cause the same drug interactions as cigarette smoking.
Table 13-11 and Table 23-11 list commonly prescribed CYP1A2 substrates that could be affected by tobacco smoke. There are no specific guidelines for how to assess, monitor, or manage pharmacokinetic drug interactions with tobacco smoke.6-13 Induction of hepatic CYP1A2 enzymes by cigarette smoke may require increased dosages of some psychotropics—such as tricyclic antidepressants, duloxetine, mirtazapine, and some first- and second-generation antipsychotics (SGAs)—to achieve serum concentrations adequate for clinical efficacy. Serum concentrations may increase to toxic levels and result in adverse effects when a person quits smoking cigarettes or if a CYP1A2 inhibitor, such as amlodipine, cimetidine, ciprofloxacin, diclofenac, fluoxetine, fluvoxamine, or nifedipine, is added.5
Table 1
Common major cytochrome P450 (CYP) 1A2 substrates
Drug | Class |
---|---|
Alosetron3,5,6 | Irritable bowel syndrome: serotonin 3 antagonist |
Aminophylline3,5 | Bronchodilator: theophylline derivative |
Betaxolol3,5 | β-1 selective adrenergic receptor blocking agent |
Caffeine3-9 | Stimulant |
Clomipramine3-11 | Tricyclic antidepressant |
Clozapine3-10 | Second-generation antipsychotic |
Cyclobenzaprine3-7 | Skeletal muscle relaxant |
Doxepin3,7,10,11 | Tricyclic antidepressant |
Duloxetine3-6 | Serotonin-norepinephrine reuptake inhibitor |
Estradiol3,5-8 | Estrogen (active) |
Estrogens: conjugated and estropipate3,5; estrone3,7 | Estrogen (derivatives) |
Fluvoxamine3,8,9 | Selective serotonin reuptake inhibitor |
Guanabenz3,5-7 | α-2 adrenergic agonist |
Mirtazapine3-7 | Antidepressant: α-2 antagonist/serotonin 2A, 2C antagonist |
Olanzapine3-11 | Second-generation antipsychotic |
Pimozide3,5,7 | First-generation antipsychotic |
Propranolol3-11 | β-adrenergic blocker |
Ramelteon3,5,10 | Melatonin receptor agonist |
Rasagiline3,5 | Antiparkinson: type B monoamine oxidase inhibitor |
Riluzole3-7,10 | Glutamate inhibitor |
Ropinirole3,5-7 | Antiparkinson: dopamine agonist |
Theophylline3-6,8-11 | Bronchodilator: methylxanthine |
Thiothixene3,5 | First-generation antipsychotic |
Trifluoperazine3,5,9 | First-generation antipsychotic |
Several classes of CYP1A2 substrates are not included and may cause toxicity with smoking cessation or require dosage increases in tobacco smokers (eg, antiarrhythmic, antifungal, antimalarial, antineoplastic, antiretroviral, and anthelmintic agents and the antibiotic quinolone). Clinicians should be most concerned about drugs with a narrow therapeutic index and those that may be toxic with smoking cessation (eg, bleeding from warfarin and clopidogrel; high serum concentrations of caffeine, clozapine, olanzapine, propranolol, and theophylline) |
Table 2
Common minor cytochrome P450 (CYP) 1A2 substrates
Drug | Class |
---|---|
Acetaminophen3-9 | Analgesic |
Almotriptan6 | Antimigraine: serotonin 1B, 1D receptor agonist |
Amitriptyline3-7,9-11 | Tricyclic antidepressant |
Asenapine9 | Second-generation antipsychotic |
Carvedilol5-7 | β and α adrenergic blocking activity |
Chlorpromazine3,4,7-9,11 | First-generation antipsychotic |
Chlorzoxazone4,7 | Skeletal muscle relaxant |
Clopidogrel5 | Antiplatelet |
Desipramine4,7,10,11 | Tricyclic antidepressant |
Diazepam4,7,9,10 | Benzodiazepine |
Diclofenac5,7 | Nonsteroidal anti-inflammatory drug |
Diphenhydramine6 | Antihistamine |
Febuxostat5 | Xanthine oxidase inhibitor |
Fluphenazine3,9 | First-generation antipsychotic |
Frovatriptan3 | Antimigraine: serotonin 1 agonist |
Haloperidol3,4,6,8,9 | First-generation antipsychotic |
Imipramine3,4,6-11 | Tricyclic antidepressant |
Maprotiline6 | Tetracyclic antidepressant |
Melatonin3,4,6,7 | Sleep-regulating hormone |
Metoclopramide3 | Antiemetic: prokinetic gastrointestinal agent |
Nabumetone6 | Nonsteroidal anti-inflammatory drug |
Naproxen3,4,6,7 | Nonsteroidal anti-inflammatory drug |
Naratriptan10 | Antimigraine: serotonin 1B, 1D receptor agonist |
Nicardipine3,7 | Calcium channel blocker |
Nortriptyline4,6,7,9-11 | Tricyclic antidepressant |
Ondansetron3,4,6,7 | Antiemetic: serotonin 3 antagonist |
Palonosetron5 | Antiemetic: serotonin 3 antagonist |
Perphenazine3,7 | First-generation antipsychotic |
Progesterone5,7 | Progestin |
Propofol4,6,7 | General anesthetic |
Ranitidine5,7 | H2 antagonist |
Rivastigmine10 | Acetylcholinesterase inhibitor |
Selegiline6,7 | Antiparkinson: type B monoamine oxidase inhibitor |
Thioridazine3,4,6 | First-generation antipsychotic |
Tizanidine3-6 | Skeletal muscle relaxant: α-2 adrenergic agonist |
Trazodone6,9 | Serotonin reuptake inhibitor and antagonist |
Triamterene6 | Diuretic: potassium sparing |
Verapamil3,4,6,7,10 | Calcium channel blocker |
Warfarin3,4,6-10 | Anticoagulant: coumarin derivative |
Zileuton3,4,6,7 | Asthma agent: 5-lipoxygenase inhibitor |
Ziprasidone3,4 | Second-generation antipsychotic |
Zolmitriptan3,6,7 | Antimigraine: serotonin 1B, 1D receptor agonist |
Zolpidem4,6,7 | Nonbenzodiazepine hypnotic |
Several classes of CYP1A2 substrates are not included and may cause toxicity with smoking cessation or require dosage increases in tobacco smokers (eg, antiarrhythmic, antifungal, antimalarial, antineoplastic, antiretroviral and anthelmintic agents and the antibiotic quinolone). Clinicians should be most concerned about drugs with a narrow therapeutic index and those that may be toxic with smoking cessation (eg, bleeding from warfarin and clopidogrel; high serum concentrations of caffeine, clozapine, olanzapine, propranolol, and theophylline) |
SGA such as clozapine and olanzapine are major substrates of CYP1A2 and dosages may need to be adjusted when smoking status changes, depending on clinical efficacy and adverse effects.10,14,15 Maximum induction of clozapine and olanzapine metabolism may occur with 7 to 12 cigarettes per day and smokers may have 40% to 50% lower serum concentrations compared with nonsmokers.14 When a patient stops smoking, clozapine and olanzapine dosages may need to be reduced by 30% to 40% (eg, a stepwise 10% reduction in daily dose until day 4) to avoid elevated serum concentrations and risk of toxicity symptoms.15
Tobacco smokers can tolerate high daily intake of caffeinated beverages because of increased metabolism and clearance of caffeine, a major substrate of CYP1A2.11 When patients stop smoking, increased caffeine serum concentrations may cause anxiety, irritability, restlessness, insomnia, tremors, palpitations, and tachycardia. Caffeine toxicity also can mimic symptoms of nicotine withdrawal; therefore, smokers should be advised to reduce their caffeine intake by half to avoid adverse effects when they stop smoking.10,11
Adjusting dosing
Factors such as the amount and frequency of tobacco smoking, how quickly CYP1A2 enzymes change when starting and stopping smoking, exposure to secondhand smoke, and other concomitant drugs contribute to variability in pharmacokinetic drug interactions. Heavy smokers (≥30 cigarettes per day) should be closely monitored because variations in drug serum concentrations may be affected significantly by changes in smoking status.4,9,11 Dosage reductions of potentially toxic drugs should be done immediately when a heavy tobacco user stops smoking.10 For CYP1A2 substrates with a narrow therapeutic range, a conservative approach is to reduce the daily dose by 10% per day for several days after smoking cessation.11,16 The impact on drug metabolism may continue for weeks to a month after the person stops smoking; therefore, there may be a delay until CYP1A2 enzymes return to normal hepatic metabolism.4,8,9,15 In most situations, smoking cessation reverses induction of hepatic CYP1A2 enzymes back to normal metabolism and causes serum drug concentrations to increase.10 Because secondhand smoke induces hepatic CYP1A2 enzymes, those exposed to smoke may have changes in drug metabolism due to environmental smoke exposure.11
Tobacco smokers who take medications and hormones that are metabolized by CYP1A2 enzymes should be closely monitored because dosage adjustments may be necessary when they start or stop smoking cigarettes. An assessment of CYP drug interactions and routine monitoring of efficacy and/or toxicity should be done to avoid potential adverse effects from medications and to determine if changes in dosages and disease state management are required.
Related Resources
- Rx for Change. Drug interactions with smoking. http://smokingcessationleadership.ucsf.edu/interactions.pdf.
- Fiore MC, Baker TB. Treating smokers in the health care setting. N Engl J Med. 2011;365(13):1222-1231.
Drug Brand Names
- Albuterol/ipratropium • Combivent
- Almotriptan • Axert
- Alosetron • Lotronex
- Aminophylline • Phyllocontin, Truphylline
- Amitriptyline • Elavil
- Amlodipine • Norvasc
- Asenapine • Saphris
- Betaxolol • Kerlone
- Carbamazepine • Carbatrol, Tegretol
- Carvedilol • Coreg
- Chlorpromazine • Thorazine
- Chlorzoxazone • Parafon Forte
- Cimetidine • Tagamet
- Ciprofloxacin • Cipro
- Clomipramine • Anafranil
- Clopidogrel • Plavix
- Clozapine • Clozaril
- Cyclobenzaprine • Flexeril
- Desipramine • Norpramin
- Diazepam • Valium
- Diclofenac • Voltaren
- Diphenhydramine • Benadryl
- Doxepin • Silenor, Sinequan
- Duloxetine • Cymbalta
- Estradiol • Estrace
- Estrogens (conjugated) • Cenestin, Premarin
- Estropipate • Ogen
- Febuxostat • Uloric
- Fluoxetine • Prozac
- Fluphenazine • Prolixin
- Fluvoxamine • Luvox
- Frovatriptan • Frova
- Guanabenz • Wytensin
- Haloperidol • Haldol
- Imipramine • Tofranil
- Maprotiline • Ludiomil
- Metoclopramide • Reglan
- Mirtazapine • Remeron
- Nabumetone • Relafen
- Naratriptan • Amerge
- Nicardipine • Cardene
- Nifedipine • Adalat, Procardia
- Nortriptyline • Aventyl, Pamelor
- Olanzapine • Zyprexa
- Omeprazole • Prilosec
- Ondansetron • Zofran
- Palonosetron • Aloxi
- Perphenazine • Trilafon
- Pimozide • Orap
- Primidone • Mysoline
- Progesterone • Prometrium
- Propofol • Diprivan
- Propranolol • Inderal
- Ramelteon • Rozerem
- Ranitidine • Zantac
- Rasagiline • Azilect
- Rifampin • Rifadin, Rimactane
- Riluzole • Rilutek
- Rivastigmine • Exelon
- Ropinirole • Requip
- Selegiline • Eldepryl, EMSAM, others
- Theophylline • Elixophyllin
- Thioridazine • Mellaril
- Thiothixene • Navane
- Tizanidine • Zanaflex
- Trazodone • Desyrel, Oleptro
- Triamterene • Dyrenium
- Trifluoperazine • Stelazine
- Verapamil • Calan, Verelan
- Warfarin • Coumadin, Jantoven
- Zileuton • Zyflo
- Ziprasidone • Geodon
- Zolmitriptan • Zomig
- Zolpidem • Ambien, Edluar
Disclosure
Ms. Fankhauser reports no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Centers for Disease Control and Prevention (CDC). Vital signs: current cigarette smoking among adults aged ≥18 years—United States 2005-2010. MMWR Morb Mortal Wkly Rep. 2011;60(35):1207-1212.
2. Ziedonis D, Hitsman B, Beckham JC, et al. Tobacco use and cessation in psychiatric disorders: National Institute of Mental Health report. Nicotine Tob Res. 2008;10(12):1691-1715.
3. Choe JY. Drug actions and interactions. New York NY: McGraw-Hill Medical; 2011.
4. Tatro DS. Drug interaction facts. St. Louis MO: Wolters Kluwer Health; 2011.
5. Lacy CF, Armstrong LL, Goldman MP, et al. eds. Drug information handbook, 20th ed. Hudson, OH: Lexicomp; 2011.
6. Zhou SF, Yang LP, Zhou ZW, et al. Insights into the substrate specificity, inhibitors, regulation, and polymorphisms and the clinical impact of human cytochrome P450 1A2. AAPS J. 2009;11(3):481-494.
7. Rendic S. Summary of information on human CYP enzymes: human P450 metabolism data. Drug Metab Rev. 2002;34(1-2):83-448.
8. Zevin S, Benowitz NL. Drug interactions with tobacco smoking. An update. Clin Pharmacokinet. 1999;36(6):425-438.
9. Desai HD, Seabolt J, Jann MW. Smoking in patients receiving psychotropic medications: a pharmacokinetic perspective. CNS Drugs. 2001;15(6):469-494.
10. Schaffer SD, Yoon S, Zadezensky I. A review of smoking cessation: potentially risky effects on prescribed medications. J Clin Nurs. 2009;18(11):1533-1540.
11. Kroon LA. Drug interactions with smoking. Am J Health Syst Pharm. 2007;64(18):1917-1921.
12. Plowchalk DR, Yeo KR. Prediction of drug clearance in a smoking population: modeling the impact of variable cigarette consumption on the induction of CYP1A2. Eur J Pharmacol. 2012;68(6):951-960.
13. Faber MS, Jetter A, Fuhr U. Assessment of CYP1A2 activity in clinical practice: why how, and when? Basic Clin Pharmacol Toxicol. 2005;97(3):125-134.
14. Haslemo T, Eikeseth PH, Tanum L, et al. The effect of variable cigarette consumption on the interaction with clozapine and olanzapine. Eur J Clin Pharmacol. 2006;62(12):1049-1053.
15. Lowe EJ, Ackman ML. Impact of tobacco smoking cessation on stable clozapine or olanzapine treatment. Ann Pharmacother. 2010;44(4):727-732.
16. Faber MS, Fuhr U. Time response of cytochrome P4501A2 activity on cessation of heavy smoking. Clin Pharmacol Ther. 2004;76(2):178-184.
1. Centers for Disease Control and Prevention (CDC). Vital signs: current cigarette smoking among adults aged ≥18 years—United States 2005-2010. MMWR Morb Mortal Wkly Rep. 2011;60(35):1207-1212.
2. Ziedonis D, Hitsman B, Beckham JC, et al. Tobacco use and cessation in psychiatric disorders: National Institute of Mental Health report. Nicotine Tob Res. 2008;10(12):1691-1715.
3. Choe JY. Drug actions and interactions. New York NY: McGraw-Hill Medical; 2011.
4. Tatro DS. Drug interaction facts. St. Louis MO: Wolters Kluwer Health; 2011.
5. Lacy CF, Armstrong LL, Goldman MP, et al. eds. Drug information handbook, 20th ed. Hudson, OH: Lexicomp; 2011.
6. Zhou SF, Yang LP, Zhou ZW, et al. Insights into the substrate specificity, inhibitors, regulation, and polymorphisms and the clinical impact of human cytochrome P450 1A2. AAPS J. 2009;11(3):481-494.
7. Rendic S. Summary of information on human CYP enzymes: human P450 metabolism data. Drug Metab Rev. 2002;34(1-2):83-448.
8. Zevin S, Benowitz NL. Drug interactions with tobacco smoking. An update. Clin Pharmacokinet. 1999;36(6):425-438.
9. Desai HD, Seabolt J, Jann MW. Smoking in patients receiving psychotropic medications: a pharmacokinetic perspective. CNS Drugs. 2001;15(6):469-494.
10. Schaffer SD, Yoon S, Zadezensky I. A review of smoking cessation: potentially risky effects on prescribed medications. J Clin Nurs. 2009;18(11):1533-1540.
11. Kroon LA. Drug interactions with smoking. Am J Health Syst Pharm. 2007;64(18):1917-1921.
12. Plowchalk DR, Yeo KR. Prediction of drug clearance in a smoking population: modeling the impact of variable cigarette consumption on the induction of CYP1A2. Eur J Pharmacol. 2012;68(6):951-960.
13. Faber MS, Jetter A, Fuhr U. Assessment of CYP1A2 activity in clinical practice: why how, and when? Basic Clin Pharmacol Toxicol. 2005;97(3):125-134.
14. Haslemo T, Eikeseth PH, Tanum L, et al. The effect of variable cigarette consumption on the interaction with clozapine and olanzapine. Eur J Clin Pharmacol. 2006;62(12):1049-1053.
15. Lowe EJ, Ackman ML. Impact of tobacco smoking cessation on stable clozapine or olanzapine treatment. Ann Pharmacother. 2010;44(4):727-732.
16. Faber MS, Fuhr U. Time response of cytochrome P4501A2 activity on cessation of heavy smoking. Clin Pharmacol Ther. 2004;76(2):178-184.
Don't Overreach for Subthreshold Pediatric Bipolar Disorder
SAN FRANCISCO – When youths get referred for help with symptoms that don’t quite meet diagnostic criteria for bipolar illness, there’s a 50-50 chance they’ll progress to a diagnosis of bipolar disorder I or II within 7 years. The odds are a coin toss.
The risk logically might be even lower in general clinical settings than in this defined group. That’s why Dr. David A. Axelson and his associates advocate using conservative criteria for diagnosing "bipolar not otherwise specified" (BP-NOS) in general clinics.
Dr. Axelson won the American Academy of Child and Adolescent Psychiatry’s Klingenstein Third Generation Foundation Award for his longitudinal research on 140 children and adolescents who met an operationalized diagnosis of BP-NOS. At a median follow-up of 5 years, 45% had converted to bipolar disorder I or II (BP I/II) within a mean of 58 weeks after intake (J. Am. Acad. Child. Adolesc. Psychiatry 2011;50:1001-16.e3).
New data from the ongoing study show that 50% progressed to BP I/II at a median follow-up of 7 years, he said at the academy’s annual meeting. Symptoms for most of the youths in the COBY (Course and Outcome of Bipolar Youth) study far exceeded the minimum BP-NOS criteria at baseline.
Very few factors predicted whether patients would convert to BP I/II or not, and even those were not strong predictors, said Dr. Axelson, medical director of Child and Adolescent Bipolar Services Outpatient Program at the University of Pittsburgh’s Western Psychiatric Institute and Clinic.
Many children and adolescents present to clinics with manic symptomatology that does not meet diagnostic criteria for BP I/II in the DSM-IV. Clinicians walk a tightrope between intervening as early as possible for best treatment results and mislabeling (and then mistreating) some youths who don’t have bipolar illness.
The criteria for BP-NOS in the DSM-IV are vague and nonspecific, Dr. Axelson said. Based on his and other studies of "subthreshold" bipolar symptoms in children and adolescents, Dr. Axelson proposed that criteria for diagnosing BP-NOS in general clinical settings include:
• Use of full DSM symptom criteria for a hypomanic or manic episode.
Almost all of the children and adolescents in his COBY study met medical criteria for symptoms, he noted.
• Having hypomanic symptoms for most of the day.
"Similar to what we think about for major depression," Dr. Axelson said. "This most-of-the-day specifier will be in DSM 5 for manic or hypomanic episodes."
• At least one episode of 2-day duration.
• At least four recurrent episodes.
Almost all the children and adolescents with BP-NOS in the COBY study already had recurrent episodes.
These criteria are "probably the best balance between sensitivity and specificity, understanding the fact that this is going to miss some kids in the early phase of illness," he said.
The COBY study enrolled 153 youths seen at three academic medical centers, 140 of whom had at least one follow-up visit. The main reasons the diagnosis was BP-NOS instead of BP I/II were because the duration of manic or hypomanic episodes was too short (only 1-3 days in 86% of patients); the youth had hypomania with no major depressive episode (11%); or the youth did not have the required number of symptoms for BP I/II (3%).
The investigators tracked at least 17 factors that they hypothesized might help predict which youths would progress to BP I/II. "Much to my surprise, very little of this actually predicted future onset," Dr. Axelson said. The main predictor was a family history of mania or hypomania and, "the effect size isn’t huge."
At intake, the 63 patients who later converted to BP I/II were significantly more likely to have a family history of mania or hypomania (64%) or depression (90%), compared with the 77 patients who did not convert to BP I/II (40% and 78%), respectively.
A total of 58% of youths with a family history of mania or hypomania converted to BP I/II by a median 5-year follow-up, compared with 36% of youths without this family history. The newest data suggest that by 8 years, two-thirds of youths with a family history of mania or hypomania convert to BP I/II, compared with just under half of youths without this family history, he reported.
"One thing that’s interesting is the progression rate keeps going up in both groups if you follow them longer," Dr. Axelson said. "These kids continue to go forward in converting to bipolar illness."
A multivariate analysis found that a family history of mania or hypomania tripled the risk for progression to BP I/II. So did white race, which "we can’t really explain," he said. Any lifetime history of psychiatric hospitalization multiplied the risk for progression 2.5 times. Higher scores on the Young Mania Rating Scale in the past month increased the risk of progression by 3%, which was statistically significant.
Any lifetime history of psychotic symptoms, however, was significantly and negatively associated with progression to BP I/II, "something we still don’t fully understand," Dr. Axelson said. Patients with a history of psychotic symptoms were 71% less likely to convert to BP I/II.
Having a family history of mania or hypomania is "a useful predictor, because more kids with family history did convert, however it’s not so strong that you can say it’s definitive," he said. "Lots of kids who had a family history didn’t progress, and a full third of the kids who didn’t have a family history progressed."
Dr. Axelson reported having no financial disclosures.
SAN FRANCISCO – When youths get referred for help with symptoms that don’t quite meet diagnostic criteria for bipolar illness, there’s a 50-50 chance they’ll progress to a diagnosis of bipolar disorder I or II within 7 years. The odds are a coin toss.
The risk logically might be even lower in general clinical settings than in this defined group. That’s why Dr. David A. Axelson and his associates advocate using conservative criteria for diagnosing "bipolar not otherwise specified" (BP-NOS) in general clinics.
Dr. Axelson won the American Academy of Child and Adolescent Psychiatry’s Klingenstein Third Generation Foundation Award for his longitudinal research on 140 children and adolescents who met an operationalized diagnosis of BP-NOS. At a median follow-up of 5 years, 45% had converted to bipolar disorder I or II (BP I/II) within a mean of 58 weeks after intake (J. Am. Acad. Child. Adolesc. Psychiatry 2011;50:1001-16.e3).
New data from the ongoing study show that 50% progressed to BP I/II at a median follow-up of 7 years, he said at the academy’s annual meeting. Symptoms for most of the youths in the COBY (Course and Outcome of Bipolar Youth) study far exceeded the minimum BP-NOS criteria at baseline.
Very few factors predicted whether patients would convert to BP I/II or not, and even those were not strong predictors, said Dr. Axelson, medical director of Child and Adolescent Bipolar Services Outpatient Program at the University of Pittsburgh’s Western Psychiatric Institute and Clinic.
Many children and adolescents present to clinics with manic symptomatology that does not meet diagnostic criteria for BP I/II in the DSM-IV. Clinicians walk a tightrope between intervening as early as possible for best treatment results and mislabeling (and then mistreating) some youths who don’t have bipolar illness.
The criteria for BP-NOS in the DSM-IV are vague and nonspecific, Dr. Axelson said. Based on his and other studies of "subthreshold" bipolar symptoms in children and adolescents, Dr. Axelson proposed that criteria for diagnosing BP-NOS in general clinical settings include:
• Use of full DSM symptom criteria for a hypomanic or manic episode.
Almost all of the children and adolescents in his COBY study met medical criteria for symptoms, he noted.
• Having hypomanic symptoms for most of the day.
"Similar to what we think about for major depression," Dr. Axelson said. "This most-of-the-day specifier will be in DSM 5 for manic or hypomanic episodes."
• At least one episode of 2-day duration.
• At least four recurrent episodes.
Almost all the children and adolescents with BP-NOS in the COBY study already had recurrent episodes.
These criteria are "probably the best balance between sensitivity and specificity, understanding the fact that this is going to miss some kids in the early phase of illness," he said.
The COBY study enrolled 153 youths seen at three academic medical centers, 140 of whom had at least one follow-up visit. The main reasons the diagnosis was BP-NOS instead of BP I/II were because the duration of manic or hypomanic episodes was too short (only 1-3 days in 86% of patients); the youth had hypomania with no major depressive episode (11%); or the youth did not have the required number of symptoms for BP I/II (3%).
The investigators tracked at least 17 factors that they hypothesized might help predict which youths would progress to BP I/II. "Much to my surprise, very little of this actually predicted future onset," Dr. Axelson said. The main predictor was a family history of mania or hypomania and, "the effect size isn’t huge."
At intake, the 63 patients who later converted to BP I/II were significantly more likely to have a family history of mania or hypomania (64%) or depression (90%), compared with the 77 patients who did not convert to BP I/II (40% and 78%), respectively.
A total of 58% of youths with a family history of mania or hypomania converted to BP I/II by a median 5-year follow-up, compared with 36% of youths without this family history. The newest data suggest that by 8 years, two-thirds of youths with a family history of mania or hypomania convert to BP I/II, compared with just under half of youths without this family history, he reported.
"One thing that’s interesting is the progression rate keeps going up in both groups if you follow them longer," Dr. Axelson said. "These kids continue to go forward in converting to bipolar illness."
A multivariate analysis found that a family history of mania or hypomania tripled the risk for progression to BP I/II. So did white race, which "we can’t really explain," he said. Any lifetime history of psychiatric hospitalization multiplied the risk for progression 2.5 times. Higher scores on the Young Mania Rating Scale in the past month increased the risk of progression by 3%, which was statistically significant.
Any lifetime history of psychotic symptoms, however, was significantly and negatively associated with progression to BP I/II, "something we still don’t fully understand," Dr. Axelson said. Patients with a history of psychotic symptoms were 71% less likely to convert to BP I/II.
Having a family history of mania or hypomania is "a useful predictor, because more kids with family history did convert, however it’s not so strong that you can say it’s definitive," he said. "Lots of kids who had a family history didn’t progress, and a full third of the kids who didn’t have a family history progressed."
Dr. Axelson reported having no financial disclosures.
SAN FRANCISCO – When youths get referred for help with symptoms that don’t quite meet diagnostic criteria for bipolar illness, there’s a 50-50 chance they’ll progress to a diagnosis of bipolar disorder I or II within 7 years. The odds are a coin toss.
The risk logically might be even lower in general clinical settings than in this defined group. That’s why Dr. David A. Axelson and his associates advocate using conservative criteria for diagnosing "bipolar not otherwise specified" (BP-NOS) in general clinics.
Dr. Axelson won the American Academy of Child and Adolescent Psychiatry’s Klingenstein Third Generation Foundation Award for his longitudinal research on 140 children and adolescents who met an operationalized diagnosis of BP-NOS. At a median follow-up of 5 years, 45% had converted to bipolar disorder I or II (BP I/II) within a mean of 58 weeks after intake (J. Am. Acad. Child. Adolesc. Psychiatry 2011;50:1001-16.e3).
New data from the ongoing study show that 50% progressed to BP I/II at a median follow-up of 7 years, he said at the academy’s annual meeting. Symptoms for most of the youths in the COBY (Course and Outcome of Bipolar Youth) study far exceeded the minimum BP-NOS criteria at baseline.
Very few factors predicted whether patients would convert to BP I/II or not, and even those were not strong predictors, said Dr. Axelson, medical director of Child and Adolescent Bipolar Services Outpatient Program at the University of Pittsburgh’s Western Psychiatric Institute and Clinic.
Many children and adolescents present to clinics with manic symptomatology that does not meet diagnostic criteria for BP I/II in the DSM-IV. Clinicians walk a tightrope between intervening as early as possible for best treatment results and mislabeling (and then mistreating) some youths who don’t have bipolar illness.
The criteria for BP-NOS in the DSM-IV are vague and nonspecific, Dr. Axelson said. Based on his and other studies of "subthreshold" bipolar symptoms in children and adolescents, Dr. Axelson proposed that criteria for diagnosing BP-NOS in general clinical settings include:
• Use of full DSM symptom criteria for a hypomanic or manic episode.
Almost all of the children and adolescents in his COBY study met medical criteria for symptoms, he noted.
• Having hypomanic symptoms for most of the day.
"Similar to what we think about for major depression," Dr. Axelson said. "This most-of-the-day specifier will be in DSM 5 for manic or hypomanic episodes."
• At least one episode of 2-day duration.
• At least four recurrent episodes.
Almost all the children and adolescents with BP-NOS in the COBY study already had recurrent episodes.
These criteria are "probably the best balance between sensitivity and specificity, understanding the fact that this is going to miss some kids in the early phase of illness," he said.
The COBY study enrolled 153 youths seen at three academic medical centers, 140 of whom had at least one follow-up visit. The main reasons the diagnosis was BP-NOS instead of BP I/II were because the duration of manic or hypomanic episodes was too short (only 1-3 days in 86% of patients); the youth had hypomania with no major depressive episode (11%); or the youth did not have the required number of symptoms for BP I/II (3%).
The investigators tracked at least 17 factors that they hypothesized might help predict which youths would progress to BP I/II. "Much to my surprise, very little of this actually predicted future onset," Dr. Axelson said. The main predictor was a family history of mania or hypomania and, "the effect size isn’t huge."
At intake, the 63 patients who later converted to BP I/II were significantly more likely to have a family history of mania or hypomania (64%) or depression (90%), compared with the 77 patients who did not convert to BP I/II (40% and 78%), respectively.
A total of 58% of youths with a family history of mania or hypomania converted to BP I/II by a median 5-year follow-up, compared with 36% of youths without this family history. The newest data suggest that by 8 years, two-thirds of youths with a family history of mania or hypomania convert to BP I/II, compared with just under half of youths without this family history, he reported.
"One thing that’s interesting is the progression rate keeps going up in both groups if you follow them longer," Dr. Axelson said. "These kids continue to go forward in converting to bipolar illness."
A multivariate analysis found that a family history of mania or hypomania tripled the risk for progression to BP I/II. So did white race, which "we can’t really explain," he said. Any lifetime history of psychiatric hospitalization multiplied the risk for progression 2.5 times. Higher scores on the Young Mania Rating Scale in the past month increased the risk of progression by 3%, which was statistically significant.
Any lifetime history of psychotic symptoms, however, was significantly and negatively associated with progression to BP I/II, "something we still don’t fully understand," Dr. Axelson said. Patients with a history of psychotic symptoms were 71% less likely to convert to BP I/II.
Having a family history of mania or hypomania is "a useful predictor, because more kids with family history did convert, however it’s not so strong that you can say it’s definitive," he said. "Lots of kids who had a family history didn’t progress, and a full third of the kids who didn’t have a family history progressed."
Dr. Axelson reported having no financial disclosures.
AT THE ANNUAL MEETING OF THE AMERICAN ACADEMY OF CHILD AND ADOLESCENT PSYCHIATRY
Major Finding: Half of youths referred for BP-NOS progressed to a diagnosis of bipolar disorder I or II within a mean of 7 years.
Data Source: Ongoing longitudinal study of 140 children and adolescents referred for symptoms that don’t quite meet diagnostic criteria for BP I/II.
Disclosures: Dr. Axelson reported having no financial disclosures.
Something smells different
CASE: Depressed and hopeless
Ms. D, age 69, has a 20-year history of bipolar II disorder, for which she is taking citalopram, 30 mg/d. She presents to her outpatient psychotherapist with a chief complaint of depressed mood. The therapist refers her for psychiatric hospitalization and electroconvulsive therapy consultation. Upon admission, Ms. D reports that her depressed mood has worsened over the past 5 weeks after a trip to the Dominican Republic. Ms. D had a negative encounter with airport security that she attributed to her 2 artificial knees and caused her to miss her flight. She endorses poor appetite, loss of energy, anhedonia, difficulty concentrating, poor memory, and feelings of hopelessness.
Ms. D reports increasingly frequent panic attacks as well as intermittent right-sided discomfort, unusual noxious smells, and increased falls. She says the falls likely are a result of new bilateral lower extremity weakness coupled with long-standing imbalance. Ms. D says she has experienced brief occasions of foul-smelling odors while showering without evidence of an offending substance. She also reports a mild, occipitally located headache.
Four years ago, Ms. D was hospitalized for a depressive episode without psychotic features and diagnosed with generalized anxiety disorder, for which she is taking clonazepam, 1.5 mg/d. Her last hypomanic episode was several years ago, and was characterized by increased energy with decreased need for sleep, flight of ideas, increased productivity, and impulsivity. Her medical history includes non-insulin dependent diabetes mellitus, chronic low back pain, hyperlipidemia, arthritis, and gastroesophageal reflux disease; her medications include pioglitazone, 30 mg/d, oxybutynin, 15 mg/d, rosuvastatin, 20 mg/d, losartan, 50 mg/d, and omeprazole, 20 mg/d. She also had bilateral knee replacements 9 years ago and an L4-S1 spinal fusion 11 years ago. She has no history of head injuries or seizures. Ms. D’s father had major depressive disorder, her mother died of a cerebrovascular accident at an unknown age, and her brother died of a myocardial infarction at age 52.
The authors’ observations
A striking aspect of Ms. D’s presenting complaints was her intermittent experience of foul smells. Although olfactory hallucinations can occur with psychotic and affective states, they also may be harbingers of an organic etiology involving the temporal lobe.1 Olfactory hallucinations associated with a psychiatric disorder often have an accompanying delusional belief regarding the cause of the smell.2
Olfactory hallucinations have been associated with migraines, epilepsy, and Parkinson’s disease.1-3 Neoplasms, cerebrovascular events, or traumatic brain injuries that result in focal mesial temporal lobe lesions can present as a partial complex seizure with olfactory or gustatory hallucinations and progress to automatisms.4 Characteristic odors in these hallucinations are unpleasant; patients with temporal lobe epilepsy describe the smells as “bad,” “rotten,” “sickening,” and “like burning food.”2 Ms. D’s report of unusual smells warranted consideration of an organic etiology for her mood change and a thorough neurologic examination.
EVALUATION: Neurologic signs
At the time of admission, Ms. D has a blood pressure of 127/68 mm Hg, heart rate of 74 beats per minute, respiratory rate of 16 breaths per minute, and temperature of 36.5°C. Neurologic examination reveals a left facial droop of unknown duration. Motor strength is weak throughout with left-sided focal weakness. Ms. D’s daughter notes that her mother’s smile appears “funny” in her admission photograph but is unsure when the asymmetry in her facial appearance began. Ms. D had been ambulatory before admission. Nursing staff observes Ms. D leans toward her left side and exhibits possible left-sided neglect during the first 12 hours of hospitalization.
When asked about her facial droop, Ms. D replies that she had not noticed any change in her appearance lately. She does not appear to be concerned about her worsening ambulation. On hospital day 2, Ms. D seems to have difficulty using utensils to eat breakfast. Ms. D is dismissive of her worsening motor function and asks to be left alone to finish her meal.
The authors’ observations
Ms. D’s focal neurologic deficits and complaint of a headache on admission were concerning because they could be caused by a cerebrovascular event or space-occupying brain lesion with potential for increased intracranial pressure. Neurologic examination with evaluation for papilledema is indicated, followed by medical transport to the closest medical center for emergent brain imaging. Neither Ms. D nor her daughter could pinpoint the onset of Ms. D’s left-sided facial droop, which precluded administering tissue plasminogen activator for a potential acute ischemic stroke.5
Ms. D’s case prompted us to consider what constitutes timely brain imaging in a patient who presents with psychiatric symptoms. Several neurologic conditions may present first with neurobehavioral symptoms before findings on physical exam. Two series of autopsies conducted >70 years ago at psychiatric hospitals found incidences of brain tumors of 3.45%6 and 13.5%.7 In a 5-year retrospective study, 21% of meningioma cases presented with psychiatric symptoms alone.8 These historical cases suggest that affective, behavioral, and psychotic symptoms may be the only clinical indicators of brain lesions that merit surgery.9-11
Imaging and radiation exposure
With the advent of CT scans in the 1970s, psychiatrists gained a new method of investigating potential structural CNS pathology in patients presenting with psychiatric symptoms. The dramatic increase in CT scan use in recent years and resulting radiation exposure is responsible for 1.5% to 2% of all cancers in the United States.12,13 Certainly, physicians must balance the advantage of early detection of brain lesions with cost-effectiveness and exposure to radiation.14
There is no consensus regarding use of brain imaging in a patient who presents with new-onset psychiatric symptoms. Certainly, patients with localizing neurologic deficits or symptoms of increased intracranial pressure should undergo brain imaging. As for psychiatric patients without neurologic findings, Filley and Kleinschmidt-DeMasters15 provide recommendations based on their 1995 case series, and other authors have recommended imaging for patients age ≥4016 vs ≥5017,18 who present with atypical mental status changes.
OUTCOME: Scan, then surgery
Ms. D’s head CT reveals a large right-sided temporoparietal low-density lesion with 8-mm left lower midline shift (Figure). She undergoes a right temporal craniotomy with resection of the mass, which is confirmed by surgical pathology to be a glioblastoma multiforme World Health Organization grade 4 tumor. Postoperative MRI shows evidence of infarction in the right posterior cerebral artery distribution and residual tumor is identified on follow-up imaging. Ms. D is referred to radiation oncology, where she receives a prognostic median life expectancy of 14 months with radiation and temozolomide treatment.19
Figure: Ms. D’s MRI results
MRI with contrast shows a large right temporal heterogeneous mass consistent with glioblastoma multiforme
The authors’ observations
Glioblastoma is a rare cancer that comprises 25% of all malignant nervous system tumors.20 It is associated with a poor prognosis, with a <30% relative survival rate for adults at 1 year and 3% at 5 years.20 Headaches, seizures, motor weakness, and progressive neurologic deficits are common symptoms of glioblastoma at diagnosis.20 Ms. D was offered the standard of care treatment for a high-grade glioma, including surgical resection followed by concomitant external-beam radiotherapy and chemotherapy.21
Consider structural brain lesions in patients who present with neurobehavioral symptoms, although most of these patients will be diagnosed with a primary psychiatric disorder. Ms. D had a known psychiatric disorder that predated the onset of neurologic symptoms and diagnosis of a rare brain cancer. Before she developed neurologic signs, Ms. D experienced symptoms uncharacteristic of her previous depressive episodes, including olfactory hallucinations, that provided an early indicator of a CNS lesion. Consider brain imaging in patients of any age who do not respond to medications targeting the presumed psychiatric diagnosis to ensure that insidious brain tumors are not missed (Table 1).15
Table 1
When to order neuroimaging for psychiatric patients
Patient’s age | Most common types of brain tumor | MRI vs CT | Indications to image |
---|---|---|---|
≥40 years | Metastases High-grade gliomas Meningiomas | Roughly equivalent for imaging common tumor types. Base on cost, availability, and relative patient contraindications | New-onset cognitive or emotional dysfunction. Patient is not responding to appropriate pharmacotherapy for psychiatric diagnosis |
<40 years | Low-grade astrocytomas Oligodendrogliomas | MRI preferred | New-onset cognitive or emotional dysfunction with associated somatic symptoms (headache, nausea, vomiting, papilledema, seizures, or focal deficits). Patient is not responding to appropriate pharmacotherapy for the psychiatric diagnosis |
Source: Reference 15 |
Compared with cerebrovascular lesions, neoplasms are more difficult to clinically correlate with their anatomic location. Neurobehavioral symptoms are more frequently associated with tumors originating in the frontal lobe or temporolimbic regions of the brain. The 3 types of frontal lobe syndromes are dorsolateral, orbitofrontal, and medial-frontal (Table 2).15 Temporolimbic tumors may present with hallucinations, mania, panic attacks, or amnesia. A meta-analysis found a statistically significant association between anorexia and hypothalamic tumors.22 Reports of neuropsychiatric symptoms that respond to pharmacologic treatment further confound the clinical picture.16
Table 2
Frontal lobe syndromes
Syndrome | Characteristics |
---|---|
Dorsolateral | Deficits in executive functioning, including organization and behavior planning |
Orbitofrontal | Prominent disinhibition |
Medial-frontal | Apathy, abulia |
Source: Reference 15 |
It is uncommon for a patient with a long-standing mood disorder to develop a primary brain cancer. However, Ms. D’s case serves as an important reminder to consider medical comorbidities in our aging psychiatric population. In particular, a patient who develops unusual symptoms or does not respond to previously effective treatments should be more closely examined and the differential diagnosis broadened.
Related Resources
- MD Anderson Cancer Center. Brain tumor videos and podcasts. www.mdanderson.org/patient-and-cancer-information/cancer-information/cancer-types/brain-tumor/videos-and-podcasts/index.html.
- Braun CM, Dumont M, Duval J, et al. Brain modules of hallucination: an analysis of multiple patients with brain lesions. J Psychiatry Neurosci. 2003;28(6):432-449.
Drug Brand Names
- Citalopram • Celexa
- Clonazepam • Klonopin
- Losartan • Cozaar
- Omeprazole • Prilosec
- Oxybutynin • Ditropan
- Pioglitazone • Actos
- Rosuvastatin • Crestor
- Temozolomide • Temodar
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Assad G, Shapiro B. Hallucinations: theoretical and clinical overview. Am J Psychiatry. 1986;143(9):1088-1097.
2. Carter JL. Visual somatosensory, olfactory, and gustatory hallucinations. Psychiatr Clin North Am. 1992;15(2):347-358.
3. Fuller GN, Guiloff RJ. Migrainous olfactory hallucinations. J Neurol Neurosurg Psychiatry. 1987;50(12):1688-1690.
4. Chang BS, Lowenstein DH. Mechanisms of disease: epilepsy. N Engl J Med. 2003;349(13):1257-1266.
5. Lansberg MG, Bluhmki E, Thijs VN. Efficacy and safety of tissue plasminogen activator 3 to 4.5 hours after acute ischemic stroke: a metaanalysis. Stroke. 2009;40(7):2438-2441.
6. Hoffman JL. Intracranial neoplasms: their incidence and mental manifestations. Psychiatr Q. 1937;11(4):561-575.
7. Larson CP. Intracranial tumors in mental hospital patients. Am J Psychiatry. 1940;97(1):49-58.
8. Gupta RK, Kumar R. Benign brain tumours and psychiatric morbidity: a 5-years retrospective data analysis. Aust N Z J Psychiatry. 2004;38(5):316-319.
9. Chambers WR. Neurosurgical conditions masquerading as psychiatric diseases. Am J Psychiatry. 1955;112(5):387-389.
10. Trimble MR, Mendez MF, Cummings JL. Neuropsychiatric symptoms from the temporolimbic lobes. J Neuropsychiatry Clin Neurosci. 1997;9(3):429-438.
11. Uribe VM. Psychiatric symptoms and brain tumor. Am Fam Physician. 1986;34(2):95-98.
12. Brenner DJ, Hall EJ. Computed tomography—an increasing source of radiation exposure. N Engl J Med. 2007;357(2):2277-2284.
13. Berrington de González A, Mahesh M, Kim KP, et al. Projected cancer risks from computed tomographic scans performed in the United States in 2007. Arch Intern Med. 2009;169(22):2071-2077.
14. Weinberger DR. Brain disease and psychiatric illness: when should a psychiatrist order a CAT scan? Am J Psychiatry. 1984;141(12):1521-1526.
15. Filley CM, Kleinschmidt-DeMasters BK. Neurobehavioral presentations of brain neoplasms. West J Med. 1995;163(1):19-25.
16. Moise D, Madhusoodanan S. Psychiatric symptoms associated with brain tumors: a clinical engima. CNS Spectr. 2006;11(1):28-31.
17. Bunevicius A, Deltuva VP, Deltuviene D, et al. Brain lesions manifesting as psychiatric disorders: eight cases. CNS Spectr. 2008;13(11):950-958.
18. Hollister LE, Boutros N. Clinical use of CT and MR scans in psychiatric patients. J Psychiatr Neurosci. 1991;16(4):194-198.
19. Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987-996.
20. Brandes AA, Tosoni A, Franceschi E, et al. Glioblastoma in adults. Crit Rev Oncol Hematol. 2008;67(2):139-152.
21. Chandana SR, Movva S, Arora M, et al. Primary brain tumors in adults. Am Fam Physician. 2008;77(10):1423-1430.
22. Madhusoodanan S, Opler MG, Moise D, et al. Brain tumor location and psychiatric symptoms: is there any association? A meta-analysis of published case studies. Expert Rev Neurother. 2010;10(10):1529-1536.
CASE: Depressed and hopeless
Ms. D, age 69, has a 20-year history of bipolar II disorder, for which she is taking citalopram, 30 mg/d. She presents to her outpatient psychotherapist with a chief complaint of depressed mood. The therapist refers her for psychiatric hospitalization and electroconvulsive therapy consultation. Upon admission, Ms. D reports that her depressed mood has worsened over the past 5 weeks after a trip to the Dominican Republic. Ms. D had a negative encounter with airport security that she attributed to her 2 artificial knees and caused her to miss her flight. She endorses poor appetite, loss of energy, anhedonia, difficulty concentrating, poor memory, and feelings of hopelessness.
Ms. D reports increasingly frequent panic attacks as well as intermittent right-sided discomfort, unusual noxious smells, and increased falls. She says the falls likely are a result of new bilateral lower extremity weakness coupled with long-standing imbalance. Ms. D says she has experienced brief occasions of foul-smelling odors while showering without evidence of an offending substance. She also reports a mild, occipitally located headache.
Four years ago, Ms. D was hospitalized for a depressive episode without psychotic features and diagnosed with generalized anxiety disorder, for which she is taking clonazepam, 1.5 mg/d. Her last hypomanic episode was several years ago, and was characterized by increased energy with decreased need for sleep, flight of ideas, increased productivity, and impulsivity. Her medical history includes non-insulin dependent diabetes mellitus, chronic low back pain, hyperlipidemia, arthritis, and gastroesophageal reflux disease; her medications include pioglitazone, 30 mg/d, oxybutynin, 15 mg/d, rosuvastatin, 20 mg/d, losartan, 50 mg/d, and omeprazole, 20 mg/d. She also had bilateral knee replacements 9 years ago and an L4-S1 spinal fusion 11 years ago. She has no history of head injuries or seizures. Ms. D’s father had major depressive disorder, her mother died of a cerebrovascular accident at an unknown age, and her brother died of a myocardial infarction at age 52.
The authors’ observations
A striking aspect of Ms. D’s presenting complaints was her intermittent experience of foul smells. Although olfactory hallucinations can occur with psychotic and affective states, they also may be harbingers of an organic etiology involving the temporal lobe.1 Olfactory hallucinations associated with a psychiatric disorder often have an accompanying delusional belief regarding the cause of the smell.2
Olfactory hallucinations have been associated with migraines, epilepsy, and Parkinson’s disease.1-3 Neoplasms, cerebrovascular events, or traumatic brain injuries that result in focal mesial temporal lobe lesions can present as a partial complex seizure with olfactory or gustatory hallucinations and progress to automatisms.4 Characteristic odors in these hallucinations are unpleasant; patients with temporal lobe epilepsy describe the smells as “bad,” “rotten,” “sickening,” and “like burning food.”2 Ms. D’s report of unusual smells warranted consideration of an organic etiology for her mood change and a thorough neurologic examination.
EVALUATION: Neurologic signs
At the time of admission, Ms. D has a blood pressure of 127/68 mm Hg, heart rate of 74 beats per minute, respiratory rate of 16 breaths per minute, and temperature of 36.5°C. Neurologic examination reveals a left facial droop of unknown duration. Motor strength is weak throughout with left-sided focal weakness. Ms. D’s daughter notes that her mother’s smile appears “funny” in her admission photograph but is unsure when the asymmetry in her facial appearance began. Ms. D had been ambulatory before admission. Nursing staff observes Ms. D leans toward her left side and exhibits possible left-sided neglect during the first 12 hours of hospitalization.
When asked about her facial droop, Ms. D replies that she had not noticed any change in her appearance lately. She does not appear to be concerned about her worsening ambulation. On hospital day 2, Ms. D seems to have difficulty using utensils to eat breakfast. Ms. D is dismissive of her worsening motor function and asks to be left alone to finish her meal.
The authors’ observations
Ms. D’s focal neurologic deficits and complaint of a headache on admission were concerning because they could be caused by a cerebrovascular event or space-occupying brain lesion with potential for increased intracranial pressure. Neurologic examination with evaluation for papilledema is indicated, followed by medical transport to the closest medical center for emergent brain imaging. Neither Ms. D nor her daughter could pinpoint the onset of Ms. D’s left-sided facial droop, which precluded administering tissue plasminogen activator for a potential acute ischemic stroke.5
Ms. D’s case prompted us to consider what constitutes timely brain imaging in a patient who presents with psychiatric symptoms. Several neurologic conditions may present first with neurobehavioral symptoms before findings on physical exam. Two series of autopsies conducted >70 years ago at psychiatric hospitals found incidences of brain tumors of 3.45%6 and 13.5%.7 In a 5-year retrospective study, 21% of meningioma cases presented with psychiatric symptoms alone.8 These historical cases suggest that affective, behavioral, and psychotic symptoms may be the only clinical indicators of brain lesions that merit surgery.9-11
Imaging and radiation exposure
With the advent of CT scans in the 1970s, psychiatrists gained a new method of investigating potential structural CNS pathology in patients presenting with psychiatric symptoms. The dramatic increase in CT scan use in recent years and resulting radiation exposure is responsible for 1.5% to 2% of all cancers in the United States.12,13 Certainly, physicians must balance the advantage of early detection of brain lesions with cost-effectiveness and exposure to radiation.14
There is no consensus regarding use of brain imaging in a patient who presents with new-onset psychiatric symptoms. Certainly, patients with localizing neurologic deficits or symptoms of increased intracranial pressure should undergo brain imaging. As for psychiatric patients without neurologic findings, Filley and Kleinschmidt-DeMasters15 provide recommendations based on their 1995 case series, and other authors have recommended imaging for patients age ≥4016 vs ≥5017,18 who present with atypical mental status changes.
OUTCOME: Scan, then surgery
Ms. D’s head CT reveals a large right-sided temporoparietal low-density lesion with 8-mm left lower midline shift (Figure). She undergoes a right temporal craniotomy with resection of the mass, which is confirmed by surgical pathology to be a glioblastoma multiforme World Health Organization grade 4 tumor. Postoperative MRI shows evidence of infarction in the right posterior cerebral artery distribution and residual tumor is identified on follow-up imaging. Ms. D is referred to radiation oncology, where she receives a prognostic median life expectancy of 14 months with radiation and temozolomide treatment.19
Figure: Ms. D’s MRI results
MRI with contrast shows a large right temporal heterogeneous mass consistent with glioblastoma multiforme
The authors’ observations
Glioblastoma is a rare cancer that comprises 25% of all malignant nervous system tumors.20 It is associated with a poor prognosis, with a <30% relative survival rate for adults at 1 year and 3% at 5 years.20 Headaches, seizures, motor weakness, and progressive neurologic deficits are common symptoms of glioblastoma at diagnosis.20 Ms. D was offered the standard of care treatment for a high-grade glioma, including surgical resection followed by concomitant external-beam radiotherapy and chemotherapy.21
Consider structural brain lesions in patients who present with neurobehavioral symptoms, although most of these patients will be diagnosed with a primary psychiatric disorder. Ms. D had a known psychiatric disorder that predated the onset of neurologic symptoms and diagnosis of a rare brain cancer. Before she developed neurologic signs, Ms. D experienced symptoms uncharacteristic of her previous depressive episodes, including olfactory hallucinations, that provided an early indicator of a CNS lesion. Consider brain imaging in patients of any age who do not respond to medications targeting the presumed psychiatric diagnosis to ensure that insidious brain tumors are not missed (Table 1).15
Table 1
When to order neuroimaging for psychiatric patients
Patient’s age | Most common types of brain tumor | MRI vs CT | Indications to image |
---|---|---|---|
≥40 years | Metastases High-grade gliomas Meningiomas | Roughly equivalent for imaging common tumor types. Base on cost, availability, and relative patient contraindications | New-onset cognitive or emotional dysfunction. Patient is not responding to appropriate pharmacotherapy for psychiatric diagnosis |
<40 years | Low-grade astrocytomas Oligodendrogliomas | MRI preferred | New-onset cognitive or emotional dysfunction with associated somatic symptoms (headache, nausea, vomiting, papilledema, seizures, or focal deficits). Patient is not responding to appropriate pharmacotherapy for the psychiatric diagnosis |
Source: Reference 15 |
Compared with cerebrovascular lesions, neoplasms are more difficult to clinically correlate with their anatomic location. Neurobehavioral symptoms are more frequently associated with tumors originating in the frontal lobe or temporolimbic regions of the brain. The 3 types of frontal lobe syndromes are dorsolateral, orbitofrontal, and medial-frontal (Table 2).15 Temporolimbic tumors may present with hallucinations, mania, panic attacks, or amnesia. A meta-analysis found a statistically significant association between anorexia and hypothalamic tumors.22 Reports of neuropsychiatric symptoms that respond to pharmacologic treatment further confound the clinical picture.16
Table 2
Frontal lobe syndromes
Syndrome | Characteristics |
---|---|
Dorsolateral | Deficits in executive functioning, including organization and behavior planning |
Orbitofrontal | Prominent disinhibition |
Medial-frontal | Apathy, abulia |
Source: Reference 15 |
It is uncommon for a patient with a long-standing mood disorder to develop a primary brain cancer. However, Ms. D’s case serves as an important reminder to consider medical comorbidities in our aging psychiatric population. In particular, a patient who develops unusual symptoms or does not respond to previously effective treatments should be more closely examined and the differential diagnosis broadened.
Related Resources
- MD Anderson Cancer Center. Brain tumor videos and podcasts. www.mdanderson.org/patient-and-cancer-information/cancer-information/cancer-types/brain-tumor/videos-and-podcasts/index.html.
- Braun CM, Dumont M, Duval J, et al. Brain modules of hallucination: an analysis of multiple patients with brain lesions. J Psychiatry Neurosci. 2003;28(6):432-449.
Drug Brand Names
- Citalopram • Celexa
- Clonazepam • Klonopin
- Losartan • Cozaar
- Omeprazole • Prilosec
- Oxybutynin • Ditropan
- Pioglitazone • Actos
- Rosuvastatin • Crestor
- Temozolomide • Temodar
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
CASE: Depressed and hopeless
Ms. D, age 69, has a 20-year history of bipolar II disorder, for which she is taking citalopram, 30 mg/d. She presents to her outpatient psychotherapist with a chief complaint of depressed mood. The therapist refers her for psychiatric hospitalization and electroconvulsive therapy consultation. Upon admission, Ms. D reports that her depressed mood has worsened over the past 5 weeks after a trip to the Dominican Republic. Ms. D had a negative encounter with airport security that she attributed to her 2 artificial knees and caused her to miss her flight. She endorses poor appetite, loss of energy, anhedonia, difficulty concentrating, poor memory, and feelings of hopelessness.
Ms. D reports increasingly frequent panic attacks as well as intermittent right-sided discomfort, unusual noxious smells, and increased falls. She says the falls likely are a result of new bilateral lower extremity weakness coupled with long-standing imbalance. Ms. D says she has experienced brief occasions of foul-smelling odors while showering without evidence of an offending substance. She also reports a mild, occipitally located headache.
Four years ago, Ms. D was hospitalized for a depressive episode without psychotic features and diagnosed with generalized anxiety disorder, for which she is taking clonazepam, 1.5 mg/d. Her last hypomanic episode was several years ago, and was characterized by increased energy with decreased need for sleep, flight of ideas, increased productivity, and impulsivity. Her medical history includes non-insulin dependent diabetes mellitus, chronic low back pain, hyperlipidemia, arthritis, and gastroesophageal reflux disease; her medications include pioglitazone, 30 mg/d, oxybutynin, 15 mg/d, rosuvastatin, 20 mg/d, losartan, 50 mg/d, and omeprazole, 20 mg/d. She also had bilateral knee replacements 9 years ago and an L4-S1 spinal fusion 11 years ago. She has no history of head injuries or seizures. Ms. D’s father had major depressive disorder, her mother died of a cerebrovascular accident at an unknown age, and her brother died of a myocardial infarction at age 52.
The authors’ observations
A striking aspect of Ms. D’s presenting complaints was her intermittent experience of foul smells. Although olfactory hallucinations can occur with psychotic and affective states, they also may be harbingers of an organic etiology involving the temporal lobe.1 Olfactory hallucinations associated with a psychiatric disorder often have an accompanying delusional belief regarding the cause of the smell.2
Olfactory hallucinations have been associated with migraines, epilepsy, and Parkinson’s disease.1-3 Neoplasms, cerebrovascular events, or traumatic brain injuries that result in focal mesial temporal lobe lesions can present as a partial complex seizure with olfactory or gustatory hallucinations and progress to automatisms.4 Characteristic odors in these hallucinations are unpleasant; patients with temporal lobe epilepsy describe the smells as “bad,” “rotten,” “sickening,” and “like burning food.”2 Ms. D’s report of unusual smells warranted consideration of an organic etiology for her mood change and a thorough neurologic examination.
EVALUATION: Neurologic signs
At the time of admission, Ms. D has a blood pressure of 127/68 mm Hg, heart rate of 74 beats per minute, respiratory rate of 16 breaths per minute, and temperature of 36.5°C. Neurologic examination reveals a left facial droop of unknown duration. Motor strength is weak throughout with left-sided focal weakness. Ms. D’s daughter notes that her mother’s smile appears “funny” in her admission photograph but is unsure when the asymmetry in her facial appearance began. Ms. D had been ambulatory before admission. Nursing staff observes Ms. D leans toward her left side and exhibits possible left-sided neglect during the first 12 hours of hospitalization.
When asked about her facial droop, Ms. D replies that she had not noticed any change in her appearance lately. She does not appear to be concerned about her worsening ambulation. On hospital day 2, Ms. D seems to have difficulty using utensils to eat breakfast. Ms. D is dismissive of her worsening motor function and asks to be left alone to finish her meal.
The authors’ observations
Ms. D’s focal neurologic deficits and complaint of a headache on admission were concerning because they could be caused by a cerebrovascular event or space-occupying brain lesion with potential for increased intracranial pressure. Neurologic examination with evaluation for papilledema is indicated, followed by medical transport to the closest medical center for emergent brain imaging. Neither Ms. D nor her daughter could pinpoint the onset of Ms. D’s left-sided facial droop, which precluded administering tissue plasminogen activator for a potential acute ischemic stroke.5
Ms. D’s case prompted us to consider what constitutes timely brain imaging in a patient who presents with psychiatric symptoms. Several neurologic conditions may present first with neurobehavioral symptoms before findings on physical exam. Two series of autopsies conducted >70 years ago at psychiatric hospitals found incidences of brain tumors of 3.45%6 and 13.5%.7 In a 5-year retrospective study, 21% of meningioma cases presented with psychiatric symptoms alone.8 These historical cases suggest that affective, behavioral, and psychotic symptoms may be the only clinical indicators of brain lesions that merit surgery.9-11
Imaging and radiation exposure
With the advent of CT scans in the 1970s, psychiatrists gained a new method of investigating potential structural CNS pathology in patients presenting with psychiatric symptoms. The dramatic increase in CT scan use in recent years and resulting radiation exposure is responsible for 1.5% to 2% of all cancers in the United States.12,13 Certainly, physicians must balance the advantage of early detection of brain lesions with cost-effectiveness and exposure to radiation.14
There is no consensus regarding use of brain imaging in a patient who presents with new-onset psychiatric symptoms. Certainly, patients with localizing neurologic deficits or symptoms of increased intracranial pressure should undergo brain imaging. As for psychiatric patients without neurologic findings, Filley and Kleinschmidt-DeMasters15 provide recommendations based on their 1995 case series, and other authors have recommended imaging for patients age ≥4016 vs ≥5017,18 who present with atypical mental status changes.
OUTCOME: Scan, then surgery
Ms. D’s head CT reveals a large right-sided temporoparietal low-density lesion with 8-mm left lower midline shift (Figure). She undergoes a right temporal craniotomy with resection of the mass, which is confirmed by surgical pathology to be a glioblastoma multiforme World Health Organization grade 4 tumor. Postoperative MRI shows evidence of infarction in the right posterior cerebral artery distribution and residual tumor is identified on follow-up imaging. Ms. D is referred to radiation oncology, where she receives a prognostic median life expectancy of 14 months with radiation and temozolomide treatment.19
Figure: Ms. D’s MRI results
MRI with contrast shows a large right temporal heterogeneous mass consistent with glioblastoma multiforme
The authors’ observations
Glioblastoma is a rare cancer that comprises 25% of all malignant nervous system tumors.20 It is associated with a poor prognosis, with a <30% relative survival rate for adults at 1 year and 3% at 5 years.20 Headaches, seizures, motor weakness, and progressive neurologic deficits are common symptoms of glioblastoma at diagnosis.20 Ms. D was offered the standard of care treatment for a high-grade glioma, including surgical resection followed by concomitant external-beam radiotherapy and chemotherapy.21
Consider structural brain lesions in patients who present with neurobehavioral symptoms, although most of these patients will be diagnosed with a primary psychiatric disorder. Ms. D had a known psychiatric disorder that predated the onset of neurologic symptoms and diagnosis of a rare brain cancer. Before she developed neurologic signs, Ms. D experienced symptoms uncharacteristic of her previous depressive episodes, including olfactory hallucinations, that provided an early indicator of a CNS lesion. Consider brain imaging in patients of any age who do not respond to medications targeting the presumed psychiatric diagnosis to ensure that insidious brain tumors are not missed (Table 1).15
Table 1
When to order neuroimaging for psychiatric patients
Patient’s age | Most common types of brain tumor | MRI vs CT | Indications to image |
---|---|---|---|
≥40 years | Metastases High-grade gliomas Meningiomas | Roughly equivalent for imaging common tumor types. Base on cost, availability, and relative patient contraindications | New-onset cognitive or emotional dysfunction. Patient is not responding to appropriate pharmacotherapy for psychiatric diagnosis |
<40 years | Low-grade astrocytomas Oligodendrogliomas | MRI preferred | New-onset cognitive or emotional dysfunction with associated somatic symptoms (headache, nausea, vomiting, papilledema, seizures, or focal deficits). Patient is not responding to appropriate pharmacotherapy for the psychiatric diagnosis |
Source: Reference 15 |
Compared with cerebrovascular lesions, neoplasms are more difficult to clinically correlate with their anatomic location. Neurobehavioral symptoms are more frequently associated with tumors originating in the frontal lobe or temporolimbic regions of the brain. The 3 types of frontal lobe syndromes are dorsolateral, orbitofrontal, and medial-frontal (Table 2).15 Temporolimbic tumors may present with hallucinations, mania, panic attacks, or amnesia. A meta-analysis found a statistically significant association between anorexia and hypothalamic tumors.22 Reports of neuropsychiatric symptoms that respond to pharmacologic treatment further confound the clinical picture.16
Table 2
Frontal lobe syndromes
Syndrome | Characteristics |
---|---|
Dorsolateral | Deficits in executive functioning, including organization and behavior planning |
Orbitofrontal | Prominent disinhibition |
Medial-frontal | Apathy, abulia |
Source: Reference 15 |
It is uncommon for a patient with a long-standing mood disorder to develop a primary brain cancer. However, Ms. D’s case serves as an important reminder to consider medical comorbidities in our aging psychiatric population. In particular, a patient who develops unusual symptoms or does not respond to previously effective treatments should be more closely examined and the differential diagnosis broadened.
Related Resources
- MD Anderson Cancer Center. Brain tumor videos and podcasts. www.mdanderson.org/patient-and-cancer-information/cancer-information/cancer-types/brain-tumor/videos-and-podcasts/index.html.
- Braun CM, Dumont M, Duval J, et al. Brain modules of hallucination: an analysis of multiple patients with brain lesions. J Psychiatry Neurosci. 2003;28(6):432-449.
Drug Brand Names
- Citalopram • Celexa
- Clonazepam • Klonopin
- Losartan • Cozaar
- Omeprazole • Prilosec
- Oxybutynin • Ditropan
- Pioglitazone • Actos
- Rosuvastatin • Crestor
- Temozolomide • Temodar
Disclosure
The authors report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.
1. Assad G, Shapiro B. Hallucinations: theoretical and clinical overview. Am J Psychiatry. 1986;143(9):1088-1097.
2. Carter JL. Visual somatosensory, olfactory, and gustatory hallucinations. Psychiatr Clin North Am. 1992;15(2):347-358.
3. Fuller GN, Guiloff RJ. Migrainous olfactory hallucinations. J Neurol Neurosurg Psychiatry. 1987;50(12):1688-1690.
4. Chang BS, Lowenstein DH. Mechanisms of disease: epilepsy. N Engl J Med. 2003;349(13):1257-1266.
5. Lansberg MG, Bluhmki E, Thijs VN. Efficacy and safety of tissue plasminogen activator 3 to 4.5 hours after acute ischemic stroke: a metaanalysis. Stroke. 2009;40(7):2438-2441.
6. Hoffman JL. Intracranial neoplasms: their incidence and mental manifestations. Psychiatr Q. 1937;11(4):561-575.
7. Larson CP. Intracranial tumors in mental hospital patients. Am J Psychiatry. 1940;97(1):49-58.
8. Gupta RK, Kumar R. Benign brain tumours and psychiatric morbidity: a 5-years retrospective data analysis. Aust N Z J Psychiatry. 2004;38(5):316-319.
9. Chambers WR. Neurosurgical conditions masquerading as psychiatric diseases. Am J Psychiatry. 1955;112(5):387-389.
10. Trimble MR, Mendez MF, Cummings JL. Neuropsychiatric symptoms from the temporolimbic lobes. J Neuropsychiatry Clin Neurosci. 1997;9(3):429-438.
11. Uribe VM. Psychiatric symptoms and brain tumor. Am Fam Physician. 1986;34(2):95-98.
12. Brenner DJ, Hall EJ. Computed tomography—an increasing source of radiation exposure. N Engl J Med. 2007;357(2):2277-2284.
13. Berrington de González A, Mahesh M, Kim KP, et al. Projected cancer risks from computed tomographic scans performed in the United States in 2007. Arch Intern Med. 2009;169(22):2071-2077.
14. Weinberger DR. Brain disease and psychiatric illness: when should a psychiatrist order a CAT scan? Am J Psychiatry. 1984;141(12):1521-1526.
15. Filley CM, Kleinschmidt-DeMasters BK. Neurobehavioral presentations of brain neoplasms. West J Med. 1995;163(1):19-25.
16. Moise D, Madhusoodanan S. Psychiatric symptoms associated with brain tumors: a clinical engima. CNS Spectr. 2006;11(1):28-31.
17. Bunevicius A, Deltuva VP, Deltuviene D, et al. Brain lesions manifesting as psychiatric disorders: eight cases. CNS Spectr. 2008;13(11):950-958.
18. Hollister LE, Boutros N. Clinical use of CT and MR scans in psychiatric patients. J Psychiatr Neurosci. 1991;16(4):194-198.
19. Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987-996.
20. Brandes AA, Tosoni A, Franceschi E, et al. Glioblastoma in adults. Crit Rev Oncol Hematol. 2008;67(2):139-152.
21. Chandana SR, Movva S, Arora M, et al. Primary brain tumors in adults. Am Fam Physician. 2008;77(10):1423-1430.
22. Madhusoodanan S, Opler MG, Moise D, et al. Brain tumor location and psychiatric symptoms: is there any association? A meta-analysis of published case studies. Expert Rev Neurother. 2010;10(10):1529-1536.
1. Assad G, Shapiro B. Hallucinations: theoretical and clinical overview. Am J Psychiatry. 1986;143(9):1088-1097.
2. Carter JL. Visual somatosensory, olfactory, and gustatory hallucinations. Psychiatr Clin North Am. 1992;15(2):347-358.
3. Fuller GN, Guiloff RJ. Migrainous olfactory hallucinations. J Neurol Neurosurg Psychiatry. 1987;50(12):1688-1690.
4. Chang BS, Lowenstein DH. Mechanisms of disease: epilepsy. N Engl J Med. 2003;349(13):1257-1266.
5. Lansberg MG, Bluhmki E, Thijs VN. Efficacy and safety of tissue plasminogen activator 3 to 4.5 hours after acute ischemic stroke: a metaanalysis. Stroke. 2009;40(7):2438-2441.
6. Hoffman JL. Intracranial neoplasms: their incidence and mental manifestations. Psychiatr Q. 1937;11(4):561-575.
7. Larson CP. Intracranial tumors in mental hospital patients. Am J Psychiatry. 1940;97(1):49-58.
8. Gupta RK, Kumar R. Benign brain tumours and psychiatric morbidity: a 5-years retrospective data analysis. Aust N Z J Psychiatry. 2004;38(5):316-319.
9. Chambers WR. Neurosurgical conditions masquerading as psychiatric diseases. Am J Psychiatry. 1955;112(5):387-389.
10. Trimble MR, Mendez MF, Cummings JL. Neuropsychiatric symptoms from the temporolimbic lobes. J Neuropsychiatry Clin Neurosci. 1997;9(3):429-438.
11. Uribe VM. Psychiatric symptoms and brain tumor. Am Fam Physician. 1986;34(2):95-98.
12. Brenner DJ, Hall EJ. Computed tomography—an increasing source of radiation exposure. N Engl J Med. 2007;357(2):2277-2284.
13. Berrington de González A, Mahesh M, Kim KP, et al. Projected cancer risks from computed tomographic scans performed in the United States in 2007. Arch Intern Med. 2009;169(22):2071-2077.
14. Weinberger DR. Brain disease and psychiatric illness: when should a psychiatrist order a CAT scan? Am J Psychiatry. 1984;141(12):1521-1526.
15. Filley CM, Kleinschmidt-DeMasters BK. Neurobehavioral presentations of brain neoplasms. West J Med. 1995;163(1):19-25.
16. Moise D, Madhusoodanan S. Psychiatric symptoms associated with brain tumors: a clinical engima. CNS Spectr. 2006;11(1):28-31.
17. Bunevicius A, Deltuva VP, Deltuviene D, et al. Brain lesions manifesting as psychiatric disorders: eight cases. CNS Spectr. 2008;13(11):950-958.
18. Hollister LE, Boutros N. Clinical use of CT and MR scans in psychiatric patients. J Psychiatr Neurosci. 1991;16(4):194-198.
19. Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987-996.
20. Brandes AA, Tosoni A, Franceschi E, et al. Glioblastoma in adults. Crit Rev Oncol Hematol. 2008;67(2):139-152.
21. Chandana SR, Movva S, Arora M, et al. Primary brain tumors in adults. Am Fam Physician. 2008;77(10):1423-1430.
22. Madhusoodanan S, Opler MG, Moise D, et al. Brain tumor location and psychiatric symptoms: is there any association? A meta-analysis of published case studies. Expert Rev Neurother. 2010;10(10):1529-1536.