Welcome to Current Psychiatry, a leading source of information, online and in print, for practitioners of psychiatry and its related subspecialties, including addiction psychiatry, child and adolescent psychiatry, and geriatric psychiatry. This Web site contains evidence-based reviews of the prevention, diagnosis, and treatment of mental illness and psychological disorders; case reports; updates on psychopharmacology; news about the specialty of psychiatry; pearls for practice; and other topics of interest and use to this audience.

Theme
medstat_cp
Top Sections
Evidence-Based Reviews
Cases That Test Your Skills
cp

Dear Drupal User: You're seeing this because you're logged in to Drupal, and not redirected to MDedge.com/psychiatry. 

Main menu
CP Main Menu
Explore menu
CP Explore Menu
Proclivity ID
18803001
Unpublish
Negative Keywords
Bipolar depression
Depression
adolescent depression
adolescent major depressive disorder
adolescent schizophrenia
adolescent with major depressive disorder
animals
autism
baby
brexpiprazole
child
child bipolar
child depression
child schizophrenia
children with bipolar disorder
children with depression
children with major depressive disorder
compulsive behaviors
cure
elderly bipolar
elderly depression
elderly major depressive disorder
elderly schizophrenia
elderly with dementia
first break
first episode
gambling
gaming
geriatric depression
geriatric major depressive disorder
geriatric schizophrenia
infant
kid
major depressive disorder
major depressive disorder in adolescents
major depressive disorder in children
parenting
pediatric
pediatric bipolar
pediatric depression
pediatric major depressive disorder
pediatric schizophrenia
pregnancy
pregnant
rexulti
skin care
teen
wine
Negative Keywords Excluded Elements
header[@id='header']
section[contains(@class, 'nav-hidden')]
footer[@id='footer']
div[contains(@class, 'pane-pub-article-current-psychiatry')]
div[contains(@class, 'pane-pub-home-current-psychiatry')]
div[contains(@class, 'pane-pub-topic-current-psychiatry')]
div[contains(@class, 'panel-panel-inner')]
div[contains(@class, 'pane-node-field-article-topics')]
section[contains(@class, 'footer-nav-section-wrapper')]
Altmetric
DSM Affiliated
Display in offset block
QuickLearn Excluded Topics/Sections
Best Practices
CME
CME Supplements
Education Center
Medical Education Library
Disqus Exclude
Best Practices
CE/CME
Education Center
Medical Education Library
Enable Disqus
Display Author and Disclosure Link
Publication Type
Clinical
Slot System
Top 25
Disable Sticky Ads
Disable Ad Block Mitigation
Featured Buckets Admin
Publication LayerRX Default ID
798,799
Show Ads on this Publication's Homepage
Consolidated Pub
Show Article Page Numbers on TOC

The model psychiatrist: 7 domains of excellence

Article Type
Changed
Mon, 04/16/2018 - 14:26
Display Headline
The model psychiatrist: 7 domains of excellence

What makes a first-class psychiatrist? What are the traits that characterize the “ideal” psychiatrist? How does a good psychiatrist become great? There are many possible answers depending on who is asked.

In my view, after observing countless fellow psychiatrists over 3 decades in various settings, I have concluded there are 7 domains that determine the caliber of psychiatrists that we all aspire to be. It may be difficult to possess all the traits across all 7 domains but I propose it as an idealized model and a road map for the journey toward peak performance in our profession.

Domain I: Personal attributes

 

  • The ability to listen “actively” and observe “comprehensively”
  • Psychological mindedness and curiosity
  • Skillfully engages and develops therapeutic alliance with patients from the first encounter
  • Compassion and empathy, but pragmatic firmness about boundaries
  • Nonjudgmental stance and cognizance of one’s own limitations
  • Impeccable integrity and ethical conduct.

Domain II: Clinical mastery

 

  • Thorough familiarity with the principles of psychiatry and models of behavior and psychopathology
  • Rigor in applying a diagnostic label by employing the skills of a physician to check patients’ medical status to rule out general medical etiologies; monitors patients’ physical and mental health and refers as needed
  • Establishes a skillful biopsychosocial treatment plan requiring extensive knowledge of psychopharmacology and psychotherapy
  • Applies evidence-based interventions wherever available
  • Always checks and deals with countertransference issues.

Domain III: Professionalism and leadership

 

  • Always well dressed and groomed with a professional appearance; projects the identity of being a physician; leadership of mental health teams
  • Involved in local, state, national, and international professional societies
  • Raises the profile of psychiatry within the medical field by networking with other physician organizations and participating in medical initiatives.

Domain IV: Organizational effectiveness

 

  • Leads balanced and well organized professional and personal lives
  • Smooth working relationship with individuals or agencies relevant to the patient’s treatment, including family members, medical and mental health professionals, hospital administrators, clinics, insurance companies, advocacy groups, and the legal system
  • Maintains an organized and complete medical record with measurement-based ratings of illness severity and side effects.

Domain V: Societal role

 

  • Establishes one’s self as a role model for those seeking advice and guidance not only for psychiatric disorders, but for various societal dilemmas and is an effective communicator
  • Feels comfortable in being a public figure in one’s community whose opinions are valued by laymen as well as other professionals
  • Serves as an ambassador for the profession by educating the public via various media outlets to erase misperceptions about mental illness or psychiatry, and to rebut and neutralize the occasional venomous assaults recklessly hurled by antipsychiatry cults.

Domain VI: Lifelong learning

 

  • Convinced of the need for continuous learning in an era of logarithmic growth of medical knowledge
  • Reads several articles a week from key journals about recent clinical and scientific advances and applies them to patients when appropriate
  • Attends or presents at continuing medical education meetings and asks questions or makes comments (active, not passive attendance).

Domain VII: Contribution to new knowledge

 

  • Recognizes the vast unmet needs in psychiatry, supports research, and understands that the research of today is the better treatment of tomorrow
  • Refers patients to clinical research protocols at the local academic institution and offers to collaborate as a rater or data collector
  • Takes the time to write up unusual clinical cases as a letter to the editor or case report and generate new clinical ideas
  • If not a full-time academic, volunteers to teach or supervise medical students or residents at the local medical school or teaching hospital.

Very few psychiatrists can excel at all 7 domains, but most meet the criteria for several of them. Every psychiatrist can move from good to great with a modicum of effort and motivation. If most of us do that, the caliber and standing of our psychiatric profession will continue to escalate.

Article PDF
Author and Disclosure Information

 


Henry A. Nasrallah, MD
Editor-in-Chief
To comment on this editorial or other topics of interest, visit http://www.facebook.com/CurrentPsychiatry, or click on the “Send Letters” link below.

Issue
Current Psychiatry - 10(11)
Publications
Topics
Page Number
5-6
Legacy Keywords
model psychiatrist; personal attributes; clinical mastery; professionalism; leadership; organizational effectiveness; societal role; lifelong learning; contribution to new knowledge; November editorial; Henry Nasrallah
Sections
Author and Disclosure Information

 


Henry A. Nasrallah, MD
Editor-in-Chief
To comment on this editorial or other topics of interest, visit http://www.facebook.com/CurrentPsychiatry, or click on the “Send Letters” link below.

Author and Disclosure Information

 


Henry A. Nasrallah, MD
Editor-in-Chief
To comment on this editorial or other topics of interest, visit http://www.facebook.com/CurrentPsychiatry, or click on the “Send Letters” link below.

Article PDF
Article PDF

What makes a first-class psychiatrist? What are the traits that characterize the “ideal” psychiatrist? How does a good psychiatrist become great? There are many possible answers depending on who is asked.

In my view, after observing countless fellow psychiatrists over 3 decades in various settings, I have concluded there are 7 domains that determine the caliber of psychiatrists that we all aspire to be. It may be difficult to possess all the traits across all 7 domains but I propose it as an idealized model and a road map for the journey toward peak performance in our profession.

Domain I: Personal attributes

 

  • The ability to listen “actively” and observe “comprehensively”
  • Psychological mindedness and curiosity
  • Skillfully engages and develops therapeutic alliance with patients from the first encounter
  • Compassion and empathy, but pragmatic firmness about boundaries
  • Nonjudgmental stance and cognizance of one’s own limitations
  • Impeccable integrity and ethical conduct.

Domain II: Clinical mastery

 

  • Thorough familiarity with the principles of psychiatry and models of behavior and psychopathology
  • Rigor in applying a diagnostic label by employing the skills of a physician to check patients’ medical status to rule out general medical etiologies; monitors patients’ physical and mental health and refers as needed
  • Establishes a skillful biopsychosocial treatment plan requiring extensive knowledge of psychopharmacology and psychotherapy
  • Applies evidence-based interventions wherever available
  • Always checks and deals with countertransference issues.

Domain III: Professionalism and leadership

 

  • Always well dressed and groomed with a professional appearance; projects the identity of being a physician; leadership of mental health teams
  • Involved in local, state, national, and international professional societies
  • Raises the profile of psychiatry within the medical field by networking with other physician organizations and participating in medical initiatives.

Domain IV: Organizational effectiveness

 

  • Leads balanced and well organized professional and personal lives
  • Smooth working relationship with individuals or agencies relevant to the patient’s treatment, including family members, medical and mental health professionals, hospital administrators, clinics, insurance companies, advocacy groups, and the legal system
  • Maintains an organized and complete medical record with measurement-based ratings of illness severity and side effects.

Domain V: Societal role

 

  • Establishes one’s self as a role model for those seeking advice and guidance not only for psychiatric disorders, but for various societal dilemmas and is an effective communicator
  • Feels comfortable in being a public figure in one’s community whose opinions are valued by laymen as well as other professionals
  • Serves as an ambassador for the profession by educating the public via various media outlets to erase misperceptions about mental illness or psychiatry, and to rebut and neutralize the occasional venomous assaults recklessly hurled by antipsychiatry cults.

Domain VI: Lifelong learning

 

  • Convinced of the need for continuous learning in an era of logarithmic growth of medical knowledge
  • Reads several articles a week from key journals about recent clinical and scientific advances and applies them to patients when appropriate
  • Attends or presents at continuing medical education meetings and asks questions or makes comments (active, not passive attendance).

Domain VII: Contribution to new knowledge

 

  • Recognizes the vast unmet needs in psychiatry, supports research, and understands that the research of today is the better treatment of tomorrow
  • Refers patients to clinical research protocols at the local academic institution and offers to collaborate as a rater or data collector
  • Takes the time to write up unusual clinical cases as a letter to the editor or case report and generate new clinical ideas
  • If not a full-time academic, volunteers to teach or supervise medical students or residents at the local medical school or teaching hospital.

Very few psychiatrists can excel at all 7 domains, but most meet the criteria for several of them. Every psychiatrist can move from good to great with a modicum of effort and motivation. If most of us do that, the caliber and standing of our psychiatric profession will continue to escalate.

What makes a first-class psychiatrist? What are the traits that characterize the “ideal” psychiatrist? How does a good psychiatrist become great? There are many possible answers depending on who is asked.

In my view, after observing countless fellow psychiatrists over 3 decades in various settings, I have concluded there are 7 domains that determine the caliber of psychiatrists that we all aspire to be. It may be difficult to possess all the traits across all 7 domains but I propose it as an idealized model and a road map for the journey toward peak performance in our profession.

Domain I: Personal attributes

 

  • The ability to listen “actively” and observe “comprehensively”
  • Psychological mindedness and curiosity
  • Skillfully engages and develops therapeutic alliance with patients from the first encounter
  • Compassion and empathy, but pragmatic firmness about boundaries
  • Nonjudgmental stance and cognizance of one’s own limitations
  • Impeccable integrity and ethical conduct.

Domain II: Clinical mastery

 

  • Thorough familiarity with the principles of psychiatry and models of behavior and psychopathology
  • Rigor in applying a diagnostic label by employing the skills of a physician to check patients’ medical status to rule out general medical etiologies; monitors patients’ physical and mental health and refers as needed
  • Establishes a skillful biopsychosocial treatment plan requiring extensive knowledge of psychopharmacology and psychotherapy
  • Applies evidence-based interventions wherever available
  • Always checks and deals with countertransference issues.

Domain III: Professionalism and leadership

 

  • Always well dressed and groomed with a professional appearance; projects the identity of being a physician; leadership of mental health teams
  • Involved in local, state, national, and international professional societies
  • Raises the profile of psychiatry within the medical field by networking with other physician organizations and participating in medical initiatives.

Domain IV: Organizational effectiveness

 

  • Leads balanced and well organized professional and personal lives
  • Smooth working relationship with individuals or agencies relevant to the patient’s treatment, including family members, medical and mental health professionals, hospital administrators, clinics, insurance companies, advocacy groups, and the legal system
  • Maintains an organized and complete medical record with measurement-based ratings of illness severity and side effects.

Domain V: Societal role

 

  • Establishes one’s self as a role model for those seeking advice and guidance not only for psychiatric disorders, but for various societal dilemmas and is an effective communicator
  • Feels comfortable in being a public figure in one’s community whose opinions are valued by laymen as well as other professionals
  • Serves as an ambassador for the profession by educating the public via various media outlets to erase misperceptions about mental illness or psychiatry, and to rebut and neutralize the occasional venomous assaults recklessly hurled by antipsychiatry cults.

Domain VI: Lifelong learning

 

  • Convinced of the need for continuous learning in an era of logarithmic growth of medical knowledge
  • Reads several articles a week from key journals about recent clinical and scientific advances and applies them to patients when appropriate
  • Attends or presents at continuing medical education meetings and asks questions or makes comments (active, not passive attendance).

Domain VII: Contribution to new knowledge

 

  • Recognizes the vast unmet needs in psychiatry, supports research, and understands that the research of today is the better treatment of tomorrow
  • Refers patients to clinical research protocols at the local academic institution and offers to collaborate as a rater or data collector
  • Takes the time to write up unusual clinical cases as a letter to the editor or case report and generate new clinical ideas
  • If not a full-time academic, volunteers to teach or supervise medical students or residents at the local medical school or teaching hospital.

Very few psychiatrists can excel at all 7 domains, but most meet the criteria for several of them. Every psychiatrist can move from good to great with a modicum of effort and motivation. If most of us do that, the caliber and standing of our psychiatric profession will continue to escalate.

Issue
Current Psychiatry - 10(11)
Issue
Current Psychiatry - 10(11)
Page Number
5-6
Page Number
5-6
Publications
Publications
Topics
Article Type
Display Headline
The model psychiatrist: 7 domains of excellence
Display Headline
The model psychiatrist: 7 domains of excellence
Legacy Keywords
model psychiatrist; personal attributes; clinical mastery; professionalism; leadership; organizational effectiveness; societal role; lifelong learning; contribution to new knowledge; November editorial; Henry Nasrallah
Legacy Keywords
model psychiatrist; personal attributes; clinical mastery; professionalism; leadership; organizational effectiveness; societal role; lifelong learning; contribution to new knowledge; November editorial; Henry Nasrallah
Sections
Article PDF Media

Treatment-resistant OCD: Options beyond first-line medications

Article Type
Changed
Tue, 12/11/2018 - 15:04
Display Headline
Treatment-resistant OCD: Options beyond first-line medications

Obsessive-compulsive disorder (OCD) is marked by recurrent and persistent anxiety-provoking thoughts (obsessions) accompanied by repetitive behaviors (compulsions) that focus on alleviating distress caused by obsessive thoughts. Although patients recognize the obsessions and compulsions are unreasonable, these thoughts and behaviors remain time-consuming and impair function. Even when they appropriately identify and treat OCD, clinicians often face “treatment-resistant” (or “treatment-refractory”) patients who do not respond adequately to standard therapies (Box).1 Several factors contribute to treatment resistance, including those related to the patient, the environment, the clinician/health system, and pathology (Table 1).2 An estimated 10% to 40% of patients with OCD are treatment-resistant.2

This article discusses the range of options for addressing resistant OCD, including augmenting first-line treatments with pharmacotherapy, psychotherapy, or reversible or irreversible forms of neuromodulation.

Defining treatment resistance in obsessive-compulsive disorder

Treatment resistance generally refers to lack of sufficient improvement despite multiple adequate and appropriate treatment trials. However, there are no universally accepted definitions or metrics of treatment resistance, and often it is operationally defined. For mood disorders, it may be defined by failure to remit or respond clinically (50% reduction in symptoms) despite ≥2 adequate antidepressant trials or failure to respond clinically despite adequate medication trials across several neurotransmitter classes. The terms treatment resistant and treatment refractory are synonymous; they refer to the same phenomenon and are used interchangeably in the literature. Including the terms “remission” and “recovery” when judging treatment efficacy for anxiety disorders can be limiting because of the chronic and often unrelenting nature of these conditions.

One review proposed categorizing obsessive-compulsive disorder treatment response into several stages along a spectrum, ranging from complete recovery (or remission) to full or partial response to non-response (or completely refractory).1 However it is defined, treatment resistance in anxiety disorders likely is characterized by minimal restoration of function despite several appropriate treatment exposures.

Table 1

Factors that contribute to treatment resistance in obsessive-compulsive disorder

Patient
Disease severity
Medical comorbidity
Psychiatric comorbidity (mood, personality, and/or substance use disorders)
Treatment nonadherence
Cultural factors
Environment
Childhood stressors (trauma, abuse)
Long-term persistent stressors (psychosocial, occupational, financial)
Life stages
Clinician/health system
Lack of knowledge in primary care (brief treatment duration, subtherapeutic dosing)
Lack of psychotherapeutic training
Limited doctor-patient relationship (eg, availability/cost of treatment)
Pathology-related
Underlying disease pathophysiology (largely unknown):
  • Multiple neurotransmitter system interactions
  • Polygenetic influences (genetic load)
  • Gene-environment interactions
  • Neural circuits (cortical and subcortical feedback loops)
Diagnostic variance (dimensional vs categorical vs target symptom approach)
Syndromal variation (differing presentations over time)
Treatment limitations (limited empirical studies, nonrepresentative study samples)
Source: Reference 2

First-line pharmacotherapy

Clomipramine or a selective serotonin reuptake inhibitor (SSRI) are considered first-line treatments for OCD. Although some evidence indicates that clomipramine may have greater efficacy than SSRIs, its poor tolerability and potential lethality in overdose make it a less practical first choice in treatment-naïve patients.3,4 SSRIs generally are well tolerated and have a favorable safety profile. Nearly all SSRIs have randomized clinical trials (RCTs) and FDA indications that support their use in OCD. SSRI choice may be guided by patient or prescriber preference because no evidence suggests that 1 SSRI is superior to another for treating OCD.5 In contrast to major depressive disorder, in OCD there is a dose-response relationship for SSRI treatment; higher doses typically are required to achieve response or remission.6,7

Augmentation and other options

Patients who have not responded to at least 2 adequate trials of first-line medications may benefit from an augmentation strategy or treatment with an unconventional agent. Such cases should be managed by a specialist who has experience in treating OCD and with careful consideration of potential risks of these interventions.

Evidence suggests the following pharmacotherapies may effectively treat OCD and may be warranted for treatment-resistant patients.

Serotonergic agents

Supratherapeutic SSRI doses. Evidence suggests that supratherapeutic doses of SSRIs may be effective, which may be a logical first step when treating patients already taking an SSRI who have not responded. In a multi-center, double-blind study comparing sertraline, 200 mg/d, to sertraline, 250 to 400 mg/d, the latter group showed significantly greater symptom improvement.8 Citalopram may not be suitable for this approach because of the recent FDA announcement regarding dose-dependent QTc prolongation associated with this medication.9

Serotonin-norepinephrine reuptake inhibitors (SNRIs). In the only double-blind, placebo-controlled study of venlafaxine for OCD, the drug was not significantly more effective than placebo.10 This study was small (N = 30). There are sufficient positive results from open-label and blinded comparator studies that venlafaxine generally is accepted as an effective and well-tolerated treatment for OCD at doses ≥225 mg/d.11

 

 

Duloxetine also may be effective in treating OCD. One case series reported improvement in 3 of 4 SSRI nonresponders who were switched to this medication and rapidly titrated to 120 mg/d.12

Clomipramine/SSRI augmentation. For patients who have not responded to an SSRI, several open-label trials support adding clomipramine.13 Conversely, SSRI augmentation for patients who have not adequately responded to clomipramine may be effective.14 With any dual therapy with serotonergic agents, monitor patients for signs and symptoms of serotonin syndrome.

IV clomipramine. By bypassing first-pass metabolism, IV clomipramine rapidly achieves high plasma levels. In a double-blind, placebo-controlled study of 54 OCD patients who were nonresponsive to oral clomipramine, IV clomipramine was more effective than placebo.15 An additional study found IV clomipramine is more effective when pulse loaded than when titrated gradually.16

Pindolol. The beta blocker pindolol acts as an antagonist of presynaptic 5-HT1A autoreceptors, increasing serotonergic signaling. A small double-blind, placebo-controlled trial (N = 14) found a significant decrease in Yale-Brown Obsessive Compulsive Scale (Y-BOCS) score with pindolol augmentation, 2.5 mg, 3 times daily, among patients who did not respond to ≥3 serotonin reuptake inhibitor (SRI) trials.17 Pindolol augmentation showed modest effects in 2 open-label studies.18,19 However, another small double-blind, placebo-controlled study (N = 15) found no difference between placebo and fluvoxamine augmented with pindolol.20

Ondansetron. A 5-HT3 receptor antagonist, ondansetron is used primarily as an antiemetic but has been shown to have anxiolytic properties in animal studies. In an open-label study of 8 patients with non–treatment refractory OCD, 3 achieved clinical response (at least 35% reduction in Y-BOCS score) with ondansetron monotherapy dosed at 1 mg, 3 times daily.21 In a subsequent single-blind trial with 14 treatment-resistant patients, 9 responded (at least 25% reduction in Y-BOCS score).22

Other medications

Antipsychotics. Most studies examining antipsychotic monotherapy for OCD have been negative. One exception was a small, open-label trial of aripiprazole monotherapy (N = 8) that found modest efficacy among non–treatment refractory patients.23 Augmentation with antipsychotics, however, has been well studied and there is good evidence of efficacy for this approach. Double-blind, placebo-controlled studies have supported the efficacy of augmenting SRIs with haloperidol, risperidone, olanzapine, quetiapine, and aripiprazole.24-26 Several case reports suggest ziprasidone may be an effective SRI adjunct, but 1 retrospective study found it was inferior to quetiapine.27

Benzodiazepines. Case reports present positive effects of clonazepam and alprazolam for OCD, but double-blind, placebo-controlled trials for monotherapy or adjunctive clonazepam have been negative.28,29 Furthermore, cognitive impairment and potential for dependence associated with benzodiazepines weigh against their use in OCD.

Opioids. A double-blind, placebo controlled crossover study of 23 patients with treatment-refractory OCD found once-weekly oral morphine added to patients’ current regimen significantly reduced Y-BOCS score vs placebo. Patients received 30 mg the first week and 15 to 45 mg the next week, depending on response or side effects.30 A case report and a small open-label trial support the efficacy of tramadol, a weak agonist of the μ opioid receptor and an inhibitor of serotonin and norepinephrine transporters, as monotherapy and as an adjunct to fluoxetine.31,32 Because patients with OCD may be particularly vulnerable to dependence and intentional or accidental overdose via opioid/benzodiazepine combinations, evaluate the risks and benefits before initiating an opioid.

Psychostimulants. Sparse but good evidence supports the efficacy of dextroamphetamine monotherapy for OCD.33,34 There are no positive studies of methylphenidate and several case reports of methylphenidate-induced OCD symptoms.35

N-methyl-D-aspartate (NMDA) antagonists. Increased glutamatergic neurotransmission has been implicated in the pathophysiology of OCD, which suggests a possible role for glutamate receptor antagonists. In an open-label trial, memantine, an NMDA antagonist used primarily to treat dementia, was associated with clinical response (>25% reduction in Y-BOCS scores) in 6 of 14 patients with treatment-refractory OCD.36 Several case reports and an open-label trial support the efficacy of riluzole—which is indicated for treating amyotrophic lateral sclerosis—as an adjunct for treatment-refractory OCD.37 Although its exact mechanism of action is unclear, riluzole’s effects are thought to be mediated via reduction in glutamatergic neurotransmission. IV ketamine has reported anti-OCD effects in a case report of a woman with treatment-resistant OCD. These effects occurred almost immediately and persisted for several days.38

Hallucinogens. Psilocybin, psilocin, and lysergic acid diethylamide have reported anti-OCD properties.39 As schedule I substances, however, they are not available outside of sanctioned research protocols and may carry substantial risk. Nonetheless, their efficacy suggests that other compounds that share their mechanism of action—namely agonism of 5-HT2A and 5-HT2C receptors—may merit investigation as potential treatments for OCD.

 

 

Psychotherapy

Cognitive-behavioral therapy (CBT) has been shown to be effective for OCD as monotherapy and augmentation to pharmacotherapy. CBT consists of cognitive and behavioral components, typically involving some form of cognitive restructuring and exposure response prevention. Although these 2 types of interventions arise from independent traditions, in CBT they are frequently intertwined, particularly when the focus of OCD patients’ anxiety is ego-dystonic thoughts.

One benefit of CBT over pharmacotherapy is that effects persist after treatment is terminated. A recent prospective study found CBT was effective for treatment-refractory OCD, with 74% of patients demonstrating clinical response after 20 to 25 sessions over 2 months and 61% maintaining clinical response 1 year after treatment.40 CBT administered remotely via teleconference, also known as “teletherapy,” has shown efficacy for OCD.41

Alternative medicine

Despite widespread use of herbal remedies for OCD, no trials have shown a strong positive effect. Both Hypericum perforatum (St. John’s wort) and Silybum marianum (milk thistle) have been used to treat obsessive and compulsive symptoms; however, placebo-controlled trials did not find any significant differences in symptoms or side effects between treatment groups.42,43 Lower-quality studies have reported modest effects for mindfulness meditation, yoga, and acupuncture.44

Because many patients continue to use complementary and alternative medicine therapies despite the lack of data on efficacy, it is important to monitor for potential interactions with prescription medications. St. John’s wort interacts with many medications because of induction of the cytochrome P450 (CYP) isoenzymes 3A4 and 2C9. This interaction may lower blood levels of alprazolam and clonazepam (3A4). Combining St. John’s wort with SSRIs increases the risk of serotonin syndrome. Milk thistle inhibits CYP450 isoenzyme 3A4, and may increase serum levels of other medications metabolized by this pathway.

Invasive therapies

Invasive options may be considered after several pharmacotherapeutic and psychotherapeutic approaches have not been effective or when significant functional impairment remains (Table 2). These therapies typically are reserved for patients whose treatment resistance is strongest.

Electroconvulsive therapy (ECT). Although ECT is an effective tool for treatment-resistant mood disorders or treatment-resistant anxiety complicated by severe depression, studies have not found ECT to be effective for OCD. One uncontrolled case series reported considerable improvements in OCD patients the year after ECT, although improvement was correlated with improved depression scores.45

Vagal nerve stimulation (VNS). In an open-label study of 7 OCD patients who received VNS, 3 were acute responders—characterized by a ≥25% improvement on the Y-BOCS—and 2 received continued benefits at 4-year follow up (2 patients dropped out).46

Repetitive transcranial magnetic stimulation (rTMS). A meta-analysis of 3 RCTs of rTMS for patients with OCD did not yield a large or statistically significant effect.47 Limitations of these trials included asymmetric stimulation sites (eg, left vs right only), limited stimulation sites (dorsolateral prefrontal cortex), different stimulation frequencies between studies, and a lack of sham stimulation conditions. A more recent RCT and subsequent review described moderate efficacy (defined by ≥25% decrease in Y-BOCS scores) compared with sham stimulations in OCD patients at 4 weeks, using the supplementary motor area as a stimulation site.48,49

The main limitation of rTMS is the inability to penetrate deeper brain structures implicated in OCD (eg, caudate nucleus, thalamus, anterior capsule fiber tracts), as well as a lack of specificity in stimulation site.

Surgical approaches. Cingulotomy is the most commonly employed surgical procedure for OCD in North America, likely because of a combination of clinical efficacy and low morbidity and mortality rates.50 Of the >1,000 cingulotomies that have been performed at Massachusetts General Hospital, no deaths or postoperative infections have been reported and 2 subdural hematomas have occurred.50 Common postsurgical side effects include transient headache, nausea, or difficulty urinating. The most serious common side effect—postoperative seizures—has been reported in 1% to 9% of cases.

Outcomes for these procedures cannot be fully assessed until at least 6 months to 2 years after the procedure, which suggests postoperative neural reorganization plays an important role in recovery. Direct comparisons of each lesion approach within studies are extremely rare. Overall, long-term outcomes of these approaches have demonstrated significant therapeutic effects of each of these procedures. Reported response rates vary between 30% to 70%, when applied to remission, response (≥35% Y-BOCS reduction), and functional improvements in quality of life.50

 

 

Deep brain stimulation (DBS). With this approach, small electrodes are inserted under precise stereotactic MRI guidance. The advantage of DBS over ablative surgery is the ability to adjust and customize neurostimulation. Following implantation, modifiable parameters of electrode stimulation include electrode polarity, intensity, frequency, and laterality. A specially trained psychiatrist can conduct parameter optimization during long-term follow-up.

The first trial of DBS for OCD was reported in 1999 (N = 4), with the initial target selected based on the site of anterior capsulotomy. Three patients derived clinically observed benefit, although no validated questionnaires were administered.51 Since then, at least 7 studies with blinded stimulation have been conducted, totaling 62 patients.52

In recent years, structures adjacent to the internal capsule also have been targeted based on the approach employed in ventral capsulotomy. Across all trials, response rates for this approach consistently have been in the 50% range, with average Y-BOCS score reductions ranging from 6.8 to 31 points.53 Some patients have reported rapid improvements in anhedonia, and this approach is being employed in treatment-resistant depression.

Postoperative complications occur more often with DBS than with lesion approaches because of the prosthetic nature of the procedure (eg, increased risk of infection, lead malfunction, etc.). Additionally, batteries must be periodically explanted and replaced. Reported stimulation-related side effects include mood changes (transient sadness, anxiety, euphoria, and hypomania), sensory disturbances (olfactory, gustatory, and motor sensations), and cognitive changes (confusion and forgetfulness). These side effects typically are stimulation-dependent and disappear after altering stimulation parameters.

Table 2

Invasive therapies for treatment-resistant OCD

TherapyQuality of evidence
Reversible
  Electroconvulsive therapyPoor
  Vagal nerve stimulationPoor
  Repetitive transcranial magnetic stimulationLimited
Irreversible (surgical)
  Anterior capsulotomy. Target: anterior limb of the internal capsuleFair
  Anterior cingulotomy. Target: anterior cingulate and cingulum bundleFair
  Subcaudate tractotomy. Target: substantia innominata, just inferior to the caudate nucleusFair
  Limbic leucotomy. Target: anterior cingulotomy combined with subcaudate tractotomyFair
  Deep brain stimulation. Multiple targetsFair
OCD: obsessive-compulsive disorder

Related Resources

  • American Psychiatric Association. Treatment of patients with obsessive-compulsive disorder. www.psychiatryonline.com/pracGuide/pracGuideTopic_10.aspx.
  • Hyman BM, Pedrick C. The OCD workbook. Your guide to breaking free from obsessive compulsive disorder. 3rd ed. Oakland, CA: New Harbinger Publications Inc; 2010.
  • Baer L. Getting control: overcoming your obsessions and compulsions. Revised ed. New York, NY: Plume; 2000.

Drug Brand Names

  • Alprazolam • Xanax
  • Aripiprazole • Abilify
  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Clonazepam • Klonopin
  • Dextroamphetamine • Adderall
  • Duloxetine • Cymbalta
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Haloperidol • Haldol
  • Ketamine • Ketalar
  • Memantine • Namenda
  • Methylphenidate • Ritalin
  • Morphine • MS Contin
  • Olanzapine • Zyprexa
  • Ondansetron • Zofran
  • Pindolol • Visken
  • Quetiapine • Seroquel
  • Riluzole • Rilutek
  • Risperidone • Risperdal
  • Sertraline • Zoloft
  • Tramadol • Ultram
  • Venlafaxine • Effexor
  • Ziprasidone • Geodon

Disclosures

Drs. Khalsa and Schiffman report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Bystritsky receives grant support from AstraZeneca, Brainways, Takeda, and Transcept and is a founder, stockholder, and consultant for BrainSonix.

References

1. Pallanti S, Quercioli L. Treatment-refractory obsessive-compulsive disorder: methodological issues operational definitions and therapeutic lines. Prog Neuropsychopharmacol Biol Psychiatry. 2006;30(3):400-412.

2. Bystritsky A. Treatment-resistant anxiety disorders. Mol Psychiatry. 2006;11(9):805-814.

3. Denys D. Pharmacotherapy of obsessive-compulsive disorder and obsessive-compulsive spectrum disorders. Psychiatr Clin North Am. 2006;29(2):553-584 xi.

4. Ackerman DL, Greenland S. Multivariate meta-analysis of controlled drug studies for obsessive-compulsive disorder. J Clin Psychopharmacol. 2002;22(3):309-317.

5. Soomro GM, Altman D, Rajagopal S, et al. Selective serotonin re-uptake inhibitors (SSRIs) versus placebo for obsessive compulsive disorder (OCD). Cochrane Database Syst Rev. 2008;(1):CD001765.-

6. Bloch MH, McGuire J, Landeros-Weisenberger A, et al. Meta-analysis of the dose-response relationship of SSRI in obsessive-compulsive disorder. Mol Psychiatry. 2010;15(8):850-855.

7. Koran LM, Hanna GL, Hollander E, et al. American Psychiatric Association. Practice guideline for the treatment of patients with obsessive-compulsive disorder. Am J Psychiatry. 2007;164(7 suppl):5-53.

8. Ninan PT, Koran LM, Kiev A, et al. High-dose sertraline strategy for nonresponders to acute treatment for obsessive-compulsive disorder: a multicenter double-blind trial. J Clin Psychiatry. 2006;67(1):15-22.

9. Food and Drug Administration. FDA drug safety communication: abnormal heart rhythms associated with high doses of Celexa (citalopram hydrobromide). http://www.fda.gov/Drugs/DrugSafety/ucm269086.htm#sa. Published August 24 2011. Accessed September 27, 2011.

10. Yaryura-Tobias JA, Neziroglu FA. Venlafaxine in obsessive-compulsive disorder. Arch Gen Psychiatry. 1996;53(7):653-654.

11. Phelps NJ, Cates ME. The role of venlafaxine in the treatment of obsessive-compulsive disorder. Ann Pharmacother. 2005;39(1):136-140.

12. Dell’osso B, Mundo E, Marazziti D, et al. Switching from serotonin reuptake inhibitors to duloxetine in patients with resistant obsessive compulsive disorder: a case series. J Psychopharmacol. 2008;22(2):210-213.

13. Pallanti S, Quercioli L, Paiva RS, et al. Citalopram for treatment-resistant obsessive-compulsive disorder. Eur Psychiatry. 1999;14:101-106.

14. Ravizza L, Barzega G, Bellino S, et al. Therapeutic effect and safety of adjunctive risperidone in refractory obsessive-compulsive disorder (OCD). Psychopharmacol Bull. 1996;32:677-682.

15. Fallon BA, Liebowitz MR, Campeas R, et al. Intravenous clomipramine for obsessive-compulsive disorder refractory to oral clomipramine: a placebo-controlled study. Arch Gen Psychiatry. 1998;55:918-924.

16. Koran LM, Pallanti S, Paiva RS, et al. Pulse loading versus gradual dosing of intravenous clomipramine in obsessive-compulsive disorder. Eur Neuropsychopharmacol. 1998;8:121-126.

17. Dannon PN, Sasson Y, Hirschmann S, et al. Pindolol augmentation in treatment-resistant obsessive compulsive disorder: a double-blind placebo controlled trial. Eur Neuropsychopharmacol. 2000;10:165-169.

18. Koran LM, Mueller K, Maloney A. Will pindolol augment the response to a serotonin reuptake inhibitor in obsessive-compulsive disorder? J Clin Psychopharmacol. 1996;16:253-254.

19. Hewlett WA, Vinogradov S, Agras WS. Clomipramine clonazepam, and clonidine treatment of obsessive-compulsive disorder. J Clin Psychopharmacol. 1992;12:420-430.

20. Mundo E, Guglielmo E, Bellodi L. Effect of adjuvant pindolol on the antiobsessional response to fluvoxamine: a double-blind placebo-controlled study. Int Clin Psychopharmacol. 1998;13:219-224.

21. Hewlett WA, Schmid SP, Salomon RM. Pilot trial of ondansetron in the treatment of 8 patients with obsessive-compulsive disorder. J Clin Psychiatry. 2003;64:1025-1030.

22. Pallanti S, Bernardi S, Antonini S, et al. Ondansetron augmentation in treatment-resistant obsessive-compulsive disorder: a preliminary, single-blind, prospective study. CNS Drugs. 2009;23(12):1047-1055.

23. Connor KM, Payne VM, Gadde KM, et al. The use of aripiprazole in obsessive-compulsive disorder: preliminary observations in 8 patients. J Clin Psychiatry. 2005;66:49-51.

24. Komossa K, Depping AM, Meyer M, et al. Second-generation antipsychotics for obsessive compulsive disorder. Cochrane Database Syst Rev. 2010;(12):CD008141.-

25. Bloch MH, Landeros-Weisenberger A, Kelmendi B, et al. A systematic review: antipsychotic augmentation with treatment refractory obsessive-compulsive disorder. Mol Psychiatry. 2006;11(7):622-632.

26. Muscatello MR, Bruno A, Pandolfo G, et al. Effect of aripiprazole augmentation of serotonin reuptake inhibitors or clomipramine in treatment-resistant obsessive-compulsive disorder: a double-blind, placebo-controlled study. J Clin Psychopharmacol. 2011;31(2):174-179.

27. Savas HA, Yumru M, Ozen ME. Quetiapine and ziprasidone as adjuncts in treatment-resistant obsessive-compulsive disorder: a retrospective comparative study. Clin Drug Investig. 2008;28(7):439-442.

28. Hollander E, Kaplan A, Stahl SM. A double-blind placebo-controlled trial of clonazepam in obsessive-compulsive disorder. World J Biol Psychiatry. 2003;4:30-34.

29. Crockett BA, Churchill E, Davidson JR. A double-blind combination study of clonazepam with sertraline in obsessive-compulsive disorder. Ann Clin Psychiatry. 2004;16(3):127-132.

30. Koran LM, Aboujaoude E, Bullock KD, et al. Double-blind treatment with oral morphine in treatment-resistant obsessive-compulsive disorder. J Clin Psychiatry. 2005;66(3):353-359.

31. Shapira NA, Keck PE Jr, Goldsmith TD, et al. Open-label pilot study of tramadol hydrochloride in treatment-refractory obsessive-compulsive disorder. Depress Anxiety. 1997;6:170-173.

32. Goldsmith TB, Shapira NA, Keck PE Jr. Rapid remission of OCD with tramadol hydrochloride. Am J Psychiatry. 1999;156(4):660-661.

33. Insel TR, Hamilton JA, Guttmacher LB, et al. D-amphetamine in obsessive-compulsive disorder. Psychopharmacology (Berl). 1983;80:231-235.

34. Joffe RT, Swinson RP, Levitt AJ. Acute psychostimulant challenge in primary obsessive-compulsive disorder. J Clin Psychopharmacol. 1991;11:237-241.

35. Woolley JB, Heyman I. Dexamphetamine for obsessive-compulsive disorder. Am J Psychiatry. 2003;160:183.-

36. Aboujaoude E, Barry JJ, Gamel N. Memantine augmentation in treatment-resistant obsessive-compulsive disorder: an open-label trial. J Clin Psychopharmacol. 2009;29(1):51-55.

37. Coric V, Taskiran S, Pittenger C, et al. Riluzole augmentation in treatment-resistant obsessive-compulsive disorder: an open-label trial. Biol Psychiatry. 2005;58(5):424-428.

38. Rodriguez CI, Kegeles LS, Flood P, et al. Rapid resolution of obsessions after an infusion of intravenous ketamine in a patient with treatment-resistant obsessive-compulsive disorder. J Clin Psychiatry. 2011;72(4):567-569.

39. Leonard HL, Rapoport JL. Relief of obsessive-compulsive symptoms by LSD and psilocin. Am J Psychiatry. 1987;144(9):1239-1240.

40. Anand N, Sudhir PM, Math SB, et al. Cognitive behavior therapy in medication non-responders with obsessive-compulsive disorder: a prospective 1-year follow-up study. J Anxiety Disord. 2011;25(7):939-945.

41. Himle JA, Fischer DJ, Muroff JR, et al. Videoconferencing-based cognitive-behavioral therapy for obsessive-compulsive disorder. Behav Res Ther. 2006;44(12):1821-1829.

42. Kobak KA, Taylor LV, Bystritsky A, et al. St John’s wort versus placebo in obsessive-compulsive disorder: results from a double-blind study. Int Clin Psychopharmacol. 2005;20(6):299-304.

43. Sayyah M, Boostani H, Pakseresht S, et al. Comparison of Silybum marianum (L.) Gaertn. with fluoxetine in the treatment of obsessive-compulsive Disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34(2):362-365.

44. Sarris J, Camfield D, Berk M. Complementary medicine self-help, and lifestyle interventions for obsessive compulsive disorder (OCD) and the OCD spectrum: a systematic review. J Affect Disord. 2011 (epub ahead of print).

45. Beale MD, Kellner CH, Pritchett JT, et al. ECT for OCD. J Clin Psychiatry. 1995;56(2):81-82.

46. George MS, Ward HE Jr, Ninan PT, et al. A pilot study of vagus nerve stimulation (VNS) for treatment-resistant anxiety disorders. Brain Stimul. 2008;1(2):112-121.

47. Slotema CW, Blom JD, Hoek HW, et al. Should we expand the toolbox of psychiatric treatment methods to include repetitive transcranial magnetic stimulation (rTMS)? A meta-analysis of the efficacy of rTMS in psychiatric disorders. J Clin Psychiatry. 2010;71(7):873-884.

48. Mantovani A, Simpson HB, Fallon BA, et al. Randomized sham-controlled trial of repetitive transcranial magnetic stimulation in treatment-resistant obsessive-compulsive disorder. Int J Neuropsychopharmacol. 2010;13(2):217-227.

49. Blom RM, Figee M, Vulink N, et al. Update on repetitive transcranial magnetic stimulation in obsessive-compulsive disorder: different targets. Curr Psychiatry Rep. 2011;13(4):289-294.

50. Greenberg BD, Rauch SL, Haber SN. Invasive circuitry-based neurotherapeutics: stereotactic ablation and deep brain stimulation for OCD. Neuropsychopharmacology. 2010;35(1):317-336.

51. Nuttin B, Cosyns P, Demeulemeester H, et al. Electrical stimulation in anterior limbs of internal capsules in patients with obsessive-compulsive disorder. Lancet. 1999;354(9189):1526.-

52. de Koning PP, Figee M, van den Munckhof P, et al. Current status of deep brain stimulation for obsessive-compulsive disorder: a clinical review of different targets. Curr Psychiatry Rep. 2011;13(4):274-282.

53. Greenberg BD, Gabriels LA, Malone DA, Jr, et al. Deep brain stimulation of the ventral internal capsule/ventral striatum for obsessive-compulsive disorder: worldwide experience. Mol Psychiatry. 2010;15(1):64-79.

Article PDF
Author and Disclosure Information

Sahib S. Khalsa, MD, PhD
Resident Physician, UCLA Anxiety Disorders Program, Department of Psychiatry, UCLA David Geffen School of Medicine, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA
Jason E. Schiffman, MD, MA, MBA
Chief Resident Physician, UCLA Anxiety Disorders Program, Department of Psychiatry, UCLA David Geffen School of Medicine, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA
Alexander Bystritsky, MD, PhD
Director, UCLA Anxiety Disorders Program, Professor of Psychiatry and Biobehavioral Sciences, Department of Psychiatry, UCLA David Geffen School of Medicine, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA

Audio / Podcast
Issue
Current Psychiatry - 10(11)
Publications
Topics
Page Number
44-52
Legacy Keywords
treatment-resistant OCD; OCD; obsessive-compulsive disorder; Sahib Khalsa; Jason Schiffman; Alexander Bystrirsky; first-line pharmacotherapy; augmentation; serotonergic agents; supratherapeutic SSRI; SNRIs; clomipramine; IV clomipramine; pindolol; ondansetron; antipsychotics; benzodiazepines; opioids; psychostimulants; NMDA antagonists; hallucinogens; psychotherapy; alternative medicine; invasive therapies; ECT; VNS; rTMS; surgical approaches; deep brain stimulation
Sections
Audio / Podcast
Audio / Podcast
Author and Disclosure Information

Sahib S. Khalsa, MD, PhD
Resident Physician, UCLA Anxiety Disorders Program, Department of Psychiatry, UCLA David Geffen School of Medicine, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA
Jason E. Schiffman, MD, MA, MBA
Chief Resident Physician, UCLA Anxiety Disorders Program, Department of Psychiatry, UCLA David Geffen School of Medicine, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA
Alexander Bystritsky, MD, PhD
Director, UCLA Anxiety Disorders Program, Professor of Psychiatry and Biobehavioral Sciences, Department of Psychiatry, UCLA David Geffen School of Medicine, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA

Author and Disclosure Information

Sahib S. Khalsa, MD, PhD
Resident Physician, UCLA Anxiety Disorders Program, Department of Psychiatry, UCLA David Geffen School of Medicine, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA
Jason E. Schiffman, MD, MA, MBA
Chief Resident Physician, UCLA Anxiety Disorders Program, Department of Psychiatry, UCLA David Geffen School of Medicine, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA
Alexander Bystritsky, MD, PhD
Director, UCLA Anxiety Disorders Program, Professor of Psychiatry and Biobehavioral Sciences, Department of Psychiatry, UCLA David Geffen School of Medicine, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA

Article PDF
Article PDF

Obsessive-compulsive disorder (OCD) is marked by recurrent and persistent anxiety-provoking thoughts (obsessions) accompanied by repetitive behaviors (compulsions) that focus on alleviating distress caused by obsessive thoughts. Although patients recognize the obsessions and compulsions are unreasonable, these thoughts and behaviors remain time-consuming and impair function. Even when they appropriately identify and treat OCD, clinicians often face “treatment-resistant” (or “treatment-refractory”) patients who do not respond adequately to standard therapies (Box).1 Several factors contribute to treatment resistance, including those related to the patient, the environment, the clinician/health system, and pathology (Table 1).2 An estimated 10% to 40% of patients with OCD are treatment-resistant.2

This article discusses the range of options for addressing resistant OCD, including augmenting first-line treatments with pharmacotherapy, psychotherapy, or reversible or irreversible forms of neuromodulation.

Defining treatment resistance in obsessive-compulsive disorder

Treatment resistance generally refers to lack of sufficient improvement despite multiple adequate and appropriate treatment trials. However, there are no universally accepted definitions or metrics of treatment resistance, and often it is operationally defined. For mood disorders, it may be defined by failure to remit or respond clinically (50% reduction in symptoms) despite ≥2 adequate antidepressant trials or failure to respond clinically despite adequate medication trials across several neurotransmitter classes. The terms treatment resistant and treatment refractory are synonymous; they refer to the same phenomenon and are used interchangeably in the literature. Including the terms “remission” and “recovery” when judging treatment efficacy for anxiety disorders can be limiting because of the chronic and often unrelenting nature of these conditions.

One review proposed categorizing obsessive-compulsive disorder treatment response into several stages along a spectrum, ranging from complete recovery (or remission) to full or partial response to non-response (or completely refractory).1 However it is defined, treatment resistance in anxiety disorders likely is characterized by minimal restoration of function despite several appropriate treatment exposures.

Table 1

Factors that contribute to treatment resistance in obsessive-compulsive disorder

Patient
Disease severity
Medical comorbidity
Psychiatric comorbidity (mood, personality, and/or substance use disorders)
Treatment nonadherence
Cultural factors
Environment
Childhood stressors (trauma, abuse)
Long-term persistent stressors (psychosocial, occupational, financial)
Life stages
Clinician/health system
Lack of knowledge in primary care (brief treatment duration, subtherapeutic dosing)
Lack of psychotherapeutic training
Limited doctor-patient relationship (eg, availability/cost of treatment)
Pathology-related
Underlying disease pathophysiology (largely unknown):
  • Multiple neurotransmitter system interactions
  • Polygenetic influences (genetic load)
  • Gene-environment interactions
  • Neural circuits (cortical and subcortical feedback loops)
Diagnostic variance (dimensional vs categorical vs target symptom approach)
Syndromal variation (differing presentations over time)
Treatment limitations (limited empirical studies, nonrepresentative study samples)
Source: Reference 2

First-line pharmacotherapy

Clomipramine or a selective serotonin reuptake inhibitor (SSRI) are considered first-line treatments for OCD. Although some evidence indicates that clomipramine may have greater efficacy than SSRIs, its poor tolerability and potential lethality in overdose make it a less practical first choice in treatment-naïve patients.3,4 SSRIs generally are well tolerated and have a favorable safety profile. Nearly all SSRIs have randomized clinical trials (RCTs) and FDA indications that support their use in OCD. SSRI choice may be guided by patient or prescriber preference because no evidence suggests that 1 SSRI is superior to another for treating OCD.5 In contrast to major depressive disorder, in OCD there is a dose-response relationship for SSRI treatment; higher doses typically are required to achieve response or remission.6,7

Augmentation and other options

Patients who have not responded to at least 2 adequate trials of first-line medications may benefit from an augmentation strategy or treatment with an unconventional agent. Such cases should be managed by a specialist who has experience in treating OCD and with careful consideration of potential risks of these interventions.

Evidence suggests the following pharmacotherapies may effectively treat OCD and may be warranted for treatment-resistant patients.

Serotonergic agents

Supratherapeutic SSRI doses. Evidence suggests that supratherapeutic doses of SSRIs may be effective, which may be a logical first step when treating patients already taking an SSRI who have not responded. In a multi-center, double-blind study comparing sertraline, 200 mg/d, to sertraline, 250 to 400 mg/d, the latter group showed significantly greater symptom improvement.8 Citalopram may not be suitable for this approach because of the recent FDA announcement regarding dose-dependent QTc prolongation associated with this medication.9

Serotonin-norepinephrine reuptake inhibitors (SNRIs). In the only double-blind, placebo-controlled study of venlafaxine for OCD, the drug was not significantly more effective than placebo.10 This study was small (N = 30). There are sufficient positive results from open-label and blinded comparator studies that venlafaxine generally is accepted as an effective and well-tolerated treatment for OCD at doses ≥225 mg/d.11

 

 

Duloxetine also may be effective in treating OCD. One case series reported improvement in 3 of 4 SSRI nonresponders who were switched to this medication and rapidly titrated to 120 mg/d.12

Clomipramine/SSRI augmentation. For patients who have not responded to an SSRI, several open-label trials support adding clomipramine.13 Conversely, SSRI augmentation for patients who have not adequately responded to clomipramine may be effective.14 With any dual therapy with serotonergic agents, monitor patients for signs and symptoms of serotonin syndrome.

IV clomipramine. By bypassing first-pass metabolism, IV clomipramine rapidly achieves high plasma levels. In a double-blind, placebo-controlled study of 54 OCD patients who were nonresponsive to oral clomipramine, IV clomipramine was more effective than placebo.15 An additional study found IV clomipramine is more effective when pulse loaded than when titrated gradually.16

Pindolol. The beta blocker pindolol acts as an antagonist of presynaptic 5-HT1A autoreceptors, increasing serotonergic signaling. A small double-blind, placebo-controlled trial (N = 14) found a significant decrease in Yale-Brown Obsessive Compulsive Scale (Y-BOCS) score with pindolol augmentation, 2.5 mg, 3 times daily, among patients who did not respond to ≥3 serotonin reuptake inhibitor (SRI) trials.17 Pindolol augmentation showed modest effects in 2 open-label studies.18,19 However, another small double-blind, placebo-controlled study (N = 15) found no difference between placebo and fluvoxamine augmented with pindolol.20

Ondansetron. A 5-HT3 receptor antagonist, ondansetron is used primarily as an antiemetic but has been shown to have anxiolytic properties in animal studies. In an open-label study of 8 patients with non–treatment refractory OCD, 3 achieved clinical response (at least 35% reduction in Y-BOCS score) with ondansetron monotherapy dosed at 1 mg, 3 times daily.21 In a subsequent single-blind trial with 14 treatment-resistant patients, 9 responded (at least 25% reduction in Y-BOCS score).22

Other medications

Antipsychotics. Most studies examining antipsychotic monotherapy for OCD have been negative. One exception was a small, open-label trial of aripiprazole monotherapy (N = 8) that found modest efficacy among non–treatment refractory patients.23 Augmentation with antipsychotics, however, has been well studied and there is good evidence of efficacy for this approach. Double-blind, placebo-controlled studies have supported the efficacy of augmenting SRIs with haloperidol, risperidone, olanzapine, quetiapine, and aripiprazole.24-26 Several case reports suggest ziprasidone may be an effective SRI adjunct, but 1 retrospective study found it was inferior to quetiapine.27

Benzodiazepines. Case reports present positive effects of clonazepam and alprazolam for OCD, but double-blind, placebo-controlled trials for monotherapy or adjunctive clonazepam have been negative.28,29 Furthermore, cognitive impairment and potential for dependence associated with benzodiazepines weigh against their use in OCD.

Opioids. A double-blind, placebo controlled crossover study of 23 patients with treatment-refractory OCD found once-weekly oral morphine added to patients’ current regimen significantly reduced Y-BOCS score vs placebo. Patients received 30 mg the first week and 15 to 45 mg the next week, depending on response or side effects.30 A case report and a small open-label trial support the efficacy of tramadol, a weak agonist of the μ opioid receptor and an inhibitor of serotonin and norepinephrine transporters, as monotherapy and as an adjunct to fluoxetine.31,32 Because patients with OCD may be particularly vulnerable to dependence and intentional or accidental overdose via opioid/benzodiazepine combinations, evaluate the risks and benefits before initiating an opioid.

Psychostimulants. Sparse but good evidence supports the efficacy of dextroamphetamine monotherapy for OCD.33,34 There are no positive studies of methylphenidate and several case reports of methylphenidate-induced OCD symptoms.35

N-methyl-D-aspartate (NMDA) antagonists. Increased glutamatergic neurotransmission has been implicated in the pathophysiology of OCD, which suggests a possible role for glutamate receptor antagonists. In an open-label trial, memantine, an NMDA antagonist used primarily to treat dementia, was associated with clinical response (>25% reduction in Y-BOCS scores) in 6 of 14 patients with treatment-refractory OCD.36 Several case reports and an open-label trial support the efficacy of riluzole—which is indicated for treating amyotrophic lateral sclerosis—as an adjunct for treatment-refractory OCD.37 Although its exact mechanism of action is unclear, riluzole’s effects are thought to be mediated via reduction in glutamatergic neurotransmission. IV ketamine has reported anti-OCD effects in a case report of a woman with treatment-resistant OCD. These effects occurred almost immediately and persisted for several days.38

Hallucinogens. Psilocybin, psilocin, and lysergic acid diethylamide have reported anti-OCD properties.39 As schedule I substances, however, they are not available outside of sanctioned research protocols and may carry substantial risk. Nonetheless, their efficacy suggests that other compounds that share their mechanism of action—namely agonism of 5-HT2A and 5-HT2C receptors—may merit investigation as potential treatments for OCD.

 

 

Psychotherapy

Cognitive-behavioral therapy (CBT) has been shown to be effective for OCD as monotherapy and augmentation to pharmacotherapy. CBT consists of cognitive and behavioral components, typically involving some form of cognitive restructuring and exposure response prevention. Although these 2 types of interventions arise from independent traditions, in CBT they are frequently intertwined, particularly when the focus of OCD patients’ anxiety is ego-dystonic thoughts.

One benefit of CBT over pharmacotherapy is that effects persist after treatment is terminated. A recent prospective study found CBT was effective for treatment-refractory OCD, with 74% of patients demonstrating clinical response after 20 to 25 sessions over 2 months and 61% maintaining clinical response 1 year after treatment.40 CBT administered remotely via teleconference, also known as “teletherapy,” has shown efficacy for OCD.41

Alternative medicine

Despite widespread use of herbal remedies for OCD, no trials have shown a strong positive effect. Both Hypericum perforatum (St. John’s wort) and Silybum marianum (milk thistle) have been used to treat obsessive and compulsive symptoms; however, placebo-controlled trials did not find any significant differences in symptoms or side effects between treatment groups.42,43 Lower-quality studies have reported modest effects for mindfulness meditation, yoga, and acupuncture.44

Because many patients continue to use complementary and alternative medicine therapies despite the lack of data on efficacy, it is important to monitor for potential interactions with prescription medications. St. John’s wort interacts with many medications because of induction of the cytochrome P450 (CYP) isoenzymes 3A4 and 2C9. This interaction may lower blood levels of alprazolam and clonazepam (3A4). Combining St. John’s wort with SSRIs increases the risk of serotonin syndrome. Milk thistle inhibits CYP450 isoenzyme 3A4, and may increase serum levels of other medications metabolized by this pathway.

Invasive therapies

Invasive options may be considered after several pharmacotherapeutic and psychotherapeutic approaches have not been effective or when significant functional impairment remains (Table 2). These therapies typically are reserved for patients whose treatment resistance is strongest.

Electroconvulsive therapy (ECT). Although ECT is an effective tool for treatment-resistant mood disorders or treatment-resistant anxiety complicated by severe depression, studies have not found ECT to be effective for OCD. One uncontrolled case series reported considerable improvements in OCD patients the year after ECT, although improvement was correlated with improved depression scores.45

Vagal nerve stimulation (VNS). In an open-label study of 7 OCD patients who received VNS, 3 were acute responders—characterized by a ≥25% improvement on the Y-BOCS—and 2 received continued benefits at 4-year follow up (2 patients dropped out).46

Repetitive transcranial magnetic stimulation (rTMS). A meta-analysis of 3 RCTs of rTMS for patients with OCD did not yield a large or statistically significant effect.47 Limitations of these trials included asymmetric stimulation sites (eg, left vs right only), limited stimulation sites (dorsolateral prefrontal cortex), different stimulation frequencies between studies, and a lack of sham stimulation conditions. A more recent RCT and subsequent review described moderate efficacy (defined by ≥25% decrease in Y-BOCS scores) compared with sham stimulations in OCD patients at 4 weeks, using the supplementary motor area as a stimulation site.48,49

The main limitation of rTMS is the inability to penetrate deeper brain structures implicated in OCD (eg, caudate nucleus, thalamus, anterior capsule fiber tracts), as well as a lack of specificity in stimulation site.

Surgical approaches. Cingulotomy is the most commonly employed surgical procedure for OCD in North America, likely because of a combination of clinical efficacy and low morbidity and mortality rates.50 Of the >1,000 cingulotomies that have been performed at Massachusetts General Hospital, no deaths or postoperative infections have been reported and 2 subdural hematomas have occurred.50 Common postsurgical side effects include transient headache, nausea, or difficulty urinating. The most serious common side effect—postoperative seizures—has been reported in 1% to 9% of cases.

Outcomes for these procedures cannot be fully assessed until at least 6 months to 2 years after the procedure, which suggests postoperative neural reorganization plays an important role in recovery. Direct comparisons of each lesion approach within studies are extremely rare. Overall, long-term outcomes of these approaches have demonstrated significant therapeutic effects of each of these procedures. Reported response rates vary between 30% to 70%, when applied to remission, response (≥35% Y-BOCS reduction), and functional improvements in quality of life.50

 

 

Deep brain stimulation (DBS). With this approach, small electrodes are inserted under precise stereotactic MRI guidance. The advantage of DBS over ablative surgery is the ability to adjust and customize neurostimulation. Following implantation, modifiable parameters of electrode stimulation include electrode polarity, intensity, frequency, and laterality. A specially trained psychiatrist can conduct parameter optimization during long-term follow-up.

The first trial of DBS for OCD was reported in 1999 (N = 4), with the initial target selected based on the site of anterior capsulotomy. Three patients derived clinically observed benefit, although no validated questionnaires were administered.51 Since then, at least 7 studies with blinded stimulation have been conducted, totaling 62 patients.52

In recent years, structures adjacent to the internal capsule also have been targeted based on the approach employed in ventral capsulotomy. Across all trials, response rates for this approach consistently have been in the 50% range, with average Y-BOCS score reductions ranging from 6.8 to 31 points.53 Some patients have reported rapid improvements in anhedonia, and this approach is being employed in treatment-resistant depression.

Postoperative complications occur more often with DBS than with lesion approaches because of the prosthetic nature of the procedure (eg, increased risk of infection, lead malfunction, etc.). Additionally, batteries must be periodically explanted and replaced. Reported stimulation-related side effects include mood changes (transient sadness, anxiety, euphoria, and hypomania), sensory disturbances (olfactory, gustatory, and motor sensations), and cognitive changes (confusion and forgetfulness). These side effects typically are stimulation-dependent and disappear after altering stimulation parameters.

Table 2

Invasive therapies for treatment-resistant OCD

TherapyQuality of evidence
Reversible
  Electroconvulsive therapyPoor
  Vagal nerve stimulationPoor
  Repetitive transcranial magnetic stimulationLimited
Irreversible (surgical)
  Anterior capsulotomy. Target: anterior limb of the internal capsuleFair
  Anterior cingulotomy. Target: anterior cingulate and cingulum bundleFair
  Subcaudate tractotomy. Target: substantia innominata, just inferior to the caudate nucleusFair
  Limbic leucotomy. Target: anterior cingulotomy combined with subcaudate tractotomyFair
  Deep brain stimulation. Multiple targetsFair
OCD: obsessive-compulsive disorder

Related Resources

  • American Psychiatric Association. Treatment of patients with obsessive-compulsive disorder. www.psychiatryonline.com/pracGuide/pracGuideTopic_10.aspx.
  • Hyman BM, Pedrick C. The OCD workbook. Your guide to breaking free from obsessive compulsive disorder. 3rd ed. Oakland, CA: New Harbinger Publications Inc; 2010.
  • Baer L. Getting control: overcoming your obsessions and compulsions. Revised ed. New York, NY: Plume; 2000.

Drug Brand Names

  • Alprazolam • Xanax
  • Aripiprazole • Abilify
  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Clonazepam • Klonopin
  • Dextroamphetamine • Adderall
  • Duloxetine • Cymbalta
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Haloperidol • Haldol
  • Ketamine • Ketalar
  • Memantine • Namenda
  • Methylphenidate • Ritalin
  • Morphine • MS Contin
  • Olanzapine • Zyprexa
  • Ondansetron • Zofran
  • Pindolol • Visken
  • Quetiapine • Seroquel
  • Riluzole • Rilutek
  • Risperidone • Risperdal
  • Sertraline • Zoloft
  • Tramadol • Ultram
  • Venlafaxine • Effexor
  • Ziprasidone • Geodon

Disclosures

Drs. Khalsa and Schiffman report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Bystritsky receives grant support from AstraZeneca, Brainways, Takeda, and Transcept and is a founder, stockholder, and consultant for BrainSonix.

Obsessive-compulsive disorder (OCD) is marked by recurrent and persistent anxiety-provoking thoughts (obsessions) accompanied by repetitive behaviors (compulsions) that focus on alleviating distress caused by obsessive thoughts. Although patients recognize the obsessions and compulsions are unreasonable, these thoughts and behaviors remain time-consuming and impair function. Even when they appropriately identify and treat OCD, clinicians often face “treatment-resistant” (or “treatment-refractory”) patients who do not respond adequately to standard therapies (Box).1 Several factors contribute to treatment resistance, including those related to the patient, the environment, the clinician/health system, and pathology (Table 1).2 An estimated 10% to 40% of patients with OCD are treatment-resistant.2

This article discusses the range of options for addressing resistant OCD, including augmenting first-line treatments with pharmacotherapy, psychotherapy, or reversible or irreversible forms of neuromodulation.

Defining treatment resistance in obsessive-compulsive disorder

Treatment resistance generally refers to lack of sufficient improvement despite multiple adequate and appropriate treatment trials. However, there are no universally accepted definitions or metrics of treatment resistance, and often it is operationally defined. For mood disorders, it may be defined by failure to remit or respond clinically (50% reduction in symptoms) despite ≥2 adequate antidepressant trials or failure to respond clinically despite adequate medication trials across several neurotransmitter classes. The terms treatment resistant and treatment refractory are synonymous; they refer to the same phenomenon and are used interchangeably in the literature. Including the terms “remission” and “recovery” when judging treatment efficacy for anxiety disorders can be limiting because of the chronic and often unrelenting nature of these conditions.

One review proposed categorizing obsessive-compulsive disorder treatment response into several stages along a spectrum, ranging from complete recovery (or remission) to full or partial response to non-response (or completely refractory).1 However it is defined, treatment resistance in anxiety disorders likely is characterized by minimal restoration of function despite several appropriate treatment exposures.

Table 1

Factors that contribute to treatment resistance in obsessive-compulsive disorder

Patient
Disease severity
Medical comorbidity
Psychiatric comorbidity (mood, personality, and/or substance use disorders)
Treatment nonadherence
Cultural factors
Environment
Childhood stressors (trauma, abuse)
Long-term persistent stressors (psychosocial, occupational, financial)
Life stages
Clinician/health system
Lack of knowledge in primary care (brief treatment duration, subtherapeutic dosing)
Lack of psychotherapeutic training
Limited doctor-patient relationship (eg, availability/cost of treatment)
Pathology-related
Underlying disease pathophysiology (largely unknown):
  • Multiple neurotransmitter system interactions
  • Polygenetic influences (genetic load)
  • Gene-environment interactions
  • Neural circuits (cortical and subcortical feedback loops)
Diagnostic variance (dimensional vs categorical vs target symptom approach)
Syndromal variation (differing presentations over time)
Treatment limitations (limited empirical studies, nonrepresentative study samples)
Source: Reference 2

First-line pharmacotherapy

Clomipramine or a selective serotonin reuptake inhibitor (SSRI) are considered first-line treatments for OCD. Although some evidence indicates that clomipramine may have greater efficacy than SSRIs, its poor tolerability and potential lethality in overdose make it a less practical first choice in treatment-naïve patients.3,4 SSRIs generally are well tolerated and have a favorable safety profile. Nearly all SSRIs have randomized clinical trials (RCTs) and FDA indications that support their use in OCD. SSRI choice may be guided by patient or prescriber preference because no evidence suggests that 1 SSRI is superior to another for treating OCD.5 In contrast to major depressive disorder, in OCD there is a dose-response relationship for SSRI treatment; higher doses typically are required to achieve response or remission.6,7

Augmentation and other options

Patients who have not responded to at least 2 adequate trials of first-line medications may benefit from an augmentation strategy or treatment with an unconventional agent. Such cases should be managed by a specialist who has experience in treating OCD and with careful consideration of potential risks of these interventions.

Evidence suggests the following pharmacotherapies may effectively treat OCD and may be warranted for treatment-resistant patients.

Serotonergic agents

Supratherapeutic SSRI doses. Evidence suggests that supratherapeutic doses of SSRIs may be effective, which may be a logical first step when treating patients already taking an SSRI who have not responded. In a multi-center, double-blind study comparing sertraline, 200 mg/d, to sertraline, 250 to 400 mg/d, the latter group showed significantly greater symptom improvement.8 Citalopram may not be suitable for this approach because of the recent FDA announcement regarding dose-dependent QTc prolongation associated with this medication.9

Serotonin-norepinephrine reuptake inhibitors (SNRIs). In the only double-blind, placebo-controlled study of venlafaxine for OCD, the drug was not significantly more effective than placebo.10 This study was small (N = 30). There are sufficient positive results from open-label and blinded comparator studies that venlafaxine generally is accepted as an effective and well-tolerated treatment for OCD at doses ≥225 mg/d.11

 

 

Duloxetine also may be effective in treating OCD. One case series reported improvement in 3 of 4 SSRI nonresponders who were switched to this medication and rapidly titrated to 120 mg/d.12

Clomipramine/SSRI augmentation. For patients who have not responded to an SSRI, several open-label trials support adding clomipramine.13 Conversely, SSRI augmentation for patients who have not adequately responded to clomipramine may be effective.14 With any dual therapy with serotonergic agents, monitor patients for signs and symptoms of serotonin syndrome.

IV clomipramine. By bypassing first-pass metabolism, IV clomipramine rapidly achieves high plasma levels. In a double-blind, placebo-controlled study of 54 OCD patients who were nonresponsive to oral clomipramine, IV clomipramine was more effective than placebo.15 An additional study found IV clomipramine is more effective when pulse loaded than when titrated gradually.16

Pindolol. The beta blocker pindolol acts as an antagonist of presynaptic 5-HT1A autoreceptors, increasing serotonergic signaling. A small double-blind, placebo-controlled trial (N = 14) found a significant decrease in Yale-Brown Obsessive Compulsive Scale (Y-BOCS) score with pindolol augmentation, 2.5 mg, 3 times daily, among patients who did not respond to ≥3 serotonin reuptake inhibitor (SRI) trials.17 Pindolol augmentation showed modest effects in 2 open-label studies.18,19 However, another small double-blind, placebo-controlled study (N = 15) found no difference between placebo and fluvoxamine augmented with pindolol.20

Ondansetron. A 5-HT3 receptor antagonist, ondansetron is used primarily as an antiemetic but has been shown to have anxiolytic properties in animal studies. In an open-label study of 8 patients with non–treatment refractory OCD, 3 achieved clinical response (at least 35% reduction in Y-BOCS score) with ondansetron monotherapy dosed at 1 mg, 3 times daily.21 In a subsequent single-blind trial with 14 treatment-resistant patients, 9 responded (at least 25% reduction in Y-BOCS score).22

Other medications

Antipsychotics. Most studies examining antipsychotic monotherapy for OCD have been negative. One exception was a small, open-label trial of aripiprazole monotherapy (N = 8) that found modest efficacy among non–treatment refractory patients.23 Augmentation with antipsychotics, however, has been well studied and there is good evidence of efficacy for this approach. Double-blind, placebo-controlled studies have supported the efficacy of augmenting SRIs with haloperidol, risperidone, olanzapine, quetiapine, and aripiprazole.24-26 Several case reports suggest ziprasidone may be an effective SRI adjunct, but 1 retrospective study found it was inferior to quetiapine.27

Benzodiazepines. Case reports present positive effects of clonazepam and alprazolam for OCD, but double-blind, placebo-controlled trials for monotherapy or adjunctive clonazepam have been negative.28,29 Furthermore, cognitive impairment and potential for dependence associated with benzodiazepines weigh against their use in OCD.

Opioids. A double-blind, placebo controlled crossover study of 23 patients with treatment-refractory OCD found once-weekly oral morphine added to patients’ current regimen significantly reduced Y-BOCS score vs placebo. Patients received 30 mg the first week and 15 to 45 mg the next week, depending on response or side effects.30 A case report and a small open-label trial support the efficacy of tramadol, a weak agonist of the μ opioid receptor and an inhibitor of serotonin and norepinephrine transporters, as monotherapy and as an adjunct to fluoxetine.31,32 Because patients with OCD may be particularly vulnerable to dependence and intentional or accidental overdose via opioid/benzodiazepine combinations, evaluate the risks and benefits before initiating an opioid.

Psychostimulants. Sparse but good evidence supports the efficacy of dextroamphetamine monotherapy for OCD.33,34 There are no positive studies of methylphenidate and several case reports of methylphenidate-induced OCD symptoms.35

N-methyl-D-aspartate (NMDA) antagonists. Increased glutamatergic neurotransmission has been implicated in the pathophysiology of OCD, which suggests a possible role for glutamate receptor antagonists. In an open-label trial, memantine, an NMDA antagonist used primarily to treat dementia, was associated with clinical response (>25% reduction in Y-BOCS scores) in 6 of 14 patients with treatment-refractory OCD.36 Several case reports and an open-label trial support the efficacy of riluzole—which is indicated for treating amyotrophic lateral sclerosis—as an adjunct for treatment-refractory OCD.37 Although its exact mechanism of action is unclear, riluzole’s effects are thought to be mediated via reduction in glutamatergic neurotransmission. IV ketamine has reported anti-OCD effects in a case report of a woman with treatment-resistant OCD. These effects occurred almost immediately and persisted for several days.38

Hallucinogens. Psilocybin, psilocin, and lysergic acid diethylamide have reported anti-OCD properties.39 As schedule I substances, however, they are not available outside of sanctioned research protocols and may carry substantial risk. Nonetheless, their efficacy suggests that other compounds that share their mechanism of action—namely agonism of 5-HT2A and 5-HT2C receptors—may merit investigation as potential treatments for OCD.

 

 

Psychotherapy

Cognitive-behavioral therapy (CBT) has been shown to be effective for OCD as monotherapy and augmentation to pharmacotherapy. CBT consists of cognitive and behavioral components, typically involving some form of cognitive restructuring and exposure response prevention. Although these 2 types of interventions arise from independent traditions, in CBT they are frequently intertwined, particularly when the focus of OCD patients’ anxiety is ego-dystonic thoughts.

One benefit of CBT over pharmacotherapy is that effects persist after treatment is terminated. A recent prospective study found CBT was effective for treatment-refractory OCD, with 74% of patients demonstrating clinical response after 20 to 25 sessions over 2 months and 61% maintaining clinical response 1 year after treatment.40 CBT administered remotely via teleconference, also known as “teletherapy,” has shown efficacy for OCD.41

Alternative medicine

Despite widespread use of herbal remedies for OCD, no trials have shown a strong positive effect. Both Hypericum perforatum (St. John’s wort) and Silybum marianum (milk thistle) have been used to treat obsessive and compulsive symptoms; however, placebo-controlled trials did not find any significant differences in symptoms or side effects between treatment groups.42,43 Lower-quality studies have reported modest effects for mindfulness meditation, yoga, and acupuncture.44

Because many patients continue to use complementary and alternative medicine therapies despite the lack of data on efficacy, it is important to monitor for potential interactions with prescription medications. St. John’s wort interacts with many medications because of induction of the cytochrome P450 (CYP) isoenzymes 3A4 and 2C9. This interaction may lower blood levels of alprazolam and clonazepam (3A4). Combining St. John’s wort with SSRIs increases the risk of serotonin syndrome. Milk thistle inhibits CYP450 isoenzyme 3A4, and may increase serum levels of other medications metabolized by this pathway.

Invasive therapies

Invasive options may be considered after several pharmacotherapeutic and psychotherapeutic approaches have not been effective or when significant functional impairment remains (Table 2). These therapies typically are reserved for patients whose treatment resistance is strongest.

Electroconvulsive therapy (ECT). Although ECT is an effective tool for treatment-resistant mood disorders or treatment-resistant anxiety complicated by severe depression, studies have not found ECT to be effective for OCD. One uncontrolled case series reported considerable improvements in OCD patients the year after ECT, although improvement was correlated with improved depression scores.45

Vagal nerve stimulation (VNS). In an open-label study of 7 OCD patients who received VNS, 3 were acute responders—characterized by a ≥25% improvement on the Y-BOCS—and 2 received continued benefits at 4-year follow up (2 patients dropped out).46

Repetitive transcranial magnetic stimulation (rTMS). A meta-analysis of 3 RCTs of rTMS for patients with OCD did not yield a large or statistically significant effect.47 Limitations of these trials included asymmetric stimulation sites (eg, left vs right only), limited stimulation sites (dorsolateral prefrontal cortex), different stimulation frequencies between studies, and a lack of sham stimulation conditions. A more recent RCT and subsequent review described moderate efficacy (defined by ≥25% decrease in Y-BOCS scores) compared with sham stimulations in OCD patients at 4 weeks, using the supplementary motor area as a stimulation site.48,49

The main limitation of rTMS is the inability to penetrate deeper brain structures implicated in OCD (eg, caudate nucleus, thalamus, anterior capsule fiber tracts), as well as a lack of specificity in stimulation site.

Surgical approaches. Cingulotomy is the most commonly employed surgical procedure for OCD in North America, likely because of a combination of clinical efficacy and low morbidity and mortality rates.50 Of the >1,000 cingulotomies that have been performed at Massachusetts General Hospital, no deaths or postoperative infections have been reported and 2 subdural hematomas have occurred.50 Common postsurgical side effects include transient headache, nausea, or difficulty urinating. The most serious common side effect—postoperative seizures—has been reported in 1% to 9% of cases.

Outcomes for these procedures cannot be fully assessed until at least 6 months to 2 years after the procedure, which suggests postoperative neural reorganization plays an important role in recovery. Direct comparisons of each lesion approach within studies are extremely rare. Overall, long-term outcomes of these approaches have demonstrated significant therapeutic effects of each of these procedures. Reported response rates vary between 30% to 70%, when applied to remission, response (≥35% Y-BOCS reduction), and functional improvements in quality of life.50

 

 

Deep brain stimulation (DBS). With this approach, small electrodes are inserted under precise stereotactic MRI guidance. The advantage of DBS over ablative surgery is the ability to adjust and customize neurostimulation. Following implantation, modifiable parameters of electrode stimulation include electrode polarity, intensity, frequency, and laterality. A specially trained psychiatrist can conduct parameter optimization during long-term follow-up.

The first trial of DBS for OCD was reported in 1999 (N = 4), with the initial target selected based on the site of anterior capsulotomy. Three patients derived clinically observed benefit, although no validated questionnaires were administered.51 Since then, at least 7 studies with blinded stimulation have been conducted, totaling 62 patients.52

In recent years, structures adjacent to the internal capsule also have been targeted based on the approach employed in ventral capsulotomy. Across all trials, response rates for this approach consistently have been in the 50% range, with average Y-BOCS score reductions ranging from 6.8 to 31 points.53 Some patients have reported rapid improvements in anhedonia, and this approach is being employed in treatment-resistant depression.

Postoperative complications occur more often with DBS than with lesion approaches because of the prosthetic nature of the procedure (eg, increased risk of infection, lead malfunction, etc.). Additionally, batteries must be periodically explanted and replaced. Reported stimulation-related side effects include mood changes (transient sadness, anxiety, euphoria, and hypomania), sensory disturbances (olfactory, gustatory, and motor sensations), and cognitive changes (confusion and forgetfulness). These side effects typically are stimulation-dependent and disappear after altering stimulation parameters.

Table 2

Invasive therapies for treatment-resistant OCD

TherapyQuality of evidence
Reversible
  Electroconvulsive therapyPoor
  Vagal nerve stimulationPoor
  Repetitive transcranial magnetic stimulationLimited
Irreversible (surgical)
  Anterior capsulotomy. Target: anterior limb of the internal capsuleFair
  Anterior cingulotomy. Target: anterior cingulate and cingulum bundleFair
  Subcaudate tractotomy. Target: substantia innominata, just inferior to the caudate nucleusFair
  Limbic leucotomy. Target: anterior cingulotomy combined with subcaudate tractotomyFair
  Deep brain stimulation. Multiple targetsFair
OCD: obsessive-compulsive disorder

Related Resources

  • American Psychiatric Association. Treatment of patients with obsessive-compulsive disorder. www.psychiatryonline.com/pracGuide/pracGuideTopic_10.aspx.
  • Hyman BM, Pedrick C. The OCD workbook. Your guide to breaking free from obsessive compulsive disorder. 3rd ed. Oakland, CA: New Harbinger Publications Inc; 2010.
  • Baer L. Getting control: overcoming your obsessions and compulsions. Revised ed. New York, NY: Plume; 2000.

Drug Brand Names

  • Alprazolam • Xanax
  • Aripiprazole • Abilify
  • Citalopram • Celexa
  • Clomipramine • Anafranil
  • Clonazepam • Klonopin
  • Dextroamphetamine • Adderall
  • Duloxetine • Cymbalta
  • Fluoxetine • Prozac
  • Fluvoxamine • Luvox
  • Haloperidol • Haldol
  • Ketamine • Ketalar
  • Memantine • Namenda
  • Methylphenidate • Ritalin
  • Morphine • MS Contin
  • Olanzapine • Zyprexa
  • Ondansetron • Zofran
  • Pindolol • Visken
  • Quetiapine • Seroquel
  • Riluzole • Rilutek
  • Risperidone • Risperdal
  • Sertraline • Zoloft
  • Tramadol • Ultram
  • Venlafaxine • Effexor
  • Ziprasidone • Geodon

Disclosures

Drs. Khalsa and Schiffman report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Bystritsky receives grant support from AstraZeneca, Brainways, Takeda, and Transcept and is a founder, stockholder, and consultant for BrainSonix.

References

1. Pallanti S, Quercioli L. Treatment-refractory obsessive-compulsive disorder: methodological issues operational definitions and therapeutic lines. Prog Neuropsychopharmacol Biol Psychiatry. 2006;30(3):400-412.

2. Bystritsky A. Treatment-resistant anxiety disorders. Mol Psychiatry. 2006;11(9):805-814.

3. Denys D. Pharmacotherapy of obsessive-compulsive disorder and obsessive-compulsive spectrum disorders. Psychiatr Clin North Am. 2006;29(2):553-584 xi.

4. Ackerman DL, Greenland S. Multivariate meta-analysis of controlled drug studies for obsessive-compulsive disorder. J Clin Psychopharmacol. 2002;22(3):309-317.

5. Soomro GM, Altman D, Rajagopal S, et al. Selective serotonin re-uptake inhibitors (SSRIs) versus placebo for obsessive compulsive disorder (OCD). Cochrane Database Syst Rev. 2008;(1):CD001765.-

6. Bloch MH, McGuire J, Landeros-Weisenberger A, et al. Meta-analysis of the dose-response relationship of SSRI in obsessive-compulsive disorder. Mol Psychiatry. 2010;15(8):850-855.

7. Koran LM, Hanna GL, Hollander E, et al. American Psychiatric Association. Practice guideline for the treatment of patients with obsessive-compulsive disorder. Am J Psychiatry. 2007;164(7 suppl):5-53.

8. Ninan PT, Koran LM, Kiev A, et al. High-dose sertraline strategy for nonresponders to acute treatment for obsessive-compulsive disorder: a multicenter double-blind trial. J Clin Psychiatry. 2006;67(1):15-22.

9. Food and Drug Administration. FDA drug safety communication: abnormal heart rhythms associated with high doses of Celexa (citalopram hydrobromide). http://www.fda.gov/Drugs/DrugSafety/ucm269086.htm#sa. Published August 24 2011. Accessed September 27, 2011.

10. Yaryura-Tobias JA, Neziroglu FA. Venlafaxine in obsessive-compulsive disorder. Arch Gen Psychiatry. 1996;53(7):653-654.

11. Phelps NJ, Cates ME. The role of venlafaxine in the treatment of obsessive-compulsive disorder. Ann Pharmacother. 2005;39(1):136-140.

12. Dell’osso B, Mundo E, Marazziti D, et al. Switching from serotonin reuptake inhibitors to duloxetine in patients with resistant obsessive compulsive disorder: a case series. J Psychopharmacol. 2008;22(2):210-213.

13. Pallanti S, Quercioli L, Paiva RS, et al. Citalopram for treatment-resistant obsessive-compulsive disorder. Eur Psychiatry. 1999;14:101-106.

14. Ravizza L, Barzega G, Bellino S, et al. Therapeutic effect and safety of adjunctive risperidone in refractory obsessive-compulsive disorder (OCD). Psychopharmacol Bull. 1996;32:677-682.

15. Fallon BA, Liebowitz MR, Campeas R, et al. Intravenous clomipramine for obsessive-compulsive disorder refractory to oral clomipramine: a placebo-controlled study. Arch Gen Psychiatry. 1998;55:918-924.

16. Koran LM, Pallanti S, Paiva RS, et al. Pulse loading versus gradual dosing of intravenous clomipramine in obsessive-compulsive disorder. Eur Neuropsychopharmacol. 1998;8:121-126.

17. Dannon PN, Sasson Y, Hirschmann S, et al. Pindolol augmentation in treatment-resistant obsessive compulsive disorder: a double-blind placebo controlled trial. Eur Neuropsychopharmacol. 2000;10:165-169.

18. Koran LM, Mueller K, Maloney A. Will pindolol augment the response to a serotonin reuptake inhibitor in obsessive-compulsive disorder? J Clin Psychopharmacol. 1996;16:253-254.

19. Hewlett WA, Vinogradov S, Agras WS. Clomipramine clonazepam, and clonidine treatment of obsessive-compulsive disorder. J Clin Psychopharmacol. 1992;12:420-430.

20. Mundo E, Guglielmo E, Bellodi L. Effect of adjuvant pindolol on the antiobsessional response to fluvoxamine: a double-blind placebo-controlled study. Int Clin Psychopharmacol. 1998;13:219-224.

21. Hewlett WA, Schmid SP, Salomon RM. Pilot trial of ondansetron in the treatment of 8 patients with obsessive-compulsive disorder. J Clin Psychiatry. 2003;64:1025-1030.

22. Pallanti S, Bernardi S, Antonini S, et al. Ondansetron augmentation in treatment-resistant obsessive-compulsive disorder: a preliminary, single-blind, prospective study. CNS Drugs. 2009;23(12):1047-1055.

23. Connor KM, Payne VM, Gadde KM, et al. The use of aripiprazole in obsessive-compulsive disorder: preliminary observations in 8 patients. J Clin Psychiatry. 2005;66:49-51.

24. Komossa K, Depping AM, Meyer M, et al. Second-generation antipsychotics for obsessive compulsive disorder. Cochrane Database Syst Rev. 2010;(12):CD008141.-

25. Bloch MH, Landeros-Weisenberger A, Kelmendi B, et al. A systematic review: antipsychotic augmentation with treatment refractory obsessive-compulsive disorder. Mol Psychiatry. 2006;11(7):622-632.

26. Muscatello MR, Bruno A, Pandolfo G, et al. Effect of aripiprazole augmentation of serotonin reuptake inhibitors or clomipramine in treatment-resistant obsessive-compulsive disorder: a double-blind, placebo-controlled study. J Clin Psychopharmacol. 2011;31(2):174-179.

27. Savas HA, Yumru M, Ozen ME. Quetiapine and ziprasidone as adjuncts in treatment-resistant obsessive-compulsive disorder: a retrospective comparative study. Clin Drug Investig. 2008;28(7):439-442.

28. Hollander E, Kaplan A, Stahl SM. A double-blind placebo-controlled trial of clonazepam in obsessive-compulsive disorder. World J Biol Psychiatry. 2003;4:30-34.

29. Crockett BA, Churchill E, Davidson JR. A double-blind combination study of clonazepam with sertraline in obsessive-compulsive disorder. Ann Clin Psychiatry. 2004;16(3):127-132.

30. Koran LM, Aboujaoude E, Bullock KD, et al. Double-blind treatment with oral morphine in treatment-resistant obsessive-compulsive disorder. J Clin Psychiatry. 2005;66(3):353-359.

31. Shapira NA, Keck PE Jr, Goldsmith TD, et al. Open-label pilot study of tramadol hydrochloride in treatment-refractory obsessive-compulsive disorder. Depress Anxiety. 1997;6:170-173.

32. Goldsmith TB, Shapira NA, Keck PE Jr. Rapid remission of OCD with tramadol hydrochloride. Am J Psychiatry. 1999;156(4):660-661.

33. Insel TR, Hamilton JA, Guttmacher LB, et al. D-amphetamine in obsessive-compulsive disorder. Psychopharmacology (Berl). 1983;80:231-235.

34. Joffe RT, Swinson RP, Levitt AJ. Acute psychostimulant challenge in primary obsessive-compulsive disorder. J Clin Psychopharmacol. 1991;11:237-241.

35. Woolley JB, Heyman I. Dexamphetamine for obsessive-compulsive disorder. Am J Psychiatry. 2003;160:183.-

36. Aboujaoude E, Barry JJ, Gamel N. Memantine augmentation in treatment-resistant obsessive-compulsive disorder: an open-label trial. J Clin Psychopharmacol. 2009;29(1):51-55.

37. Coric V, Taskiran S, Pittenger C, et al. Riluzole augmentation in treatment-resistant obsessive-compulsive disorder: an open-label trial. Biol Psychiatry. 2005;58(5):424-428.

38. Rodriguez CI, Kegeles LS, Flood P, et al. Rapid resolution of obsessions after an infusion of intravenous ketamine in a patient with treatment-resistant obsessive-compulsive disorder. J Clin Psychiatry. 2011;72(4):567-569.

39. Leonard HL, Rapoport JL. Relief of obsessive-compulsive symptoms by LSD and psilocin. Am J Psychiatry. 1987;144(9):1239-1240.

40. Anand N, Sudhir PM, Math SB, et al. Cognitive behavior therapy in medication non-responders with obsessive-compulsive disorder: a prospective 1-year follow-up study. J Anxiety Disord. 2011;25(7):939-945.

41. Himle JA, Fischer DJ, Muroff JR, et al. Videoconferencing-based cognitive-behavioral therapy for obsessive-compulsive disorder. Behav Res Ther. 2006;44(12):1821-1829.

42. Kobak KA, Taylor LV, Bystritsky A, et al. St John’s wort versus placebo in obsessive-compulsive disorder: results from a double-blind study. Int Clin Psychopharmacol. 2005;20(6):299-304.

43. Sayyah M, Boostani H, Pakseresht S, et al. Comparison of Silybum marianum (L.) Gaertn. with fluoxetine in the treatment of obsessive-compulsive Disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34(2):362-365.

44. Sarris J, Camfield D, Berk M. Complementary medicine self-help, and lifestyle interventions for obsessive compulsive disorder (OCD) and the OCD spectrum: a systematic review. J Affect Disord. 2011 (epub ahead of print).

45. Beale MD, Kellner CH, Pritchett JT, et al. ECT for OCD. J Clin Psychiatry. 1995;56(2):81-82.

46. George MS, Ward HE Jr, Ninan PT, et al. A pilot study of vagus nerve stimulation (VNS) for treatment-resistant anxiety disorders. Brain Stimul. 2008;1(2):112-121.

47. Slotema CW, Blom JD, Hoek HW, et al. Should we expand the toolbox of psychiatric treatment methods to include repetitive transcranial magnetic stimulation (rTMS)? A meta-analysis of the efficacy of rTMS in psychiatric disorders. J Clin Psychiatry. 2010;71(7):873-884.

48. Mantovani A, Simpson HB, Fallon BA, et al. Randomized sham-controlled trial of repetitive transcranial magnetic stimulation in treatment-resistant obsessive-compulsive disorder. Int J Neuropsychopharmacol. 2010;13(2):217-227.

49. Blom RM, Figee M, Vulink N, et al. Update on repetitive transcranial magnetic stimulation in obsessive-compulsive disorder: different targets. Curr Psychiatry Rep. 2011;13(4):289-294.

50. Greenberg BD, Rauch SL, Haber SN. Invasive circuitry-based neurotherapeutics: stereotactic ablation and deep brain stimulation for OCD. Neuropsychopharmacology. 2010;35(1):317-336.

51. Nuttin B, Cosyns P, Demeulemeester H, et al. Electrical stimulation in anterior limbs of internal capsules in patients with obsessive-compulsive disorder. Lancet. 1999;354(9189):1526.-

52. de Koning PP, Figee M, van den Munckhof P, et al. Current status of deep brain stimulation for obsessive-compulsive disorder: a clinical review of different targets. Curr Psychiatry Rep. 2011;13(4):274-282.

53. Greenberg BD, Gabriels LA, Malone DA, Jr, et al. Deep brain stimulation of the ventral internal capsule/ventral striatum for obsessive-compulsive disorder: worldwide experience. Mol Psychiatry. 2010;15(1):64-79.

References

1. Pallanti S, Quercioli L. Treatment-refractory obsessive-compulsive disorder: methodological issues operational definitions and therapeutic lines. Prog Neuropsychopharmacol Biol Psychiatry. 2006;30(3):400-412.

2. Bystritsky A. Treatment-resistant anxiety disorders. Mol Psychiatry. 2006;11(9):805-814.

3. Denys D. Pharmacotherapy of obsessive-compulsive disorder and obsessive-compulsive spectrum disorders. Psychiatr Clin North Am. 2006;29(2):553-584 xi.

4. Ackerman DL, Greenland S. Multivariate meta-analysis of controlled drug studies for obsessive-compulsive disorder. J Clin Psychopharmacol. 2002;22(3):309-317.

5. Soomro GM, Altman D, Rajagopal S, et al. Selective serotonin re-uptake inhibitors (SSRIs) versus placebo for obsessive compulsive disorder (OCD). Cochrane Database Syst Rev. 2008;(1):CD001765.-

6. Bloch MH, McGuire J, Landeros-Weisenberger A, et al. Meta-analysis of the dose-response relationship of SSRI in obsessive-compulsive disorder. Mol Psychiatry. 2010;15(8):850-855.

7. Koran LM, Hanna GL, Hollander E, et al. American Psychiatric Association. Practice guideline for the treatment of patients with obsessive-compulsive disorder. Am J Psychiatry. 2007;164(7 suppl):5-53.

8. Ninan PT, Koran LM, Kiev A, et al. High-dose sertraline strategy for nonresponders to acute treatment for obsessive-compulsive disorder: a multicenter double-blind trial. J Clin Psychiatry. 2006;67(1):15-22.

9. Food and Drug Administration. FDA drug safety communication: abnormal heart rhythms associated with high doses of Celexa (citalopram hydrobromide). http://www.fda.gov/Drugs/DrugSafety/ucm269086.htm#sa. Published August 24 2011. Accessed September 27, 2011.

10. Yaryura-Tobias JA, Neziroglu FA. Venlafaxine in obsessive-compulsive disorder. Arch Gen Psychiatry. 1996;53(7):653-654.

11. Phelps NJ, Cates ME. The role of venlafaxine in the treatment of obsessive-compulsive disorder. Ann Pharmacother. 2005;39(1):136-140.

12. Dell’osso B, Mundo E, Marazziti D, et al. Switching from serotonin reuptake inhibitors to duloxetine in patients with resistant obsessive compulsive disorder: a case series. J Psychopharmacol. 2008;22(2):210-213.

13. Pallanti S, Quercioli L, Paiva RS, et al. Citalopram for treatment-resistant obsessive-compulsive disorder. Eur Psychiatry. 1999;14:101-106.

14. Ravizza L, Barzega G, Bellino S, et al. Therapeutic effect and safety of adjunctive risperidone in refractory obsessive-compulsive disorder (OCD). Psychopharmacol Bull. 1996;32:677-682.

15. Fallon BA, Liebowitz MR, Campeas R, et al. Intravenous clomipramine for obsessive-compulsive disorder refractory to oral clomipramine: a placebo-controlled study. Arch Gen Psychiatry. 1998;55:918-924.

16. Koran LM, Pallanti S, Paiva RS, et al. Pulse loading versus gradual dosing of intravenous clomipramine in obsessive-compulsive disorder. Eur Neuropsychopharmacol. 1998;8:121-126.

17. Dannon PN, Sasson Y, Hirschmann S, et al. Pindolol augmentation in treatment-resistant obsessive compulsive disorder: a double-blind placebo controlled trial. Eur Neuropsychopharmacol. 2000;10:165-169.

18. Koran LM, Mueller K, Maloney A. Will pindolol augment the response to a serotonin reuptake inhibitor in obsessive-compulsive disorder? J Clin Psychopharmacol. 1996;16:253-254.

19. Hewlett WA, Vinogradov S, Agras WS. Clomipramine clonazepam, and clonidine treatment of obsessive-compulsive disorder. J Clin Psychopharmacol. 1992;12:420-430.

20. Mundo E, Guglielmo E, Bellodi L. Effect of adjuvant pindolol on the antiobsessional response to fluvoxamine: a double-blind placebo-controlled study. Int Clin Psychopharmacol. 1998;13:219-224.

21. Hewlett WA, Schmid SP, Salomon RM. Pilot trial of ondansetron in the treatment of 8 patients with obsessive-compulsive disorder. J Clin Psychiatry. 2003;64:1025-1030.

22. Pallanti S, Bernardi S, Antonini S, et al. Ondansetron augmentation in treatment-resistant obsessive-compulsive disorder: a preliminary, single-blind, prospective study. CNS Drugs. 2009;23(12):1047-1055.

23. Connor KM, Payne VM, Gadde KM, et al. The use of aripiprazole in obsessive-compulsive disorder: preliminary observations in 8 patients. J Clin Psychiatry. 2005;66:49-51.

24. Komossa K, Depping AM, Meyer M, et al. Second-generation antipsychotics for obsessive compulsive disorder. Cochrane Database Syst Rev. 2010;(12):CD008141.-

25. Bloch MH, Landeros-Weisenberger A, Kelmendi B, et al. A systematic review: antipsychotic augmentation with treatment refractory obsessive-compulsive disorder. Mol Psychiatry. 2006;11(7):622-632.

26. Muscatello MR, Bruno A, Pandolfo G, et al. Effect of aripiprazole augmentation of serotonin reuptake inhibitors or clomipramine in treatment-resistant obsessive-compulsive disorder: a double-blind, placebo-controlled study. J Clin Psychopharmacol. 2011;31(2):174-179.

27. Savas HA, Yumru M, Ozen ME. Quetiapine and ziprasidone as adjuncts in treatment-resistant obsessive-compulsive disorder: a retrospective comparative study. Clin Drug Investig. 2008;28(7):439-442.

28. Hollander E, Kaplan A, Stahl SM. A double-blind placebo-controlled trial of clonazepam in obsessive-compulsive disorder. World J Biol Psychiatry. 2003;4:30-34.

29. Crockett BA, Churchill E, Davidson JR. A double-blind combination study of clonazepam with sertraline in obsessive-compulsive disorder. Ann Clin Psychiatry. 2004;16(3):127-132.

30. Koran LM, Aboujaoude E, Bullock KD, et al. Double-blind treatment with oral morphine in treatment-resistant obsessive-compulsive disorder. J Clin Psychiatry. 2005;66(3):353-359.

31. Shapira NA, Keck PE Jr, Goldsmith TD, et al. Open-label pilot study of tramadol hydrochloride in treatment-refractory obsessive-compulsive disorder. Depress Anxiety. 1997;6:170-173.

32. Goldsmith TB, Shapira NA, Keck PE Jr. Rapid remission of OCD with tramadol hydrochloride. Am J Psychiatry. 1999;156(4):660-661.

33. Insel TR, Hamilton JA, Guttmacher LB, et al. D-amphetamine in obsessive-compulsive disorder. Psychopharmacology (Berl). 1983;80:231-235.

34. Joffe RT, Swinson RP, Levitt AJ. Acute psychostimulant challenge in primary obsessive-compulsive disorder. J Clin Psychopharmacol. 1991;11:237-241.

35. Woolley JB, Heyman I. Dexamphetamine for obsessive-compulsive disorder. Am J Psychiatry. 2003;160:183.-

36. Aboujaoude E, Barry JJ, Gamel N. Memantine augmentation in treatment-resistant obsessive-compulsive disorder: an open-label trial. J Clin Psychopharmacol. 2009;29(1):51-55.

37. Coric V, Taskiran S, Pittenger C, et al. Riluzole augmentation in treatment-resistant obsessive-compulsive disorder: an open-label trial. Biol Psychiatry. 2005;58(5):424-428.

38. Rodriguez CI, Kegeles LS, Flood P, et al. Rapid resolution of obsessions after an infusion of intravenous ketamine in a patient with treatment-resistant obsessive-compulsive disorder. J Clin Psychiatry. 2011;72(4):567-569.

39. Leonard HL, Rapoport JL. Relief of obsessive-compulsive symptoms by LSD and psilocin. Am J Psychiatry. 1987;144(9):1239-1240.

40. Anand N, Sudhir PM, Math SB, et al. Cognitive behavior therapy in medication non-responders with obsessive-compulsive disorder: a prospective 1-year follow-up study. J Anxiety Disord. 2011;25(7):939-945.

41. Himle JA, Fischer DJ, Muroff JR, et al. Videoconferencing-based cognitive-behavioral therapy for obsessive-compulsive disorder. Behav Res Ther. 2006;44(12):1821-1829.

42. Kobak KA, Taylor LV, Bystritsky A, et al. St John’s wort versus placebo in obsessive-compulsive disorder: results from a double-blind study. Int Clin Psychopharmacol. 2005;20(6):299-304.

43. Sayyah M, Boostani H, Pakseresht S, et al. Comparison of Silybum marianum (L.) Gaertn. with fluoxetine in the treatment of obsessive-compulsive Disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34(2):362-365.

44. Sarris J, Camfield D, Berk M. Complementary medicine self-help, and lifestyle interventions for obsessive compulsive disorder (OCD) and the OCD spectrum: a systematic review. J Affect Disord. 2011 (epub ahead of print).

45. Beale MD, Kellner CH, Pritchett JT, et al. ECT for OCD. J Clin Psychiatry. 1995;56(2):81-82.

46. George MS, Ward HE Jr, Ninan PT, et al. A pilot study of vagus nerve stimulation (VNS) for treatment-resistant anxiety disorders. Brain Stimul. 2008;1(2):112-121.

47. Slotema CW, Blom JD, Hoek HW, et al. Should we expand the toolbox of psychiatric treatment methods to include repetitive transcranial magnetic stimulation (rTMS)? A meta-analysis of the efficacy of rTMS in psychiatric disorders. J Clin Psychiatry. 2010;71(7):873-884.

48. Mantovani A, Simpson HB, Fallon BA, et al. Randomized sham-controlled trial of repetitive transcranial magnetic stimulation in treatment-resistant obsessive-compulsive disorder. Int J Neuropsychopharmacol. 2010;13(2):217-227.

49. Blom RM, Figee M, Vulink N, et al. Update on repetitive transcranial magnetic stimulation in obsessive-compulsive disorder: different targets. Curr Psychiatry Rep. 2011;13(4):289-294.

50. Greenberg BD, Rauch SL, Haber SN. Invasive circuitry-based neurotherapeutics: stereotactic ablation and deep brain stimulation for OCD. Neuropsychopharmacology. 2010;35(1):317-336.

51. Nuttin B, Cosyns P, Demeulemeester H, et al. Electrical stimulation in anterior limbs of internal capsules in patients with obsessive-compulsive disorder. Lancet. 1999;354(9189):1526.-

52. de Koning PP, Figee M, van den Munckhof P, et al. Current status of deep brain stimulation for obsessive-compulsive disorder: a clinical review of different targets. Curr Psychiatry Rep. 2011;13(4):274-282.

53. Greenberg BD, Gabriels LA, Malone DA, Jr, et al. Deep brain stimulation of the ventral internal capsule/ventral striatum for obsessive-compulsive disorder: worldwide experience. Mol Psychiatry. 2010;15(1):64-79.

Issue
Current Psychiatry - 10(11)
Issue
Current Psychiatry - 10(11)
Page Number
44-52
Page Number
44-52
Publications
Publications
Topics
Article Type
Display Headline
Treatment-resistant OCD: Options beyond first-line medications
Display Headline
Treatment-resistant OCD: Options beyond first-line medications
Legacy Keywords
treatment-resistant OCD; OCD; obsessive-compulsive disorder; Sahib Khalsa; Jason Schiffman; Alexander Bystrirsky; first-line pharmacotherapy; augmentation; serotonergic agents; supratherapeutic SSRI; SNRIs; clomipramine; IV clomipramine; pindolol; ondansetron; antipsychotics; benzodiazepines; opioids; psychostimulants; NMDA antagonists; hallucinogens; psychotherapy; alternative medicine; invasive therapies; ECT; VNS; rTMS; surgical approaches; deep brain stimulation
Legacy Keywords
treatment-resistant OCD; OCD; obsessive-compulsive disorder; Sahib Khalsa; Jason Schiffman; Alexander Bystrirsky; first-line pharmacotherapy; augmentation; serotonergic agents; supratherapeutic SSRI; SNRIs; clomipramine; IV clomipramine; pindolol; ondansetron; antipsychotics; benzodiazepines; opioids; psychostimulants; NMDA antagonists; hallucinogens; psychotherapy; alternative medicine; invasive therapies; ECT; VNS; rTMS; surgical approaches; deep brain stimulation
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Hallucinations: Common features and causes

Article Type
Changed
Tue, 12/11/2018 - 15:04
Display Headline
Hallucinations: Common features and causes

Not all patients who experience hallucinations have a psychotic disorder. Many physical and psychiatric disorders can manifest with hallucinations, and some patients have >1 disorder that could cause different types of hallucinations. To avoid providing unnecessary or ineffective treatments—and to ensure that patients receive proper care for nonpsychiatric conditions—it is important to accurately diagnose the disorder causing a patient’s hallucinations.

In this article we describe common features and psychiatric and nonpsychiatric causes of auditory, visual, olfactory, gustatory, tactile, and somatic hallucinations. Awareness of typical presentations of hallucinations associated with specific disorders can help narrow the diagnosis and provide appropriate treatment.

Auditory hallucinations

Also known as paracusia, auditory hallucinations are perceptions of sounds without identifiable external stimuli. This type of hallucination has various causes (Table 1).1 A frequent symptom of schizophrenia, auditory hallucinations can cause substantial distress and functional disability.2 Approximately 60% to 90% of patients with schizophrenia and up to 80% of those with affective psychoses experience auditory hallucinations.1

Auditory hallucinations in psychosis usually are formed and complex.3 A common manifestation is hearing ≥1 voices. A patient might experience 2 voices talking about him in the third person. The voices may be perceived as coming from inside or outside the patient’s head. Some might hear their own thoughts spoken aloud. According to DSM-IV-TR, “hearing voices” is sufficient to diagnose schizophrenia if the hallucinations consist of a voice keeping up a running commentary on the person’s behavior or ≥2 voices conversing with each other.4 Auditory hallucinations also are seen in mood disorders but tend to be milder than their psychosis-induced counterparts.

Simple (unformed) auditory hallucinations—referred to as tinnitus—can be caused by disease of the middle ear (otosclerosis) or inner ear. These unformed hallucinations consist of buzzing or tones of varying pitch and timbre.1

Partial seizures may cause auditory hallucinations. Perceptions of music have been associated with partial seizures.5 Curie and colleagues found that 17% of 514 patients with temporal lobe epilepsy had auditory hallucinations as a component of their seizures.6 These hallucinations typically are brief, stereotyped sensory impressions and, if formed, may be trivial sentences, previously heard phrases, or commands.

Alcoholic hallucinosis is a hallucinatory syndrome caused by alcohol withdrawal. These hallucinations usually are vocal and typically consist of accusatory, threatening, and/or critical voices directed at the patient.1 Patients with alcohol hallucinosis also may experience musical auditory hallucinations.7,8

CNS neoplasms can produce auditory hallucinations in 3% to 10% of patients.9 Hemorrhages and arteriovenous malformations in the pontine tegmentum and lower midbrain have been associated with acute onset of auditory hallucinations. The sounds typically are unformed mechanical or seashell-like noises or music.10

Patients with migraines rarely report auditory hallucinations. When they occur, they typically consist of perceived unilateral tinnitus, phonophobia, or hearing loss.

Table 1

Common causes of auditory hallucinations

Peripheral lesions
Middle ear disease
Inner ear disease
Auditory nerve disease
CNS disorders
Temporal lobe epilepsy
Pontine lesions
Stroke
Arteriovenous malformations
Syncope
Toxic metabolic disturbances
Alcoholic hallucinosis
Delirium
Hallucinogens
Schizophrenia
Mania
Psychotic depression
Dissociative identity disorder
Posttraumatic stress disorder
Source: Reference 1

Visual hallucinations

Visual hallucinations manifest as visual sensory perceptions in the absence of external stimuli.11 These false perceptions may consist of formed images (eg, people) or unformed images (eg, flashes of light).12 Visual hallucinations occur in numerous ophthalmologic, neurologic, medical, and psychiatric disorders (Table 2).13

DSM-IV-TR lists visual hallucinations as a primary diagnostic criterion for several psychotic disorders, including schizophrenia and schizoaffective disorder,4 and they occur in 16% to 72% of patients with these conditions.14,15 Patients with major depressive disorder or bipolar disorder also may experience visual hallucinations. Visual hallucinations in those with schizophrenia tend to involve vivid scenes with family members, religious figures, and/or animals.16

Delirium is a transient, reversible cause of cerebral dysfunction that often presents with hallucinations. Several studies have shown that visual hallucinations are the most common type among patients with delirium. Webster and Holroyd found visual hallucinations in 27% of 227 delirium patients.17

Delirium tremens typically is accompanied by visual hallucinations. Visions of small animals and crawling insects are common.18 Hallucinations due to drug intoxication or withdrawal generally vary in duration from brief to continuous; such experiences often contribute to agitation.19

Migraines are a well-recognized cause of visual hallucinations. Up to 31% of those with migraines experience an aura, and nearly 99% of those with aura have visual symptoms.20,21 The classic visual aura starts as an irregular colored crescent of light with multi-colored edges in the center of the visual field that gradually progresses toward the periphery, lasting <60 minutes. These simple visual hallucinations are most common; more complex hallucinations are seen more frequently in migraine coma and familial hemiplegic migraine.

 

 

Approximately 5% of patients with epilepsy have occipital seizures, which almost always have visual manifestations. Epileptic visual hallucinations often are simple, brief, stereotyped, and fragmentary. They usually consist of small, brightly colored spots or shapes that flash.22 Complex visual hallucinations in epilepsy are similar to hypnagogic hallucinations but are rare. Intracranial electroencephalography recordings have shown that pathological excitation of visual cortical areas may be responsible for complex visual hallucinations in epilepsy.19

Dementia with Lewy bodies (DLB) is associated with visual hallucinations.23 Visual hallucinations occur in >20% of patients with DLB.24 Patients with DLB may see complex scenarios of people and items that are not present. Visual hallucinations have an 83% positive predictive value for distinguishing DLB from dementia of the Alzheimer’s type.25 There is a strong correlation between Lewy bodies located in the amygdala and parahippocampus and well-formed visual hallucinations.26

Visual hallucinations are common in Parkinson’s disease and may occur in up to one-half of patients.27 Patients with Parkinson’s disease may experience hallucinations similar to those observed in DLB, which can range from seeing a person or animal to more complex, formed, and mobile people, animals, or objects.

Table 2

Common causes of visual hallucinations

Neurologic disorders
Migraine
Epilepsy
Hemispheric lesions
Optic nerve disorders
Brain stem lesions (peduncular hallucinosis)
Narcolepsy
Ophthalmologic diseases
Glaucoma
Retinal disease
Enucleation
Cataract formation
Choroidal disorder
Macular abnormalities
Toxic and metabolic conditions
Toxic-metabolic encephalopathy
Drug and alcohol withdrawal syndromes
Hallucinogens
Schizophrenia
Affective disorders
Conversion disorders
Sensory deprivation
Sleep deprivation
Hypnosis
Intense emotional experiences
Source: Reference 13

Olfactory hallucinations

Also known as phantosmia, olfactory hallucinations involve smelling odors that are not derived from any physical stimulus. They can occur with several psychiatric conditions, including schizophrenia, depression, bipolar disorder, eating disorders, and substance abuse.28 Olfactory hallucinations caused by epileptic activity are rare. They constitute approximately 0.9% of all auras and typically are described as unpleasant. Tumors that affect the medial temporal lobe and mesial temporal sclerosis are associated with olfactory hallucinations.29 Olfactory hallucinations also have been reported in patients with multi-infarct dementia, Alzheimer’s disease, and alcoholic psychosyndromes. In patients with schizophrenia, the smell may be perceived as coming from an external source, whereas patients with depression may perceive the source as internal.30 Patients who perceive that they are the source of an offensive odor—a condition known as olfactory reference syndrome—may wash excessively, overuse deodorants and perfumes, or become socially withdrawn.30

Gustatory hallucinations

Patients with gustatory hallucinations may experience salivation, sensation of thirst, or taste alterations. These hallucinations can be observed when the sylvian fissure that extends to the insula is stimulated electrically.31 Similar to olfactory hallucinations, gustatory hallucinations are associated with temporal lobe disease and parietal operculum lesions.31,32 Sinus diseases have been associated with olfactory and gustatory hallucinations.33 Brief gustatory hallucinations can be elicited with stimulation of the right rolandic operculum, parietal operculum, amygdala, hippocampus, medial temporal gyrus, and anterior part of right temporal gyrus.34

Tactile hallucinations

These hallucinations may include perceptions of insects crawling over or under the skin (formication) or simulation of pressure on skin.35 They have been associated with substance abuse, toxicity, or withdrawal.28 Tactile hallucinations are characteristic of cocaine or amphetamine intoxication.35

Tactile hallucinations are a rare symptom of schizophrenia. Heveling and colleagues reported a case of a woman, age 68, with chronic schizophrenia who experienced touching and being touched by a “shadow man” several times a day in addition to auditory and visual hallucinations.36 Her symptoms disappeared after 4 weeks of antipsychotic and mood stabilizer therapy.

Tactile hallucinations have been associated with obsessive-compulsive disorder (OCD).37 Fontenelle and colleagues suggested that OCD and psychotic disorders may share dysfunctional dopaminergic circuits.37

Somatic hallucinations

Patients who have somatic hallucinations report perceptions of abnormal body sensations or physical experiences. For example, a patient may have sense of not having a stomach while eating.35

This type of hallucination has been associated with activation of postcentral gyrus, parietal operculum, insula, and inferior parietal lobule on stereoelectroencephalography.34 In a study of cerebral blood flow in 20 geriatric patients with delusional disorder, somatic type who were experiencing somatic hallucinations, positron emission testing scan demonstrated increased perfusion in somatic sensory processing regions, particularly the left postcentral gyrus and the right paracentral lobule.38 Other researchers have linked somatic hallucinations with activation in the primary somatosensory and posterior parietal cortex, areas that normally mediate tactile perception.39

Related Resource

  • Teeple RC, Caplan JP, Stern TA. Visual hallucinations: differential diagnosis and treatment. Prim Care Companion J Clin Psychiatry. 2009;11(1):26-32.
 

 

Disclosures

Drs. Ali, Patel, Avenido, Bailey, and Jabeen report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Riley is on the board of directors for Vertex Pharmaceuticals.

Acknowledgment

The authors would like to thank Marwah Shahid, Research Associate, Vanderbilt University, Nashville, TN.

References

1. Cummings JL, Mega MS. Hallucinations. In: Cummings JL Mega MS, eds. Neuropsychiatry and behavioral neuroscience. New York, NY: Oxford University Press; 2003: 187–199.

2. Shergill SS, Murray RM, McGuire PK. Auditory hallucinations: a review of psychological treatments. Schizophr Res. 1998;32(3):137-150.

3. Goodwin DW, Alderson P, Rosenthal R. Clinical significance of hallucinations in psychiatric disorders. A study of 116 hallucinatory patients. Arch Gen Psychiatry. 1971;24(1):76-80.

4. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.

5. Kasper BS, Kasper EM, Pauli E, et al. Phenomenology of hallucinations, illusions, and delusions as part of seizure semiology. Epilepsy Behav. 2010;18(1-2):13-23.

6. Currie S, Heathfield KW, Henson RA, et al. Clinical course and prognosis of temporal lobe epilepsy. A survey of 666 patients. Brain. 1971;94(1):173-190.

7. Keshavan MS, David AS, Steingard S, et al. Musical hallucinations: a review and synthesis. Cogn Behav Neurol. 1992;5(3):211-223.

8. Duncan R, Mitchell JD, Critchley EMR. Hallucinations and music. Behav Neurol. 1989;2(2):115-124.

9. Tarachow S. The clinical value of hallucinations in localizing brain tumors. Am J Psychiatry. 1941;97:1434-1442.

10. Lanska DJ, Lanska MJ, Mendez MF. Brainstem auditory hallucinosis. Neurology. 1987;37(10):1685.-

11. Norton JW, Corbett JJ. Visual perceptual abnormalities: hallucinations and illusions. Semin Neurol. 2000;20(1):111-121.

12. Kaplan HI, Sadock BJ, Grebb JA. Typical signs and symptoms of psychiatric illness defined. In: Kaplan HI Sadock BJ, Grebb JA, eds. Kaplan and Sadock’s synopsis of psychiatry: behavioral sciences, clinical psychiatry. Baltimore, MD: Williams and Wilkins; 1994:300.

13. Cummings JL, Miller BL. Visual hallucinations. Clinical occurrence and use in differential diagnosis. West J Med. 1987;146(1):46-51.

14. First MB, Tasman A. Schizophrenia and other psychoses. In: First MB Tasman A, eds. Clinical guide to the diagnosis and treatment of mental disorders. San Francisco, CA: John Wiley and Sons; 2009:245–278.

15. Mueser KT, Bellack AS, Brady EU. Hallucinations in schizophrenia. Acta Psychiatr Scand. 1990;82(1):26-29.

16. Small IF, Small JG, Andersen JM. Clinical characteristics of hallucinations of schizophrenia. Dis Nerv Syst. 1966;27(5):349-353.

17. Webster R, Holroyd S. Prevalence of psychotic symptoms in delirium. Psychosomatics. 2000;41(6):519-522.

18. Gastfriend DR, Renner JA, Hackett TP. Alcoholic patients: acute and chronic. In: Stern TA Fricchione G, Cassem NH, et al, eds. Massachusetts General Hospital handbook of general hospital psychiatry. 5th ed. Philadelphia, PA: Mosby; 2004:203–216.

19. Manford M, Andermann F. Complex visual hallucinations. Clinical and neurobiological insights. Brain. 1998;121(Pt 10):1819-1840.

20. Goadsby PJ, Lipton RB, Ferrari MD. Migraine—current understanding and treatment. N Engl J Med. 2002;346(4):257-270.

21. Russell MB, Olesen J. A nosographic analysis of the migraine aura in a general population. Brain. 1996;119(Pt 2):355-361.

22. Panayiotopoulos CP. Elementary visual hallucinations blindness, and headache in idiopathic occipital epilepsy: differentiation from migraine. J Neurol Neurosurg Psychiatry. 1999;66(4):536-540.

23. Ballard CG, O’Brien JT, Swann AG, et al. The natural history of psychosis and depression in dementia with Lewy bodies and Alzheimer’s disease: persistence and new cases over 1 year of follow-up. J Clin Psychiatry. 2001;62(1):46-49.

24. Ala TA, Yang KH, Sung JH, et al. Hallucinations and signs of parkinsonism help distinguish patients with dementia and cortical Lewy bodies from patients with Alzheimer’s disease at presentation: a clinicopathological study. J Neurol Neurosurg Psychiatry. 1997;62(1):16-21.

25. Tiraboschi P, Salmon DP, Hansen LA, et al. What best differentiates Lewy body from Alzheimer’s disease in early-stage dementia? Brain. 2006;129(Pt 3):729-735.

26. Harding AJ, Broe GA, Halliday GM. Visual hallucinations in Lewy body disease relate to Lewy bodies in the temporal lobe. Brain. 2002;125(Pt 2):391-403.

27. Williams DR, Lees AJ. Visual hallucinations in the diagnosis of idiopathic Parkinson’s disease: a retrospective autopsy study. Lancet Neurol. 2005;4(10):605-610.

28. Lewandowski KE, DePaola J, Camsari GB, et al. Tactile, olfactory, and gustatory hallucinations in psychotic disorders: a descriptive study. Ann Acad Med Singapore. 2009;38(5):383-385.

29. Acharya V, Acharya J, Lüders H. Olfactory epileptic auras. Neurology. 1998;51(1):56-61.

30. Ropper AH, Samuels MA. Disorders of smell and taste. In: Ropper AH Samuels MA, eds. Adams and Victor’s principles of neurology. 9th ed. New York, NY: McGraw-Hill Companies; 2009:216–224.

31. Ropper AH, Samuels MA. Epilepsy and other seizure disorders. In: Ropper AH Samuels MA, eds. Adams and Victor’s principles of neurology. 9th ed. New York, NY: McGraw-Hill Companies; 2009:304–338.

32. Capampangan DJ, Hoerth MT, Drazkowski JF, et al. Olfactory and gustatory hallucinations presenting as partial status epilepticus because of glioblastoma multiforme. Ann Emerg Med. 2010;56(4):374-377.

33. Frasnelli J, Reden J, Landis BN, et al. Comment on “Olfactory hallucinations as a manifestation of hidden rhinosinusitis”. J Clin Neurosci. 2010;17(4):543.-

34. Elliott B, Joyce E, Shorvon S. Delusions illusions and hallucinations in epilepsy: 1. Elementary phenomena. Epilepsy Res. 2009;85(2-3):162-171.

35. Nurcombe B, Ebert MH. The psychiatric interview. In: Ebert MH Nurcombe B, Loosen PT, et al, eds. Current diagnosis and treatment: psychiatry. 2nd ed. New York, NY: McGraw-Hill Companies; 2008:95–114.

36. Heveling T, Emrich HM, Dietrich DE. Treatment of a rare psychopathological phenomenon: tactile hallucinations and the delusional other. Eur Psychiatry. 2004;19(6):387-388.

37. Fontenelle LF, Lopes AP, Borges MC, et al. Auditory, visual, tactile, olfactory, and bodily hallucinations in patients with obsessive-compulsive disorder. CNS Spectr. 2008;13(2):125-130.

38. Nemoto K, Mizukami K, Hori T, et al. Hyperperfusion in primary somatosensory region related to somatic hallucination in the elderly. Psychiatry Clin Neurosci. 2010;64(4):421-425.

39. Shergill SS, Cameron LA, Brammer MJ, et al. Modality specific neural correlates of auditory and somatic hallucinations. J Neurol Neurosurg Psychiatry. 2001;71(5):688-690.

Article PDF
Author and Disclosure Information

Shahid Ali, MD
Assistant Professor, Clinical Psychiatry, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN
Milapkumar Patel, MD
Research Associate, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN
Jaymie Avenido, MD
Research/Forensic Psychiatry Associate, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN
Rahn K. Bailey, MD, FAPA
Associate Professor, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN
Shagufta Jabeen, MD
Assistant Professor, Clinical Psychiatry, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN
Wayne J. Riley, MD, MPH, MBA, MACP
Professor of Family Medicine, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN

Issue
Current Psychiatry - 10(11)
Publications
Topics
Page Number
22-29
Legacy Keywords
hallucinations; features; causes; Shahid Ali; Milapkumar Patel; Jaymie Avenido; Rahn Bailey; Shagufta Jabeen; Wayne Riley; auditory hallucinations; visual hallucinations; olfactory hallucinations; gustatory hallucinations; tactile hallucinations; somatic hallucinations
Sections
Author and Disclosure Information

Shahid Ali, MD
Assistant Professor, Clinical Psychiatry, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN
Milapkumar Patel, MD
Research Associate, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN
Jaymie Avenido, MD
Research/Forensic Psychiatry Associate, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN
Rahn K. Bailey, MD, FAPA
Associate Professor, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN
Shagufta Jabeen, MD
Assistant Professor, Clinical Psychiatry, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN
Wayne J. Riley, MD, MPH, MBA, MACP
Professor of Family Medicine, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN

Author and Disclosure Information

Shahid Ali, MD
Assistant Professor, Clinical Psychiatry, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN
Milapkumar Patel, MD
Research Associate, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN
Jaymie Avenido, MD
Research/Forensic Psychiatry Associate, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN
Rahn K. Bailey, MD, FAPA
Associate Professor, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN
Shagufta Jabeen, MD
Assistant Professor, Clinical Psychiatry, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN
Wayne J. Riley, MD, MPH, MBA, MACP
Professor of Family Medicine, Department of Psychiatry and Behavioral Sciences, Meharry Medical College, Nashville, TN

Article PDF
Article PDF

Not all patients who experience hallucinations have a psychotic disorder. Many physical and psychiatric disorders can manifest with hallucinations, and some patients have >1 disorder that could cause different types of hallucinations. To avoid providing unnecessary or ineffective treatments—and to ensure that patients receive proper care for nonpsychiatric conditions—it is important to accurately diagnose the disorder causing a patient’s hallucinations.

In this article we describe common features and psychiatric and nonpsychiatric causes of auditory, visual, olfactory, gustatory, tactile, and somatic hallucinations. Awareness of typical presentations of hallucinations associated with specific disorders can help narrow the diagnosis and provide appropriate treatment.

Auditory hallucinations

Also known as paracusia, auditory hallucinations are perceptions of sounds without identifiable external stimuli. This type of hallucination has various causes (Table 1).1 A frequent symptom of schizophrenia, auditory hallucinations can cause substantial distress and functional disability.2 Approximately 60% to 90% of patients with schizophrenia and up to 80% of those with affective psychoses experience auditory hallucinations.1

Auditory hallucinations in psychosis usually are formed and complex.3 A common manifestation is hearing ≥1 voices. A patient might experience 2 voices talking about him in the third person. The voices may be perceived as coming from inside or outside the patient’s head. Some might hear their own thoughts spoken aloud. According to DSM-IV-TR, “hearing voices” is sufficient to diagnose schizophrenia if the hallucinations consist of a voice keeping up a running commentary on the person’s behavior or ≥2 voices conversing with each other.4 Auditory hallucinations also are seen in mood disorders but tend to be milder than their psychosis-induced counterparts.

Simple (unformed) auditory hallucinations—referred to as tinnitus—can be caused by disease of the middle ear (otosclerosis) or inner ear. These unformed hallucinations consist of buzzing or tones of varying pitch and timbre.1

Partial seizures may cause auditory hallucinations. Perceptions of music have been associated with partial seizures.5 Curie and colleagues found that 17% of 514 patients with temporal lobe epilepsy had auditory hallucinations as a component of their seizures.6 These hallucinations typically are brief, stereotyped sensory impressions and, if formed, may be trivial sentences, previously heard phrases, or commands.

Alcoholic hallucinosis is a hallucinatory syndrome caused by alcohol withdrawal. These hallucinations usually are vocal and typically consist of accusatory, threatening, and/or critical voices directed at the patient.1 Patients with alcohol hallucinosis also may experience musical auditory hallucinations.7,8

CNS neoplasms can produce auditory hallucinations in 3% to 10% of patients.9 Hemorrhages and arteriovenous malformations in the pontine tegmentum and lower midbrain have been associated with acute onset of auditory hallucinations. The sounds typically are unformed mechanical or seashell-like noises or music.10

Patients with migraines rarely report auditory hallucinations. When they occur, they typically consist of perceived unilateral tinnitus, phonophobia, or hearing loss.

Table 1

Common causes of auditory hallucinations

Peripheral lesions
Middle ear disease
Inner ear disease
Auditory nerve disease
CNS disorders
Temporal lobe epilepsy
Pontine lesions
Stroke
Arteriovenous malformations
Syncope
Toxic metabolic disturbances
Alcoholic hallucinosis
Delirium
Hallucinogens
Schizophrenia
Mania
Psychotic depression
Dissociative identity disorder
Posttraumatic stress disorder
Source: Reference 1

Visual hallucinations

Visual hallucinations manifest as visual sensory perceptions in the absence of external stimuli.11 These false perceptions may consist of formed images (eg, people) or unformed images (eg, flashes of light).12 Visual hallucinations occur in numerous ophthalmologic, neurologic, medical, and psychiatric disorders (Table 2).13

DSM-IV-TR lists visual hallucinations as a primary diagnostic criterion for several psychotic disorders, including schizophrenia and schizoaffective disorder,4 and they occur in 16% to 72% of patients with these conditions.14,15 Patients with major depressive disorder or bipolar disorder also may experience visual hallucinations. Visual hallucinations in those with schizophrenia tend to involve vivid scenes with family members, religious figures, and/or animals.16

Delirium is a transient, reversible cause of cerebral dysfunction that often presents with hallucinations. Several studies have shown that visual hallucinations are the most common type among patients with delirium. Webster and Holroyd found visual hallucinations in 27% of 227 delirium patients.17

Delirium tremens typically is accompanied by visual hallucinations. Visions of small animals and crawling insects are common.18 Hallucinations due to drug intoxication or withdrawal generally vary in duration from brief to continuous; such experiences often contribute to agitation.19

Migraines are a well-recognized cause of visual hallucinations. Up to 31% of those with migraines experience an aura, and nearly 99% of those with aura have visual symptoms.20,21 The classic visual aura starts as an irregular colored crescent of light with multi-colored edges in the center of the visual field that gradually progresses toward the periphery, lasting <60 minutes. These simple visual hallucinations are most common; more complex hallucinations are seen more frequently in migraine coma and familial hemiplegic migraine.

 

 

Approximately 5% of patients with epilepsy have occipital seizures, which almost always have visual manifestations. Epileptic visual hallucinations often are simple, brief, stereotyped, and fragmentary. They usually consist of small, brightly colored spots or shapes that flash.22 Complex visual hallucinations in epilepsy are similar to hypnagogic hallucinations but are rare. Intracranial electroencephalography recordings have shown that pathological excitation of visual cortical areas may be responsible for complex visual hallucinations in epilepsy.19

Dementia with Lewy bodies (DLB) is associated with visual hallucinations.23 Visual hallucinations occur in >20% of patients with DLB.24 Patients with DLB may see complex scenarios of people and items that are not present. Visual hallucinations have an 83% positive predictive value for distinguishing DLB from dementia of the Alzheimer’s type.25 There is a strong correlation between Lewy bodies located in the amygdala and parahippocampus and well-formed visual hallucinations.26

Visual hallucinations are common in Parkinson’s disease and may occur in up to one-half of patients.27 Patients with Parkinson’s disease may experience hallucinations similar to those observed in DLB, which can range from seeing a person or animal to more complex, formed, and mobile people, animals, or objects.

Table 2

Common causes of visual hallucinations

Neurologic disorders
Migraine
Epilepsy
Hemispheric lesions
Optic nerve disorders
Brain stem lesions (peduncular hallucinosis)
Narcolepsy
Ophthalmologic diseases
Glaucoma
Retinal disease
Enucleation
Cataract formation
Choroidal disorder
Macular abnormalities
Toxic and metabolic conditions
Toxic-metabolic encephalopathy
Drug and alcohol withdrawal syndromes
Hallucinogens
Schizophrenia
Affective disorders
Conversion disorders
Sensory deprivation
Sleep deprivation
Hypnosis
Intense emotional experiences
Source: Reference 13

Olfactory hallucinations

Also known as phantosmia, olfactory hallucinations involve smelling odors that are not derived from any physical stimulus. They can occur with several psychiatric conditions, including schizophrenia, depression, bipolar disorder, eating disorders, and substance abuse.28 Olfactory hallucinations caused by epileptic activity are rare. They constitute approximately 0.9% of all auras and typically are described as unpleasant. Tumors that affect the medial temporal lobe and mesial temporal sclerosis are associated with olfactory hallucinations.29 Olfactory hallucinations also have been reported in patients with multi-infarct dementia, Alzheimer’s disease, and alcoholic psychosyndromes. In patients with schizophrenia, the smell may be perceived as coming from an external source, whereas patients with depression may perceive the source as internal.30 Patients who perceive that they are the source of an offensive odor—a condition known as olfactory reference syndrome—may wash excessively, overuse deodorants and perfumes, or become socially withdrawn.30

Gustatory hallucinations

Patients with gustatory hallucinations may experience salivation, sensation of thirst, or taste alterations. These hallucinations can be observed when the sylvian fissure that extends to the insula is stimulated electrically.31 Similar to olfactory hallucinations, gustatory hallucinations are associated with temporal lobe disease and parietal operculum lesions.31,32 Sinus diseases have been associated with olfactory and gustatory hallucinations.33 Brief gustatory hallucinations can be elicited with stimulation of the right rolandic operculum, parietal operculum, amygdala, hippocampus, medial temporal gyrus, and anterior part of right temporal gyrus.34

Tactile hallucinations

These hallucinations may include perceptions of insects crawling over or under the skin (formication) or simulation of pressure on skin.35 They have been associated with substance abuse, toxicity, or withdrawal.28 Tactile hallucinations are characteristic of cocaine or amphetamine intoxication.35

Tactile hallucinations are a rare symptom of schizophrenia. Heveling and colleagues reported a case of a woman, age 68, with chronic schizophrenia who experienced touching and being touched by a “shadow man” several times a day in addition to auditory and visual hallucinations.36 Her symptoms disappeared after 4 weeks of antipsychotic and mood stabilizer therapy.

Tactile hallucinations have been associated with obsessive-compulsive disorder (OCD).37 Fontenelle and colleagues suggested that OCD and psychotic disorders may share dysfunctional dopaminergic circuits.37

Somatic hallucinations

Patients who have somatic hallucinations report perceptions of abnormal body sensations or physical experiences. For example, a patient may have sense of not having a stomach while eating.35

This type of hallucination has been associated with activation of postcentral gyrus, parietal operculum, insula, and inferior parietal lobule on stereoelectroencephalography.34 In a study of cerebral blood flow in 20 geriatric patients with delusional disorder, somatic type who were experiencing somatic hallucinations, positron emission testing scan demonstrated increased perfusion in somatic sensory processing regions, particularly the left postcentral gyrus and the right paracentral lobule.38 Other researchers have linked somatic hallucinations with activation in the primary somatosensory and posterior parietal cortex, areas that normally mediate tactile perception.39

Related Resource

  • Teeple RC, Caplan JP, Stern TA. Visual hallucinations: differential diagnosis and treatment. Prim Care Companion J Clin Psychiatry. 2009;11(1):26-32.
 

 

Disclosures

Drs. Ali, Patel, Avenido, Bailey, and Jabeen report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Riley is on the board of directors for Vertex Pharmaceuticals.

Acknowledgment

The authors would like to thank Marwah Shahid, Research Associate, Vanderbilt University, Nashville, TN.

Not all patients who experience hallucinations have a psychotic disorder. Many physical and psychiatric disorders can manifest with hallucinations, and some patients have >1 disorder that could cause different types of hallucinations. To avoid providing unnecessary or ineffective treatments—and to ensure that patients receive proper care for nonpsychiatric conditions—it is important to accurately diagnose the disorder causing a patient’s hallucinations.

In this article we describe common features and psychiatric and nonpsychiatric causes of auditory, visual, olfactory, gustatory, tactile, and somatic hallucinations. Awareness of typical presentations of hallucinations associated with specific disorders can help narrow the diagnosis and provide appropriate treatment.

Auditory hallucinations

Also known as paracusia, auditory hallucinations are perceptions of sounds without identifiable external stimuli. This type of hallucination has various causes (Table 1).1 A frequent symptom of schizophrenia, auditory hallucinations can cause substantial distress and functional disability.2 Approximately 60% to 90% of patients with schizophrenia and up to 80% of those with affective psychoses experience auditory hallucinations.1

Auditory hallucinations in psychosis usually are formed and complex.3 A common manifestation is hearing ≥1 voices. A patient might experience 2 voices talking about him in the third person. The voices may be perceived as coming from inside or outside the patient’s head. Some might hear their own thoughts spoken aloud. According to DSM-IV-TR, “hearing voices” is sufficient to diagnose schizophrenia if the hallucinations consist of a voice keeping up a running commentary on the person’s behavior or ≥2 voices conversing with each other.4 Auditory hallucinations also are seen in mood disorders but tend to be milder than their psychosis-induced counterparts.

Simple (unformed) auditory hallucinations—referred to as tinnitus—can be caused by disease of the middle ear (otosclerosis) or inner ear. These unformed hallucinations consist of buzzing or tones of varying pitch and timbre.1

Partial seizures may cause auditory hallucinations. Perceptions of music have been associated with partial seizures.5 Curie and colleagues found that 17% of 514 patients with temporal lobe epilepsy had auditory hallucinations as a component of their seizures.6 These hallucinations typically are brief, stereotyped sensory impressions and, if formed, may be trivial sentences, previously heard phrases, or commands.

Alcoholic hallucinosis is a hallucinatory syndrome caused by alcohol withdrawal. These hallucinations usually are vocal and typically consist of accusatory, threatening, and/or critical voices directed at the patient.1 Patients with alcohol hallucinosis also may experience musical auditory hallucinations.7,8

CNS neoplasms can produce auditory hallucinations in 3% to 10% of patients.9 Hemorrhages and arteriovenous malformations in the pontine tegmentum and lower midbrain have been associated with acute onset of auditory hallucinations. The sounds typically are unformed mechanical or seashell-like noises or music.10

Patients with migraines rarely report auditory hallucinations. When they occur, they typically consist of perceived unilateral tinnitus, phonophobia, or hearing loss.

Table 1

Common causes of auditory hallucinations

Peripheral lesions
Middle ear disease
Inner ear disease
Auditory nerve disease
CNS disorders
Temporal lobe epilepsy
Pontine lesions
Stroke
Arteriovenous malformations
Syncope
Toxic metabolic disturbances
Alcoholic hallucinosis
Delirium
Hallucinogens
Schizophrenia
Mania
Psychotic depression
Dissociative identity disorder
Posttraumatic stress disorder
Source: Reference 1

Visual hallucinations

Visual hallucinations manifest as visual sensory perceptions in the absence of external stimuli.11 These false perceptions may consist of formed images (eg, people) or unformed images (eg, flashes of light).12 Visual hallucinations occur in numerous ophthalmologic, neurologic, medical, and psychiatric disorders (Table 2).13

DSM-IV-TR lists visual hallucinations as a primary diagnostic criterion for several psychotic disorders, including schizophrenia and schizoaffective disorder,4 and they occur in 16% to 72% of patients with these conditions.14,15 Patients with major depressive disorder or bipolar disorder also may experience visual hallucinations. Visual hallucinations in those with schizophrenia tend to involve vivid scenes with family members, religious figures, and/or animals.16

Delirium is a transient, reversible cause of cerebral dysfunction that often presents with hallucinations. Several studies have shown that visual hallucinations are the most common type among patients with delirium. Webster and Holroyd found visual hallucinations in 27% of 227 delirium patients.17

Delirium tremens typically is accompanied by visual hallucinations. Visions of small animals and crawling insects are common.18 Hallucinations due to drug intoxication or withdrawal generally vary in duration from brief to continuous; such experiences often contribute to agitation.19

Migraines are a well-recognized cause of visual hallucinations. Up to 31% of those with migraines experience an aura, and nearly 99% of those with aura have visual symptoms.20,21 The classic visual aura starts as an irregular colored crescent of light with multi-colored edges in the center of the visual field that gradually progresses toward the periphery, lasting <60 minutes. These simple visual hallucinations are most common; more complex hallucinations are seen more frequently in migraine coma and familial hemiplegic migraine.

 

 

Approximately 5% of patients with epilepsy have occipital seizures, which almost always have visual manifestations. Epileptic visual hallucinations often are simple, brief, stereotyped, and fragmentary. They usually consist of small, brightly colored spots or shapes that flash.22 Complex visual hallucinations in epilepsy are similar to hypnagogic hallucinations but are rare. Intracranial electroencephalography recordings have shown that pathological excitation of visual cortical areas may be responsible for complex visual hallucinations in epilepsy.19

Dementia with Lewy bodies (DLB) is associated with visual hallucinations.23 Visual hallucinations occur in >20% of patients with DLB.24 Patients with DLB may see complex scenarios of people and items that are not present. Visual hallucinations have an 83% positive predictive value for distinguishing DLB from dementia of the Alzheimer’s type.25 There is a strong correlation between Lewy bodies located in the amygdala and parahippocampus and well-formed visual hallucinations.26

Visual hallucinations are common in Parkinson’s disease and may occur in up to one-half of patients.27 Patients with Parkinson’s disease may experience hallucinations similar to those observed in DLB, which can range from seeing a person or animal to more complex, formed, and mobile people, animals, or objects.

Table 2

Common causes of visual hallucinations

Neurologic disorders
Migraine
Epilepsy
Hemispheric lesions
Optic nerve disorders
Brain stem lesions (peduncular hallucinosis)
Narcolepsy
Ophthalmologic diseases
Glaucoma
Retinal disease
Enucleation
Cataract formation
Choroidal disorder
Macular abnormalities
Toxic and metabolic conditions
Toxic-metabolic encephalopathy
Drug and alcohol withdrawal syndromes
Hallucinogens
Schizophrenia
Affective disorders
Conversion disorders
Sensory deprivation
Sleep deprivation
Hypnosis
Intense emotional experiences
Source: Reference 13

Olfactory hallucinations

Also known as phantosmia, olfactory hallucinations involve smelling odors that are not derived from any physical stimulus. They can occur with several psychiatric conditions, including schizophrenia, depression, bipolar disorder, eating disorders, and substance abuse.28 Olfactory hallucinations caused by epileptic activity are rare. They constitute approximately 0.9% of all auras and typically are described as unpleasant. Tumors that affect the medial temporal lobe and mesial temporal sclerosis are associated with olfactory hallucinations.29 Olfactory hallucinations also have been reported in patients with multi-infarct dementia, Alzheimer’s disease, and alcoholic psychosyndromes. In patients with schizophrenia, the smell may be perceived as coming from an external source, whereas patients with depression may perceive the source as internal.30 Patients who perceive that they are the source of an offensive odor—a condition known as olfactory reference syndrome—may wash excessively, overuse deodorants and perfumes, or become socially withdrawn.30

Gustatory hallucinations

Patients with gustatory hallucinations may experience salivation, sensation of thirst, or taste alterations. These hallucinations can be observed when the sylvian fissure that extends to the insula is stimulated electrically.31 Similar to olfactory hallucinations, gustatory hallucinations are associated with temporal lobe disease and parietal operculum lesions.31,32 Sinus diseases have been associated with olfactory and gustatory hallucinations.33 Brief gustatory hallucinations can be elicited with stimulation of the right rolandic operculum, parietal operculum, amygdala, hippocampus, medial temporal gyrus, and anterior part of right temporal gyrus.34

Tactile hallucinations

These hallucinations may include perceptions of insects crawling over or under the skin (formication) or simulation of pressure on skin.35 They have been associated with substance abuse, toxicity, or withdrawal.28 Tactile hallucinations are characteristic of cocaine or amphetamine intoxication.35

Tactile hallucinations are a rare symptom of schizophrenia. Heveling and colleagues reported a case of a woman, age 68, with chronic schizophrenia who experienced touching and being touched by a “shadow man” several times a day in addition to auditory and visual hallucinations.36 Her symptoms disappeared after 4 weeks of antipsychotic and mood stabilizer therapy.

Tactile hallucinations have been associated with obsessive-compulsive disorder (OCD).37 Fontenelle and colleagues suggested that OCD and psychotic disorders may share dysfunctional dopaminergic circuits.37

Somatic hallucinations

Patients who have somatic hallucinations report perceptions of abnormal body sensations or physical experiences. For example, a patient may have sense of not having a stomach while eating.35

This type of hallucination has been associated with activation of postcentral gyrus, parietal operculum, insula, and inferior parietal lobule on stereoelectroencephalography.34 In a study of cerebral blood flow in 20 geriatric patients with delusional disorder, somatic type who were experiencing somatic hallucinations, positron emission testing scan demonstrated increased perfusion in somatic sensory processing regions, particularly the left postcentral gyrus and the right paracentral lobule.38 Other researchers have linked somatic hallucinations with activation in the primary somatosensory and posterior parietal cortex, areas that normally mediate tactile perception.39

Related Resource

  • Teeple RC, Caplan JP, Stern TA. Visual hallucinations: differential diagnosis and treatment. Prim Care Companion J Clin Psychiatry. 2009;11(1):26-32.
 

 

Disclosures

Drs. Ali, Patel, Avenido, Bailey, and Jabeen report no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. Riley is on the board of directors for Vertex Pharmaceuticals.

Acknowledgment

The authors would like to thank Marwah Shahid, Research Associate, Vanderbilt University, Nashville, TN.

References

1. Cummings JL, Mega MS. Hallucinations. In: Cummings JL Mega MS, eds. Neuropsychiatry and behavioral neuroscience. New York, NY: Oxford University Press; 2003: 187–199.

2. Shergill SS, Murray RM, McGuire PK. Auditory hallucinations: a review of psychological treatments. Schizophr Res. 1998;32(3):137-150.

3. Goodwin DW, Alderson P, Rosenthal R. Clinical significance of hallucinations in psychiatric disorders. A study of 116 hallucinatory patients. Arch Gen Psychiatry. 1971;24(1):76-80.

4. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.

5. Kasper BS, Kasper EM, Pauli E, et al. Phenomenology of hallucinations, illusions, and delusions as part of seizure semiology. Epilepsy Behav. 2010;18(1-2):13-23.

6. Currie S, Heathfield KW, Henson RA, et al. Clinical course and prognosis of temporal lobe epilepsy. A survey of 666 patients. Brain. 1971;94(1):173-190.

7. Keshavan MS, David AS, Steingard S, et al. Musical hallucinations: a review and synthesis. Cogn Behav Neurol. 1992;5(3):211-223.

8. Duncan R, Mitchell JD, Critchley EMR. Hallucinations and music. Behav Neurol. 1989;2(2):115-124.

9. Tarachow S. The clinical value of hallucinations in localizing brain tumors. Am J Psychiatry. 1941;97:1434-1442.

10. Lanska DJ, Lanska MJ, Mendez MF. Brainstem auditory hallucinosis. Neurology. 1987;37(10):1685.-

11. Norton JW, Corbett JJ. Visual perceptual abnormalities: hallucinations and illusions. Semin Neurol. 2000;20(1):111-121.

12. Kaplan HI, Sadock BJ, Grebb JA. Typical signs and symptoms of psychiatric illness defined. In: Kaplan HI Sadock BJ, Grebb JA, eds. Kaplan and Sadock’s synopsis of psychiatry: behavioral sciences, clinical psychiatry. Baltimore, MD: Williams and Wilkins; 1994:300.

13. Cummings JL, Miller BL. Visual hallucinations. Clinical occurrence and use in differential diagnosis. West J Med. 1987;146(1):46-51.

14. First MB, Tasman A. Schizophrenia and other psychoses. In: First MB Tasman A, eds. Clinical guide to the diagnosis and treatment of mental disorders. San Francisco, CA: John Wiley and Sons; 2009:245–278.

15. Mueser KT, Bellack AS, Brady EU. Hallucinations in schizophrenia. Acta Psychiatr Scand. 1990;82(1):26-29.

16. Small IF, Small JG, Andersen JM. Clinical characteristics of hallucinations of schizophrenia. Dis Nerv Syst. 1966;27(5):349-353.

17. Webster R, Holroyd S. Prevalence of psychotic symptoms in delirium. Psychosomatics. 2000;41(6):519-522.

18. Gastfriend DR, Renner JA, Hackett TP. Alcoholic patients: acute and chronic. In: Stern TA Fricchione G, Cassem NH, et al, eds. Massachusetts General Hospital handbook of general hospital psychiatry. 5th ed. Philadelphia, PA: Mosby; 2004:203–216.

19. Manford M, Andermann F. Complex visual hallucinations. Clinical and neurobiological insights. Brain. 1998;121(Pt 10):1819-1840.

20. Goadsby PJ, Lipton RB, Ferrari MD. Migraine—current understanding and treatment. N Engl J Med. 2002;346(4):257-270.

21. Russell MB, Olesen J. A nosographic analysis of the migraine aura in a general population. Brain. 1996;119(Pt 2):355-361.

22. Panayiotopoulos CP. Elementary visual hallucinations blindness, and headache in idiopathic occipital epilepsy: differentiation from migraine. J Neurol Neurosurg Psychiatry. 1999;66(4):536-540.

23. Ballard CG, O’Brien JT, Swann AG, et al. The natural history of psychosis and depression in dementia with Lewy bodies and Alzheimer’s disease: persistence and new cases over 1 year of follow-up. J Clin Psychiatry. 2001;62(1):46-49.

24. Ala TA, Yang KH, Sung JH, et al. Hallucinations and signs of parkinsonism help distinguish patients with dementia and cortical Lewy bodies from patients with Alzheimer’s disease at presentation: a clinicopathological study. J Neurol Neurosurg Psychiatry. 1997;62(1):16-21.

25. Tiraboschi P, Salmon DP, Hansen LA, et al. What best differentiates Lewy body from Alzheimer’s disease in early-stage dementia? Brain. 2006;129(Pt 3):729-735.

26. Harding AJ, Broe GA, Halliday GM. Visual hallucinations in Lewy body disease relate to Lewy bodies in the temporal lobe. Brain. 2002;125(Pt 2):391-403.

27. Williams DR, Lees AJ. Visual hallucinations in the diagnosis of idiopathic Parkinson’s disease: a retrospective autopsy study. Lancet Neurol. 2005;4(10):605-610.

28. Lewandowski KE, DePaola J, Camsari GB, et al. Tactile, olfactory, and gustatory hallucinations in psychotic disorders: a descriptive study. Ann Acad Med Singapore. 2009;38(5):383-385.

29. Acharya V, Acharya J, Lüders H. Olfactory epileptic auras. Neurology. 1998;51(1):56-61.

30. Ropper AH, Samuels MA. Disorders of smell and taste. In: Ropper AH Samuels MA, eds. Adams and Victor’s principles of neurology. 9th ed. New York, NY: McGraw-Hill Companies; 2009:216–224.

31. Ropper AH, Samuels MA. Epilepsy and other seizure disorders. In: Ropper AH Samuels MA, eds. Adams and Victor’s principles of neurology. 9th ed. New York, NY: McGraw-Hill Companies; 2009:304–338.

32. Capampangan DJ, Hoerth MT, Drazkowski JF, et al. Olfactory and gustatory hallucinations presenting as partial status epilepticus because of glioblastoma multiforme. Ann Emerg Med. 2010;56(4):374-377.

33. Frasnelli J, Reden J, Landis BN, et al. Comment on “Olfactory hallucinations as a manifestation of hidden rhinosinusitis”. J Clin Neurosci. 2010;17(4):543.-

34. Elliott B, Joyce E, Shorvon S. Delusions illusions and hallucinations in epilepsy: 1. Elementary phenomena. Epilepsy Res. 2009;85(2-3):162-171.

35. Nurcombe B, Ebert MH. The psychiatric interview. In: Ebert MH Nurcombe B, Loosen PT, et al, eds. Current diagnosis and treatment: psychiatry. 2nd ed. New York, NY: McGraw-Hill Companies; 2008:95–114.

36. Heveling T, Emrich HM, Dietrich DE. Treatment of a rare psychopathological phenomenon: tactile hallucinations and the delusional other. Eur Psychiatry. 2004;19(6):387-388.

37. Fontenelle LF, Lopes AP, Borges MC, et al. Auditory, visual, tactile, olfactory, and bodily hallucinations in patients with obsessive-compulsive disorder. CNS Spectr. 2008;13(2):125-130.

38. Nemoto K, Mizukami K, Hori T, et al. Hyperperfusion in primary somatosensory region related to somatic hallucination in the elderly. Psychiatry Clin Neurosci. 2010;64(4):421-425.

39. Shergill SS, Cameron LA, Brammer MJ, et al. Modality specific neural correlates of auditory and somatic hallucinations. J Neurol Neurosurg Psychiatry. 2001;71(5):688-690.

References

1. Cummings JL, Mega MS. Hallucinations. In: Cummings JL Mega MS, eds. Neuropsychiatry and behavioral neuroscience. New York, NY: Oxford University Press; 2003: 187–199.

2. Shergill SS, Murray RM, McGuire PK. Auditory hallucinations: a review of psychological treatments. Schizophr Res. 1998;32(3):137-150.

3. Goodwin DW, Alderson P, Rosenthal R. Clinical significance of hallucinations in psychiatric disorders. A study of 116 hallucinatory patients. Arch Gen Psychiatry. 1971;24(1):76-80.

4. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington DC: American Psychiatric Association; 2000.

5. Kasper BS, Kasper EM, Pauli E, et al. Phenomenology of hallucinations, illusions, and delusions as part of seizure semiology. Epilepsy Behav. 2010;18(1-2):13-23.

6. Currie S, Heathfield KW, Henson RA, et al. Clinical course and prognosis of temporal lobe epilepsy. A survey of 666 patients. Brain. 1971;94(1):173-190.

7. Keshavan MS, David AS, Steingard S, et al. Musical hallucinations: a review and synthesis. Cogn Behav Neurol. 1992;5(3):211-223.

8. Duncan R, Mitchell JD, Critchley EMR. Hallucinations and music. Behav Neurol. 1989;2(2):115-124.

9. Tarachow S. The clinical value of hallucinations in localizing brain tumors. Am J Psychiatry. 1941;97:1434-1442.

10. Lanska DJ, Lanska MJ, Mendez MF. Brainstem auditory hallucinosis. Neurology. 1987;37(10):1685.-

11. Norton JW, Corbett JJ. Visual perceptual abnormalities: hallucinations and illusions. Semin Neurol. 2000;20(1):111-121.

12. Kaplan HI, Sadock BJ, Grebb JA. Typical signs and symptoms of psychiatric illness defined. In: Kaplan HI Sadock BJ, Grebb JA, eds. Kaplan and Sadock’s synopsis of psychiatry: behavioral sciences, clinical psychiatry. Baltimore, MD: Williams and Wilkins; 1994:300.

13. Cummings JL, Miller BL. Visual hallucinations. Clinical occurrence and use in differential diagnosis. West J Med. 1987;146(1):46-51.

14. First MB, Tasman A. Schizophrenia and other psychoses. In: First MB Tasman A, eds. Clinical guide to the diagnosis and treatment of mental disorders. San Francisco, CA: John Wiley and Sons; 2009:245–278.

15. Mueser KT, Bellack AS, Brady EU. Hallucinations in schizophrenia. Acta Psychiatr Scand. 1990;82(1):26-29.

16. Small IF, Small JG, Andersen JM. Clinical characteristics of hallucinations of schizophrenia. Dis Nerv Syst. 1966;27(5):349-353.

17. Webster R, Holroyd S. Prevalence of psychotic symptoms in delirium. Psychosomatics. 2000;41(6):519-522.

18. Gastfriend DR, Renner JA, Hackett TP. Alcoholic patients: acute and chronic. In: Stern TA Fricchione G, Cassem NH, et al, eds. Massachusetts General Hospital handbook of general hospital psychiatry. 5th ed. Philadelphia, PA: Mosby; 2004:203–216.

19. Manford M, Andermann F. Complex visual hallucinations. Clinical and neurobiological insights. Brain. 1998;121(Pt 10):1819-1840.

20. Goadsby PJ, Lipton RB, Ferrari MD. Migraine—current understanding and treatment. N Engl J Med. 2002;346(4):257-270.

21. Russell MB, Olesen J. A nosographic analysis of the migraine aura in a general population. Brain. 1996;119(Pt 2):355-361.

22. Panayiotopoulos CP. Elementary visual hallucinations blindness, and headache in idiopathic occipital epilepsy: differentiation from migraine. J Neurol Neurosurg Psychiatry. 1999;66(4):536-540.

23. Ballard CG, O’Brien JT, Swann AG, et al. The natural history of psychosis and depression in dementia with Lewy bodies and Alzheimer’s disease: persistence and new cases over 1 year of follow-up. J Clin Psychiatry. 2001;62(1):46-49.

24. Ala TA, Yang KH, Sung JH, et al. Hallucinations and signs of parkinsonism help distinguish patients with dementia and cortical Lewy bodies from patients with Alzheimer’s disease at presentation: a clinicopathological study. J Neurol Neurosurg Psychiatry. 1997;62(1):16-21.

25. Tiraboschi P, Salmon DP, Hansen LA, et al. What best differentiates Lewy body from Alzheimer’s disease in early-stage dementia? Brain. 2006;129(Pt 3):729-735.

26. Harding AJ, Broe GA, Halliday GM. Visual hallucinations in Lewy body disease relate to Lewy bodies in the temporal lobe. Brain. 2002;125(Pt 2):391-403.

27. Williams DR, Lees AJ. Visual hallucinations in the diagnosis of idiopathic Parkinson’s disease: a retrospective autopsy study. Lancet Neurol. 2005;4(10):605-610.

28. Lewandowski KE, DePaola J, Camsari GB, et al. Tactile, olfactory, and gustatory hallucinations in psychotic disorders: a descriptive study. Ann Acad Med Singapore. 2009;38(5):383-385.

29. Acharya V, Acharya J, Lüders H. Olfactory epileptic auras. Neurology. 1998;51(1):56-61.

30. Ropper AH, Samuels MA. Disorders of smell and taste. In: Ropper AH Samuels MA, eds. Adams and Victor’s principles of neurology. 9th ed. New York, NY: McGraw-Hill Companies; 2009:216–224.

31. Ropper AH, Samuels MA. Epilepsy and other seizure disorders. In: Ropper AH Samuels MA, eds. Adams and Victor’s principles of neurology. 9th ed. New York, NY: McGraw-Hill Companies; 2009:304–338.

32. Capampangan DJ, Hoerth MT, Drazkowski JF, et al. Olfactory and gustatory hallucinations presenting as partial status epilepticus because of glioblastoma multiforme. Ann Emerg Med. 2010;56(4):374-377.

33. Frasnelli J, Reden J, Landis BN, et al. Comment on “Olfactory hallucinations as a manifestation of hidden rhinosinusitis”. J Clin Neurosci. 2010;17(4):543.-

34. Elliott B, Joyce E, Shorvon S. Delusions illusions and hallucinations in epilepsy: 1. Elementary phenomena. Epilepsy Res. 2009;85(2-3):162-171.

35. Nurcombe B, Ebert MH. The psychiatric interview. In: Ebert MH Nurcombe B, Loosen PT, et al, eds. Current diagnosis and treatment: psychiatry. 2nd ed. New York, NY: McGraw-Hill Companies; 2008:95–114.

36. Heveling T, Emrich HM, Dietrich DE. Treatment of a rare psychopathological phenomenon: tactile hallucinations and the delusional other. Eur Psychiatry. 2004;19(6):387-388.

37. Fontenelle LF, Lopes AP, Borges MC, et al. Auditory, visual, tactile, olfactory, and bodily hallucinations in patients with obsessive-compulsive disorder. CNS Spectr. 2008;13(2):125-130.

38. Nemoto K, Mizukami K, Hori T, et al. Hyperperfusion in primary somatosensory region related to somatic hallucination in the elderly. Psychiatry Clin Neurosci. 2010;64(4):421-425.

39. Shergill SS, Cameron LA, Brammer MJ, et al. Modality specific neural correlates of auditory and somatic hallucinations. J Neurol Neurosurg Psychiatry. 2001;71(5):688-690.

Issue
Current Psychiatry - 10(11)
Issue
Current Psychiatry - 10(11)
Page Number
22-29
Page Number
22-29
Publications
Publications
Topics
Article Type
Display Headline
Hallucinations: Common features and causes
Display Headline
Hallucinations: Common features and causes
Legacy Keywords
hallucinations; features; causes; Shahid Ali; Milapkumar Patel; Jaymie Avenido; Rahn Bailey; Shagufta Jabeen; Wayne Riley; auditory hallucinations; visual hallucinations; olfactory hallucinations; gustatory hallucinations; tactile hallucinations; somatic hallucinations
Legacy Keywords
hallucinations; features; causes; Shahid Ali; Milapkumar Patel; Jaymie Avenido; Rahn Bailey; Shagufta Jabeen; Wayne Riley; auditory hallucinations; visual hallucinations; olfactory hallucinations; gustatory hallucinations; tactile hallucinations; somatic hallucinations
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Steps to take when a patient develops tardive dyskinesia

Article Type
Changed
Wed, 09/26/2018 - 11:48
Display Headline
Steps to take when a patient develops tardive dyskinesia

Issue
Current Psychiatry - 10(10)
Publications
Topics
Sections

Issue
Current Psychiatry - 10(10)
Issue
Current Psychiatry - 10(10)
Publications
Publications
Topics
Article Type
Display Headline
Steps to take when a patient develops tardive dyskinesia
Display Headline
Steps to take when a patient develops tardive dyskinesia
Sections
Disallow All Ads
Alternative CME
Use ProPublica

A curious case of depression

Article Type
Changed
Tue, 12/11/2018 - 15:30
Display Headline
A curious case of depression

Discuss this article at www.facebook.com/CurrentPsychiatry

Mr. Z, age 61, is referred by his primary care clinician to the hospital’s medical service with increasing depressive symptoms and non-pruritic rash. He has a history of bipolar I disorder for >30 years. When the primary care physician evaluated Mr. Z, his vitals were normal, but blood work revealed mild anemia and thrombocytopenia of 34 x103/μL, which prompted referral to the hospital. During admission, the psychiatric consultation service is called to evaluate Mr. Z’s depressive symptoms.

Mr. Z reports having chronic sleep problems and feeling cold and tired, shivering at times, but has no pain. He says he’s worried because he feels severely depressed, worthless, and hopeless, but denies suicidal ideation and psychosis. Mr. Z says he started experiencing increasingly depressed mood, anhedonia, insomnia, fatigue, poor appetite, and concentration 2 months ago. At that time his outpatient psychiatrist started Mr. Z on risperidone, 6 mg/d, and divalproex, 1,500 mg at bedtime because of emerging mood symptoms, after he was off medication for 7 months. Mr. Z attributed his worsened mood symptoms to being overwhelmed by several psychosocial stressors, including going through a complicated divorce, financial problems, and homelessness after being evicted from his apartment.

A review of Mr. Z’s psychiatric history reveals several remote hospitalizations—the last was 7 years ago—for escalated manic symptoms after he stopped taking his medication. He denies past suicide attempts. Mr. Z says he is compliant with his current medication regimen—risperidone, 6 mg/d, and divalproex, 1,500 mg at bedtime. He denies illicit drug use and says he drinks “a couple of beers, mostly on weekends.” Family history is positive for depression and bipolar II disorder.

His medical history is significant for hypothyroidism after goiter removal 6 years ago, for which he takes levothyroxine, 150 mcg/d, and a sports injury-related splenectomy in childhood. He reports no allergies. Vital signs at the time of admission are temperature, 99.1°F; pulse, 98 beats per minute; respiration, 16 breaths per minute; blood pressure, 123/73 mm/Hg; and oxygen saturation, 97%.

During the interview, Mr. Z presents with tired facies and exhibits psychomotor retardation. He has to force himself to stay engaged in the evaluation and maintain eye contact. His speech is clear, regular, and soft. Mr. Z says he is “very depressed”; his affect is constricted, almost flat, stable, and consistent with depressed mood. His thought process is linear and somewhat concrete and his thought content is notable for hopelessness, although Mr. Z continues to deny suicidal or homicidal ideations. No hallucinations or apparent delusions are noted. Insight and judgment are fair. Mr. Z understands his current mental state; however, he displays some lack of knowledge regarding his current hospitalization. Cognition is intact.

The authors’ observations

The differential diagnosis in patients presenting with mood changes is extensive (Table 1)1 and in Mr. Z’s case includes several precipitating and perpetuating factors. Mr. Z presents with severe depressive symptoms and meets DSM-IV-TR criteria for a major depressive episode (MDE). This presentation is not typical of his bipolar I disorder because Mr. Z has never experienced an MDE and usually presents with escalating hypomanic/manic symptoms in the context of medication nonadherence. Nevertheless, Mr. Z has several risk factors for severe depression, including a family psychiatric history, multiple enduring social stressors and life crises, and medical conditions.

In the general population, the lifetime risk for developing depression is 8% to 17%.2 The risk of developing a mood disorder increases significantly if a first-degree relative is diagnosed with a mood disorder; the relative risk is 10.3 for bipolar disorder and 3.2 for depression.3 Additionally, Mr. Z is going through a complicated divorce, has financial problems, and is homeless, all of which could trigger an MDE. Furthermore, hypothyroidism shares many symptoms of depression, including fatigue, lethargy, anhedonia, cold intolerance, and low mood; mental status changes frequently are the initial presentation of thyroid problems.4 Physicians started Mr. Z on a new medication regimen (risperidone and divalproex) to control mood instability, which coincided with symptom onset. Atypical antipsychotics have been reported to precipitate depressive symptoms; their side effect profile includes extrapyramidal effects, such as flat affect, which can be mistaken for depression.5 Rapid valproate titration can mimic neurovegetative symptoms of depression and cause dose-dependent thrombocytopenia and rash, which could explain his initial presentation.6 Finally, Mr. Z’s history of traumatic splenectomy, change in mental status, and thrombocytopenia suggest an infectious etiology.

Table 1

Differential diagnosis in patients presenting with mood changes

 

 

Cerebrovascular disease
Degenerative disorders (Parkinson’s disease, Huntington’s disease, Wilson’s disease)
Demyelinating disorders (multiple sclerosis, amyotrophic lateral sclerosis, lipid storage disease)
Endocrine disorders (Addison’s disease, Cushing’s disease, hyperthyroidism, hypothyroidism, hyperparathyroidism, pituitary dysfunction)
Epilepsy
Infectious diseases
Immune diseases
Metabolic encephalopathy
Neoplasm
Nutritional deficits (thiamine, niacin, vitamin B12)
Primary psychiatric disorders (mood disorders, dementia, sleep disorders)
Substance use
Toxins/medications
Traumatic brain injury
Source: Reference 1

Possible infectious causes

The increased prevalence of immune suppression due to human immunodeficiency virus/acquired immune deficiency syndrome (HIV/AIDS) or from therapeutic modalities such as cancer therapy or splenectomy has led to an increased number of chronic CNS infections, manifesting with an array of neuropsychiatric symptoms and nonspecific physiological reactions.1,7 Mr. Z complains of a 2-month period of worsening depression that could suggest an infectious process with an insidious onset. Some infectious agents that can cause chronic CNS infection and encephalopathy are presented in Table 2.8 HIV, tuberculosis, syphilis, Lyme disease, and herpes simplex virus are becoming more prevalent and can present with neuropsychiatric symptoms.1 For example, in addition to thrombocytopenia and low-grade fever, patients with HIV may exhibit a broad range of neuropsychiatric symptoms such as cognitive problems, impaired executive and motor functioning, sleep disturbance, and anxiety. These patients frequently present with low mood and neurovegetative symptoms of depression.7 Similarly, the same tick responsible for Lyme disease infection can transmit other infectious agents that can cause thrombocytopenia, including Babesia, Ehrlichia chaffeensis, Anaplasma phagocytophilum, and human Ewingii ehrlichiosis.

The authors’ observations

Diagnosis of a mood change, particularly an MDE, is clinical, based on careful psychiatric evaluation using standardized criteria rather than a specific lab test. However, some laboratory studies (Table 3)1 are useful in differentiating medical illnesses that may present with depression. Mr. Z’s presentation warrants investigating these tests. His history of traumatic splenectomy and night sweats suggests an infection. The team’s initial recommendations include laboratory tests, discontinuing divalproex because it may be causing thrombocytopenia, and decreasing risperidone to 2 mg/d to improve his fatigue and possibly developed extrapyramidal symptoms.

Table 2

Potential infectious causes of chronic encephalopathy

Type of infectionOrganism/disease
MycobacterialMycobacterium tuberculosis
SpirochetalTreponema pallidum (syphilis), Borrelia burgdorferi (Lyme disease), Leptospira
BacterialBrucella, Listeria, Nocardia, Actinomyces israelii, Whipple’s disease
ViralHIV/AIDS, cytomegalovirus, varicella zoster virus, herpes simplex virus, enterovirus
FungalHistoplasmosis, coccidiosis, sporothrix, Blastomyces, Cryptococcus
ParasiticToxoplasmosis, taenia solium (cysticercosis), Schistosoma, Acanthamoeba
AIDS: acquired immunodeficiency syndrome; HIV: human immunodeficiency virus
Source: Reference 8

Table 3

Differentiating medical illnesses that may mimic depression

Laboratory tests
  • CBC, thyroid-stimulating hormone, antinuclear antibody, erythrocyte sedimentation rate, vitamin B12, rapid plasma reagin, HIV test, electrolytes and calcium levels and renal function test, liver function tests, blood alcohol, blood, and urine toxicology screen, ABG, Lyme antibody test (ELISA), dexamethasone suppression test (Cushing’s disease), cosyntropin stimulation test (Addison’s disease)
Imaging studies
  • CT scan or MRI of the brain
Other tests
  • EEG
Procedures
  • Lumbar puncture for VDRL, Lyme antibody, cell count, chemistry, and protein electrophoresis
ABG: arterial blood gases; CBC: complete blood count;
EEG: electroencephalogram; ELISA: enzyme-linked immunosorbent assay; HIV: human immunodeficiency virus; VDRL: venereal disease research laboratory
Source: Reference 1

TREATMENT: Cause revealed

Mr. Z develops a persistent fever of 102°F with continuous profuse sweating and a hypotensive episode. Blood work reveals mild anemia, thrombocytopenia, and increased coagulation parameters with high D-dimer and low fibrinogen, consistent with diagnosis of disseminated intravascular coagulation (DIC) secondary to infectious etiology. Thyroid and HIV tests are negative. After further evaluation, Mr. Z remembers that 4 months earlier he removed several ticks from his legs after hunting; he also remembers experiencing shivering and night sweats several weeks before he was hospitalized. His blood smear is positive for babesiosis and further testing confirms positive Lyme antibodies. Mr. Z is started on aggressive hemodynamic stabilization and a pathogen-tailored course of antibiotics for several weeks. This results in improvement and discharge home in a stable condition. His depression and fatigue improve but do not fully remit by the time he is discharged.

The authors’ observations

Lyme disease is one of the fastest-growing infectious diseases in the United States.9 The prevalence of positive Lyme antibodies is 30% higher in psychiatric populations than the general population.10 Lyme disease is transmitted by deer tick bite, often undetected, that is infected with spirochete Borrelia burgdorferi. To be infectious, ticks need to be attached to the skin for 24 to 48 hours,11,12 although individual cases have reported transmission in <24 hours. The clinical manifestations of Lyme disease can be divided into 3 phases:

  • early localized phase, characterized by the distinctive skin lesion erythema migrans with or without constitutional symptoms
  • early disseminated phase, characterized by multiple erythema migrans lesions and neurologic and/or cardiac findings
  • late or chronic disease associated with intermittent/persistent arthritis and/or neurologic problems.11,13
 

 

The clinical features of each stage frequently overlap and some patients in a later stage of Lyme disease do not have prior signs or symptoms of the disease. Because it is a multisystem disease, Lyme disease can attack the CNS in the form of neuroborreliosis, a clinical diagnosis, without involving other systems, and its neuropsychiatric manifestations can resemble neurosyphilis because both organisms are spirochetes.11,13,14 CNS disorders have been found in up to 40% of Lyme disease cases.11 In neuroborreliosis, cognitive problems usually predominate; however, neuroborreliosis can mimic multiple brain diseases presenting with various neurologic and psychiatric symptoms (Table 4)14,15 and can present at any time after the tick bite. Furthermore neuroborreliosis is difficult to diagnose because symptoms may remain dormant and emerge after several years.11,13,14Borrelia burgdorferi is challenging to isolate and grow in the lab, and enzyme-linked immunosorbent assay (ELISA) testing for antibodies is highly specific but not very sensitive,16 frequently giving false negative results. Western blot confirms the diagnosis.

Table 4

Late-stage neuropsychiatric symptoms of Lyme disease

Cognitive problems, memory problems, forgetfulness, slowing of thought processing, dysfunction in visuospatial orientation, dyslexia
Depression
Mood swings
Psychosis
Violent behavior/irritability
OCD
Anxiety
Panic attacks
Sleep disorders
Seizures
ADHD-like symptoms
Autism-like behavior
Chronic fatigue syndrome
Fibromyalgia
ADHD: attention-deficit/hyperactivity disorder; OCD: obsessive-compulsive disorder
Source: References 14,15

Mr. Z’s presentation also reflects co-infection with babesiosis. Babesia is malaria-like protozoa diagnosed by blood smear that can cause a fatal illness in immuno-compromised patients. The clinical picture varies from mild symptoms such as night sweats, chills, arthralgias, and anorexia with thrombocytopenia to severe and potentially fatal outcomes in immunocompromised patients, including DIC, acute renal failure, sepsis, congestive heart failure, and myocardial infarction.11 Risk factors for the severest forms of babesiosis are age >50, co-infection with Lyme disease, and splenectomy,11 all of which were present in Mr. Z. Co-infected patients experience fatigue, headache, anorexia, and emotional lability more frequently than those with Lyme disease alone.12

Treatment options

Treatment of Lyme disease/neuroborreliosis is complex. The mainstay approach is antibiotics. Despite adequate treatment, many patients experience continued impairment, including chronic pain, fatigue, and cognitive and psychiatric symptoms.11,14 There is some evidence Borrelia burgdorferi can persist and re-emerge after adequate treatment.14,17 The National Institute of Health sponsored several clinical trials of prolonged antibiotic treatment for chronic Lyme disease. Some results suggested improvement in fatigue and cognitive function, although these results were not sustained.18

There is a strong link between mental illness and increased prevalence of positive Lyme disease antibodies.10 Several studies report increased risk of infection during psychological stress that may be related to an altered immune system response.19 Evidence suggests that Borrelia burgdorferi can alter immune system response, making T cells more reactive not only to Borrelia burgdorferi antigens but also to host antigens,20 creating autoimmune inflammatory reactions that could explain chronic neuropsychiatric symptoms. It appears Lyme disease antigens can mimic certain autoantigens (for example, in the thyroid gland).21 Whether there is a role for autoimmune therapy in treating chronic symptoms needs to be investigated.

Once Lyme disease is diagnosed, educating patients and families becomes an important part of treatment because many patients report feeling stigmatized by the diagnosis. Referral to a Lyme disease support group may be beneficial. Patients with neuropsychiatric symptoms that persist after antibiotic treatment should be offered symptom-based treatment, including medications and therapy.

Related Resource

Drug Brand Names

  • Divalproex sodium • Depakote
  • Levothyroxine • Levoxyl, Synthroid
  • Risperidone • Risperdal

Disclosure

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

References

1. Stern TA, Rosenbaum JF, Fava M, et al. eds. Massachusetts General Hospital comprehensive clinical psychiatry. Philadelphia, PA: Mosby; 2008;257-277.

2. Andrade L, Caraveo-Anduaga JJ, Berglund P, et al. The epidemiology of major depressive episodes: results from the International Consortium of Psychiatric Epidemiology (ICPE) Surveys. Int J Methods Psychiatr Res. 2003;12(1):3-21.

3. Merikangas KR, Low NC. The epidemiology of mood disorders. Curr Psychiatry Rep. 2004;6:411-421.

4. Brown GM. Psychiatric and neurologic aspects of endocrine disease. In Krieger DT Hughes JC, eds. Neuroendocrinology. Sunderland, MA: Sinauer Associates; 1980;185-194.

5. Maguire GA. Comprehensive understanding of schizophrenia and its treatment. Am J Health Syst Pharm. 2002;59(17 suppl 5):S4-S11.

6. Dreifuss FE, Langer DH. Side effects of valproate. Am J Med. 1988;84(1A):34-41.

7. Perry S, Jacobsen P. Neuropsychiatric manifestations of AIDS-spectrum disorders. Hosp Community Psychiatry. 1986;37:135-142.

8. Hildebrand J, Aoun M. Chronic meningitis: still a diagnostic challenge. J Neurol. 2003;250(6):653-660.

9. Bacon RM, Kugeler KJ, Mead PS. Centers for Disease Control and Prevention (CDC). Surveillance for Lyme disease—United States 1992-2006. MMWR Surveill Summ. 2008;57(10):1-9.

10. Hájek T, Pasková B, Janovská D, et al. Higher prevalence of antibodies to Borrelia burgdorferi in psychiatric patients than in healthy subjects. Am J Psychiatry. 2002;159:297-301.

11. Wormser GP, Dattwyler RJ, Shapiro ED, et al. The clinical assessment, treatment, and prevention of lyme disease, human granulocytic anaplasmosis, and babesiosis: clinical practice guidelines by the Infectious Diseases Society of America. Clin Infect Dis. 2006;43(9):1089-1134.

12. Krause PJ, Telford SR 3rd, Spielman A, et al. Concurrent Lyme disease and babesiosis. Evidence for increased severity and duration of illness. JAMA. 1996;275(21):1657-1660.

13. Hildenbrand P, Craven DE, Jones R, et al. Lyme neuroborreliosis: manifestations of a rapidly emerging zoonosis. AJNR Am J Neuroradiol. 2009;30:1079-1087.

14. Fallon BA, Nields JA. Lyme disease: a neuropsychiatric illness. Am J Psychiatry. 1994;151:1571-1583.

15. Fallon BA, Nields JA, Burrascano JJ, et al. The neuropsychiatric manifestations of Lyme borreliosis. Psychiatr Q. 1992;63(1):95-117.

16. Stricker RB, Johnson L. Lyme wars: let’s tackle the testing. BMJ. 2007;335:1008.-

17. Hodzic E, Feng S, Holden K, et al. Persistence of Borrelia burgdorferi following antibiotic treatment in mice. Antimicrob Agents Chemother. 2008;52(5):1728-1736.

18. Fallon BA, Keilp JG, Corbera KM, et al. A randomized, placebo-controlled trial of repeated IV antibiotic therapy for Lyme encephalopathy. Neurology. 2008;70(13):992-1003.

19. Sheridan JF, Dobbs C, Brown D, et al. Psychoneuroimmunology: stress effects on pathogenesis and immunity during infection. Clin Microbiol Rev. 1994;7(2):200-212.

20. Yssel H, Shanafelt MC, Soderberg C, et al. Borrelia burgdorferi activates a T helper 1-like T cell subset in Lyme arthritis. J Exp Med. 1991;174:593-601.

21. Benvenga S, Guarneri F, Vaccaro M, et al. Homologies between proteins of Borrelia burgdorferi and thyroid autoantigens. Thyroid. 2004;14(11):964-966.

Article PDF
Author and Disclosure Information

Pavle Joksovic, MD
Dr. Joksovic is Senior Resident, Department of Psychiatry, Yale University School of Medicine, New Haven, CT
Pieter Joost van Wattum, MD, MA
Dr. van Wattum is Associate Clinical Professor, Department of Psychiatry and Yale Child Study Center, Yale University School of Medicine, New Haven, CT
Catherine Chiles, MD
Dr. Chiles is Associate Clinical Professor, Department of Psychiatry, Yale University School of Medicine, New Haven, CT

Issue
Current Psychiatry - 10(10)
Publications
Topics
Page Number
53-58
Legacy Keywords
Cases that test your skills; depression; Pavle Joksovic; Pieter Joost van Wattum; Catherine Chiles; bipolar I disorder; B; depression; fatigue; thrombocytopenia; rash; Lyme disease
Sections
Author and Disclosure Information

Pavle Joksovic, MD
Dr. Joksovic is Senior Resident, Department of Psychiatry, Yale University School of Medicine, New Haven, CT
Pieter Joost van Wattum, MD, MA
Dr. van Wattum is Associate Clinical Professor, Department of Psychiatry and Yale Child Study Center, Yale University School of Medicine, New Haven, CT
Catherine Chiles, MD
Dr. Chiles is Associate Clinical Professor, Department of Psychiatry, Yale University School of Medicine, New Haven, CT

Author and Disclosure Information

Pavle Joksovic, MD
Dr. Joksovic is Senior Resident, Department of Psychiatry, Yale University School of Medicine, New Haven, CT
Pieter Joost van Wattum, MD, MA
Dr. van Wattum is Associate Clinical Professor, Department of Psychiatry and Yale Child Study Center, Yale University School of Medicine, New Haven, CT
Catherine Chiles, MD
Dr. Chiles is Associate Clinical Professor, Department of Psychiatry, Yale University School of Medicine, New Haven, CT

Article PDF
Article PDF

Discuss this article at www.facebook.com/CurrentPsychiatry

Mr. Z, age 61, is referred by his primary care clinician to the hospital’s medical service with increasing depressive symptoms and non-pruritic rash. He has a history of bipolar I disorder for >30 years. When the primary care physician evaluated Mr. Z, his vitals were normal, but blood work revealed mild anemia and thrombocytopenia of 34 x103/μL, which prompted referral to the hospital. During admission, the psychiatric consultation service is called to evaluate Mr. Z’s depressive symptoms.

Mr. Z reports having chronic sleep problems and feeling cold and tired, shivering at times, but has no pain. He says he’s worried because he feels severely depressed, worthless, and hopeless, but denies suicidal ideation and psychosis. Mr. Z says he started experiencing increasingly depressed mood, anhedonia, insomnia, fatigue, poor appetite, and concentration 2 months ago. At that time his outpatient psychiatrist started Mr. Z on risperidone, 6 mg/d, and divalproex, 1,500 mg at bedtime because of emerging mood symptoms, after he was off medication for 7 months. Mr. Z attributed his worsened mood symptoms to being overwhelmed by several psychosocial stressors, including going through a complicated divorce, financial problems, and homelessness after being evicted from his apartment.

A review of Mr. Z’s psychiatric history reveals several remote hospitalizations—the last was 7 years ago—for escalated manic symptoms after he stopped taking his medication. He denies past suicide attempts. Mr. Z says he is compliant with his current medication regimen—risperidone, 6 mg/d, and divalproex, 1,500 mg at bedtime. He denies illicit drug use and says he drinks “a couple of beers, mostly on weekends.” Family history is positive for depression and bipolar II disorder.

His medical history is significant for hypothyroidism after goiter removal 6 years ago, for which he takes levothyroxine, 150 mcg/d, and a sports injury-related splenectomy in childhood. He reports no allergies. Vital signs at the time of admission are temperature, 99.1°F; pulse, 98 beats per minute; respiration, 16 breaths per minute; blood pressure, 123/73 mm/Hg; and oxygen saturation, 97%.

During the interview, Mr. Z presents with tired facies and exhibits psychomotor retardation. He has to force himself to stay engaged in the evaluation and maintain eye contact. His speech is clear, regular, and soft. Mr. Z says he is “very depressed”; his affect is constricted, almost flat, stable, and consistent with depressed mood. His thought process is linear and somewhat concrete and his thought content is notable for hopelessness, although Mr. Z continues to deny suicidal or homicidal ideations. No hallucinations or apparent delusions are noted. Insight and judgment are fair. Mr. Z understands his current mental state; however, he displays some lack of knowledge regarding his current hospitalization. Cognition is intact.

The authors’ observations

The differential diagnosis in patients presenting with mood changes is extensive (Table 1)1 and in Mr. Z’s case includes several precipitating and perpetuating factors. Mr. Z presents with severe depressive symptoms and meets DSM-IV-TR criteria for a major depressive episode (MDE). This presentation is not typical of his bipolar I disorder because Mr. Z has never experienced an MDE and usually presents with escalating hypomanic/manic symptoms in the context of medication nonadherence. Nevertheless, Mr. Z has several risk factors for severe depression, including a family psychiatric history, multiple enduring social stressors and life crises, and medical conditions.

In the general population, the lifetime risk for developing depression is 8% to 17%.2 The risk of developing a mood disorder increases significantly if a first-degree relative is diagnosed with a mood disorder; the relative risk is 10.3 for bipolar disorder and 3.2 for depression.3 Additionally, Mr. Z is going through a complicated divorce, has financial problems, and is homeless, all of which could trigger an MDE. Furthermore, hypothyroidism shares many symptoms of depression, including fatigue, lethargy, anhedonia, cold intolerance, and low mood; mental status changes frequently are the initial presentation of thyroid problems.4 Physicians started Mr. Z on a new medication regimen (risperidone and divalproex) to control mood instability, which coincided with symptom onset. Atypical antipsychotics have been reported to precipitate depressive symptoms; their side effect profile includes extrapyramidal effects, such as flat affect, which can be mistaken for depression.5 Rapid valproate titration can mimic neurovegetative symptoms of depression and cause dose-dependent thrombocytopenia and rash, which could explain his initial presentation.6 Finally, Mr. Z’s history of traumatic splenectomy, change in mental status, and thrombocytopenia suggest an infectious etiology.

Table 1

Differential diagnosis in patients presenting with mood changes

 

 

Cerebrovascular disease
Degenerative disorders (Parkinson’s disease, Huntington’s disease, Wilson’s disease)
Demyelinating disorders (multiple sclerosis, amyotrophic lateral sclerosis, lipid storage disease)
Endocrine disorders (Addison’s disease, Cushing’s disease, hyperthyroidism, hypothyroidism, hyperparathyroidism, pituitary dysfunction)
Epilepsy
Infectious diseases
Immune diseases
Metabolic encephalopathy
Neoplasm
Nutritional deficits (thiamine, niacin, vitamin B12)
Primary psychiatric disorders (mood disorders, dementia, sleep disorders)
Substance use
Toxins/medications
Traumatic brain injury
Source: Reference 1

Possible infectious causes

The increased prevalence of immune suppression due to human immunodeficiency virus/acquired immune deficiency syndrome (HIV/AIDS) or from therapeutic modalities such as cancer therapy or splenectomy has led to an increased number of chronic CNS infections, manifesting with an array of neuropsychiatric symptoms and nonspecific physiological reactions.1,7 Mr. Z complains of a 2-month period of worsening depression that could suggest an infectious process with an insidious onset. Some infectious agents that can cause chronic CNS infection and encephalopathy are presented in Table 2.8 HIV, tuberculosis, syphilis, Lyme disease, and herpes simplex virus are becoming more prevalent and can present with neuropsychiatric symptoms.1 For example, in addition to thrombocytopenia and low-grade fever, patients with HIV may exhibit a broad range of neuropsychiatric symptoms such as cognitive problems, impaired executive and motor functioning, sleep disturbance, and anxiety. These patients frequently present with low mood and neurovegetative symptoms of depression.7 Similarly, the same tick responsible for Lyme disease infection can transmit other infectious agents that can cause thrombocytopenia, including Babesia, Ehrlichia chaffeensis, Anaplasma phagocytophilum, and human Ewingii ehrlichiosis.

The authors’ observations

Diagnosis of a mood change, particularly an MDE, is clinical, based on careful psychiatric evaluation using standardized criteria rather than a specific lab test. However, some laboratory studies (Table 3)1 are useful in differentiating medical illnesses that may present with depression. Mr. Z’s presentation warrants investigating these tests. His history of traumatic splenectomy and night sweats suggests an infection. The team’s initial recommendations include laboratory tests, discontinuing divalproex because it may be causing thrombocytopenia, and decreasing risperidone to 2 mg/d to improve his fatigue and possibly developed extrapyramidal symptoms.

Table 2

Potential infectious causes of chronic encephalopathy

Type of infectionOrganism/disease
MycobacterialMycobacterium tuberculosis
SpirochetalTreponema pallidum (syphilis), Borrelia burgdorferi (Lyme disease), Leptospira
BacterialBrucella, Listeria, Nocardia, Actinomyces israelii, Whipple’s disease
ViralHIV/AIDS, cytomegalovirus, varicella zoster virus, herpes simplex virus, enterovirus
FungalHistoplasmosis, coccidiosis, sporothrix, Blastomyces, Cryptococcus
ParasiticToxoplasmosis, taenia solium (cysticercosis), Schistosoma, Acanthamoeba
AIDS: acquired immunodeficiency syndrome; HIV: human immunodeficiency virus
Source: Reference 8

Table 3

Differentiating medical illnesses that may mimic depression

Laboratory tests
  • CBC, thyroid-stimulating hormone, antinuclear antibody, erythrocyte sedimentation rate, vitamin B12, rapid plasma reagin, HIV test, electrolytes and calcium levels and renal function test, liver function tests, blood alcohol, blood, and urine toxicology screen, ABG, Lyme antibody test (ELISA), dexamethasone suppression test (Cushing’s disease), cosyntropin stimulation test (Addison’s disease)
Imaging studies
  • CT scan or MRI of the brain
Other tests
  • EEG
Procedures
  • Lumbar puncture for VDRL, Lyme antibody, cell count, chemistry, and protein electrophoresis
ABG: arterial blood gases; CBC: complete blood count;
EEG: electroencephalogram; ELISA: enzyme-linked immunosorbent assay; HIV: human immunodeficiency virus; VDRL: venereal disease research laboratory
Source: Reference 1

TREATMENT: Cause revealed

Mr. Z develops a persistent fever of 102°F with continuous profuse sweating and a hypotensive episode. Blood work reveals mild anemia, thrombocytopenia, and increased coagulation parameters with high D-dimer and low fibrinogen, consistent with diagnosis of disseminated intravascular coagulation (DIC) secondary to infectious etiology. Thyroid and HIV tests are negative. After further evaluation, Mr. Z remembers that 4 months earlier he removed several ticks from his legs after hunting; he also remembers experiencing shivering and night sweats several weeks before he was hospitalized. His blood smear is positive for babesiosis and further testing confirms positive Lyme antibodies. Mr. Z is started on aggressive hemodynamic stabilization and a pathogen-tailored course of antibiotics for several weeks. This results in improvement and discharge home in a stable condition. His depression and fatigue improve but do not fully remit by the time he is discharged.

The authors’ observations

Lyme disease is one of the fastest-growing infectious diseases in the United States.9 The prevalence of positive Lyme antibodies is 30% higher in psychiatric populations than the general population.10 Lyme disease is transmitted by deer tick bite, often undetected, that is infected with spirochete Borrelia burgdorferi. To be infectious, ticks need to be attached to the skin for 24 to 48 hours,11,12 although individual cases have reported transmission in <24 hours. The clinical manifestations of Lyme disease can be divided into 3 phases:

  • early localized phase, characterized by the distinctive skin lesion erythema migrans with or without constitutional symptoms
  • early disseminated phase, characterized by multiple erythema migrans lesions and neurologic and/or cardiac findings
  • late or chronic disease associated with intermittent/persistent arthritis and/or neurologic problems.11,13
 

 

The clinical features of each stage frequently overlap and some patients in a later stage of Lyme disease do not have prior signs or symptoms of the disease. Because it is a multisystem disease, Lyme disease can attack the CNS in the form of neuroborreliosis, a clinical diagnosis, without involving other systems, and its neuropsychiatric manifestations can resemble neurosyphilis because both organisms are spirochetes.11,13,14 CNS disorders have been found in up to 40% of Lyme disease cases.11 In neuroborreliosis, cognitive problems usually predominate; however, neuroborreliosis can mimic multiple brain diseases presenting with various neurologic and psychiatric symptoms (Table 4)14,15 and can present at any time after the tick bite. Furthermore neuroborreliosis is difficult to diagnose because symptoms may remain dormant and emerge after several years.11,13,14Borrelia burgdorferi is challenging to isolate and grow in the lab, and enzyme-linked immunosorbent assay (ELISA) testing for antibodies is highly specific but not very sensitive,16 frequently giving false negative results. Western blot confirms the diagnosis.

Table 4

Late-stage neuropsychiatric symptoms of Lyme disease

Cognitive problems, memory problems, forgetfulness, slowing of thought processing, dysfunction in visuospatial orientation, dyslexia
Depression
Mood swings
Psychosis
Violent behavior/irritability
OCD
Anxiety
Panic attacks
Sleep disorders
Seizures
ADHD-like symptoms
Autism-like behavior
Chronic fatigue syndrome
Fibromyalgia
ADHD: attention-deficit/hyperactivity disorder; OCD: obsessive-compulsive disorder
Source: References 14,15

Mr. Z’s presentation also reflects co-infection with babesiosis. Babesia is malaria-like protozoa diagnosed by blood smear that can cause a fatal illness in immuno-compromised patients. The clinical picture varies from mild symptoms such as night sweats, chills, arthralgias, and anorexia with thrombocytopenia to severe and potentially fatal outcomes in immunocompromised patients, including DIC, acute renal failure, sepsis, congestive heart failure, and myocardial infarction.11 Risk factors for the severest forms of babesiosis are age >50, co-infection with Lyme disease, and splenectomy,11 all of which were present in Mr. Z. Co-infected patients experience fatigue, headache, anorexia, and emotional lability more frequently than those with Lyme disease alone.12

Treatment options

Treatment of Lyme disease/neuroborreliosis is complex. The mainstay approach is antibiotics. Despite adequate treatment, many patients experience continued impairment, including chronic pain, fatigue, and cognitive and psychiatric symptoms.11,14 There is some evidence Borrelia burgdorferi can persist and re-emerge after adequate treatment.14,17 The National Institute of Health sponsored several clinical trials of prolonged antibiotic treatment for chronic Lyme disease. Some results suggested improvement in fatigue and cognitive function, although these results were not sustained.18

There is a strong link between mental illness and increased prevalence of positive Lyme disease antibodies.10 Several studies report increased risk of infection during psychological stress that may be related to an altered immune system response.19 Evidence suggests that Borrelia burgdorferi can alter immune system response, making T cells more reactive not only to Borrelia burgdorferi antigens but also to host antigens,20 creating autoimmune inflammatory reactions that could explain chronic neuropsychiatric symptoms. It appears Lyme disease antigens can mimic certain autoantigens (for example, in the thyroid gland).21 Whether there is a role for autoimmune therapy in treating chronic symptoms needs to be investigated.

Once Lyme disease is diagnosed, educating patients and families becomes an important part of treatment because many patients report feeling stigmatized by the diagnosis. Referral to a Lyme disease support group may be beneficial. Patients with neuropsychiatric symptoms that persist after antibiotic treatment should be offered symptom-based treatment, including medications and therapy.

Related Resource

Drug Brand Names

  • Divalproex sodium • Depakote
  • Levothyroxine • Levoxyl, Synthroid
  • Risperidone • Risperdal

Disclosure

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

Discuss this article at www.facebook.com/CurrentPsychiatry

Mr. Z, age 61, is referred by his primary care clinician to the hospital’s medical service with increasing depressive symptoms and non-pruritic rash. He has a history of bipolar I disorder for >30 years. When the primary care physician evaluated Mr. Z, his vitals were normal, but blood work revealed mild anemia and thrombocytopenia of 34 x103/μL, which prompted referral to the hospital. During admission, the psychiatric consultation service is called to evaluate Mr. Z’s depressive symptoms.

Mr. Z reports having chronic sleep problems and feeling cold and tired, shivering at times, but has no pain. He says he’s worried because he feels severely depressed, worthless, and hopeless, but denies suicidal ideation and psychosis. Mr. Z says he started experiencing increasingly depressed mood, anhedonia, insomnia, fatigue, poor appetite, and concentration 2 months ago. At that time his outpatient psychiatrist started Mr. Z on risperidone, 6 mg/d, and divalproex, 1,500 mg at bedtime because of emerging mood symptoms, after he was off medication for 7 months. Mr. Z attributed his worsened mood symptoms to being overwhelmed by several psychosocial stressors, including going through a complicated divorce, financial problems, and homelessness after being evicted from his apartment.

A review of Mr. Z’s psychiatric history reveals several remote hospitalizations—the last was 7 years ago—for escalated manic symptoms after he stopped taking his medication. He denies past suicide attempts. Mr. Z says he is compliant with his current medication regimen—risperidone, 6 mg/d, and divalproex, 1,500 mg at bedtime. He denies illicit drug use and says he drinks “a couple of beers, mostly on weekends.” Family history is positive for depression and bipolar II disorder.

His medical history is significant for hypothyroidism after goiter removal 6 years ago, for which he takes levothyroxine, 150 mcg/d, and a sports injury-related splenectomy in childhood. He reports no allergies. Vital signs at the time of admission are temperature, 99.1°F; pulse, 98 beats per minute; respiration, 16 breaths per minute; blood pressure, 123/73 mm/Hg; and oxygen saturation, 97%.

During the interview, Mr. Z presents with tired facies and exhibits psychomotor retardation. He has to force himself to stay engaged in the evaluation and maintain eye contact. His speech is clear, regular, and soft. Mr. Z says he is “very depressed”; his affect is constricted, almost flat, stable, and consistent with depressed mood. His thought process is linear and somewhat concrete and his thought content is notable for hopelessness, although Mr. Z continues to deny suicidal or homicidal ideations. No hallucinations or apparent delusions are noted. Insight and judgment are fair. Mr. Z understands his current mental state; however, he displays some lack of knowledge regarding his current hospitalization. Cognition is intact.

The authors’ observations

The differential diagnosis in patients presenting with mood changes is extensive (Table 1)1 and in Mr. Z’s case includes several precipitating and perpetuating factors. Mr. Z presents with severe depressive symptoms and meets DSM-IV-TR criteria for a major depressive episode (MDE). This presentation is not typical of his bipolar I disorder because Mr. Z has never experienced an MDE and usually presents with escalating hypomanic/manic symptoms in the context of medication nonadherence. Nevertheless, Mr. Z has several risk factors for severe depression, including a family psychiatric history, multiple enduring social stressors and life crises, and medical conditions.

In the general population, the lifetime risk for developing depression is 8% to 17%.2 The risk of developing a mood disorder increases significantly if a first-degree relative is diagnosed with a mood disorder; the relative risk is 10.3 for bipolar disorder and 3.2 for depression.3 Additionally, Mr. Z is going through a complicated divorce, has financial problems, and is homeless, all of which could trigger an MDE. Furthermore, hypothyroidism shares many symptoms of depression, including fatigue, lethargy, anhedonia, cold intolerance, and low mood; mental status changes frequently are the initial presentation of thyroid problems.4 Physicians started Mr. Z on a new medication regimen (risperidone and divalproex) to control mood instability, which coincided with symptom onset. Atypical antipsychotics have been reported to precipitate depressive symptoms; their side effect profile includes extrapyramidal effects, such as flat affect, which can be mistaken for depression.5 Rapid valproate titration can mimic neurovegetative symptoms of depression and cause dose-dependent thrombocytopenia and rash, which could explain his initial presentation.6 Finally, Mr. Z’s history of traumatic splenectomy, change in mental status, and thrombocytopenia suggest an infectious etiology.

Table 1

Differential diagnosis in patients presenting with mood changes

 

 

Cerebrovascular disease
Degenerative disorders (Parkinson’s disease, Huntington’s disease, Wilson’s disease)
Demyelinating disorders (multiple sclerosis, amyotrophic lateral sclerosis, lipid storage disease)
Endocrine disorders (Addison’s disease, Cushing’s disease, hyperthyroidism, hypothyroidism, hyperparathyroidism, pituitary dysfunction)
Epilepsy
Infectious diseases
Immune diseases
Metabolic encephalopathy
Neoplasm
Nutritional deficits (thiamine, niacin, vitamin B12)
Primary psychiatric disorders (mood disorders, dementia, sleep disorders)
Substance use
Toxins/medications
Traumatic brain injury
Source: Reference 1

Possible infectious causes

The increased prevalence of immune suppression due to human immunodeficiency virus/acquired immune deficiency syndrome (HIV/AIDS) or from therapeutic modalities such as cancer therapy or splenectomy has led to an increased number of chronic CNS infections, manifesting with an array of neuropsychiatric symptoms and nonspecific physiological reactions.1,7 Mr. Z complains of a 2-month period of worsening depression that could suggest an infectious process with an insidious onset. Some infectious agents that can cause chronic CNS infection and encephalopathy are presented in Table 2.8 HIV, tuberculosis, syphilis, Lyme disease, and herpes simplex virus are becoming more prevalent and can present with neuropsychiatric symptoms.1 For example, in addition to thrombocytopenia and low-grade fever, patients with HIV may exhibit a broad range of neuropsychiatric symptoms such as cognitive problems, impaired executive and motor functioning, sleep disturbance, and anxiety. These patients frequently present with low mood and neurovegetative symptoms of depression.7 Similarly, the same tick responsible for Lyme disease infection can transmit other infectious agents that can cause thrombocytopenia, including Babesia, Ehrlichia chaffeensis, Anaplasma phagocytophilum, and human Ewingii ehrlichiosis.

The authors’ observations

Diagnosis of a mood change, particularly an MDE, is clinical, based on careful psychiatric evaluation using standardized criteria rather than a specific lab test. However, some laboratory studies (Table 3)1 are useful in differentiating medical illnesses that may present with depression. Mr. Z’s presentation warrants investigating these tests. His history of traumatic splenectomy and night sweats suggests an infection. The team’s initial recommendations include laboratory tests, discontinuing divalproex because it may be causing thrombocytopenia, and decreasing risperidone to 2 mg/d to improve his fatigue and possibly developed extrapyramidal symptoms.

Table 2

Potential infectious causes of chronic encephalopathy

Type of infectionOrganism/disease
MycobacterialMycobacterium tuberculosis
SpirochetalTreponema pallidum (syphilis), Borrelia burgdorferi (Lyme disease), Leptospira
BacterialBrucella, Listeria, Nocardia, Actinomyces israelii, Whipple’s disease
ViralHIV/AIDS, cytomegalovirus, varicella zoster virus, herpes simplex virus, enterovirus
FungalHistoplasmosis, coccidiosis, sporothrix, Blastomyces, Cryptococcus
ParasiticToxoplasmosis, taenia solium (cysticercosis), Schistosoma, Acanthamoeba
AIDS: acquired immunodeficiency syndrome; HIV: human immunodeficiency virus
Source: Reference 8

Table 3

Differentiating medical illnesses that may mimic depression

Laboratory tests
  • CBC, thyroid-stimulating hormone, antinuclear antibody, erythrocyte sedimentation rate, vitamin B12, rapid plasma reagin, HIV test, electrolytes and calcium levels and renal function test, liver function tests, blood alcohol, blood, and urine toxicology screen, ABG, Lyme antibody test (ELISA), dexamethasone suppression test (Cushing’s disease), cosyntropin stimulation test (Addison’s disease)
Imaging studies
  • CT scan or MRI of the brain
Other tests
  • EEG
Procedures
  • Lumbar puncture for VDRL, Lyme antibody, cell count, chemistry, and protein electrophoresis
ABG: arterial blood gases; CBC: complete blood count;
EEG: electroencephalogram; ELISA: enzyme-linked immunosorbent assay; HIV: human immunodeficiency virus; VDRL: venereal disease research laboratory
Source: Reference 1

TREATMENT: Cause revealed

Mr. Z develops a persistent fever of 102°F with continuous profuse sweating and a hypotensive episode. Blood work reveals mild anemia, thrombocytopenia, and increased coagulation parameters with high D-dimer and low fibrinogen, consistent with diagnosis of disseminated intravascular coagulation (DIC) secondary to infectious etiology. Thyroid and HIV tests are negative. After further evaluation, Mr. Z remembers that 4 months earlier he removed several ticks from his legs after hunting; he also remembers experiencing shivering and night sweats several weeks before he was hospitalized. His blood smear is positive for babesiosis and further testing confirms positive Lyme antibodies. Mr. Z is started on aggressive hemodynamic stabilization and a pathogen-tailored course of antibiotics for several weeks. This results in improvement and discharge home in a stable condition. His depression and fatigue improve but do not fully remit by the time he is discharged.

The authors’ observations

Lyme disease is one of the fastest-growing infectious diseases in the United States.9 The prevalence of positive Lyme antibodies is 30% higher in psychiatric populations than the general population.10 Lyme disease is transmitted by deer tick bite, often undetected, that is infected with spirochete Borrelia burgdorferi. To be infectious, ticks need to be attached to the skin for 24 to 48 hours,11,12 although individual cases have reported transmission in <24 hours. The clinical manifestations of Lyme disease can be divided into 3 phases:

  • early localized phase, characterized by the distinctive skin lesion erythema migrans with or without constitutional symptoms
  • early disseminated phase, characterized by multiple erythema migrans lesions and neurologic and/or cardiac findings
  • late or chronic disease associated with intermittent/persistent arthritis and/or neurologic problems.11,13
 

 

The clinical features of each stage frequently overlap and some patients in a later stage of Lyme disease do not have prior signs or symptoms of the disease. Because it is a multisystem disease, Lyme disease can attack the CNS in the form of neuroborreliosis, a clinical diagnosis, without involving other systems, and its neuropsychiatric manifestations can resemble neurosyphilis because both organisms are spirochetes.11,13,14 CNS disorders have been found in up to 40% of Lyme disease cases.11 In neuroborreliosis, cognitive problems usually predominate; however, neuroborreliosis can mimic multiple brain diseases presenting with various neurologic and psychiatric symptoms (Table 4)14,15 and can present at any time after the tick bite. Furthermore neuroborreliosis is difficult to diagnose because symptoms may remain dormant and emerge after several years.11,13,14Borrelia burgdorferi is challenging to isolate and grow in the lab, and enzyme-linked immunosorbent assay (ELISA) testing for antibodies is highly specific but not very sensitive,16 frequently giving false negative results. Western blot confirms the diagnosis.

Table 4

Late-stage neuropsychiatric symptoms of Lyme disease

Cognitive problems, memory problems, forgetfulness, slowing of thought processing, dysfunction in visuospatial orientation, dyslexia
Depression
Mood swings
Psychosis
Violent behavior/irritability
OCD
Anxiety
Panic attacks
Sleep disorders
Seizures
ADHD-like symptoms
Autism-like behavior
Chronic fatigue syndrome
Fibromyalgia
ADHD: attention-deficit/hyperactivity disorder; OCD: obsessive-compulsive disorder
Source: References 14,15

Mr. Z’s presentation also reflects co-infection with babesiosis. Babesia is malaria-like protozoa diagnosed by blood smear that can cause a fatal illness in immuno-compromised patients. The clinical picture varies from mild symptoms such as night sweats, chills, arthralgias, and anorexia with thrombocytopenia to severe and potentially fatal outcomes in immunocompromised patients, including DIC, acute renal failure, sepsis, congestive heart failure, and myocardial infarction.11 Risk factors for the severest forms of babesiosis are age >50, co-infection with Lyme disease, and splenectomy,11 all of which were present in Mr. Z. Co-infected patients experience fatigue, headache, anorexia, and emotional lability more frequently than those with Lyme disease alone.12

Treatment options

Treatment of Lyme disease/neuroborreliosis is complex. The mainstay approach is antibiotics. Despite adequate treatment, many patients experience continued impairment, including chronic pain, fatigue, and cognitive and psychiatric symptoms.11,14 There is some evidence Borrelia burgdorferi can persist and re-emerge after adequate treatment.14,17 The National Institute of Health sponsored several clinical trials of prolonged antibiotic treatment for chronic Lyme disease. Some results suggested improvement in fatigue and cognitive function, although these results were not sustained.18

There is a strong link between mental illness and increased prevalence of positive Lyme disease antibodies.10 Several studies report increased risk of infection during psychological stress that may be related to an altered immune system response.19 Evidence suggests that Borrelia burgdorferi can alter immune system response, making T cells more reactive not only to Borrelia burgdorferi antigens but also to host antigens,20 creating autoimmune inflammatory reactions that could explain chronic neuropsychiatric symptoms. It appears Lyme disease antigens can mimic certain autoantigens (for example, in the thyroid gland).21 Whether there is a role for autoimmune therapy in treating chronic symptoms needs to be investigated.

Once Lyme disease is diagnosed, educating patients and families becomes an important part of treatment because many patients report feeling stigmatized by the diagnosis. Referral to a Lyme disease support group may be beneficial. Patients with neuropsychiatric symptoms that persist after antibiotic treatment should be offered symptom-based treatment, including medications and therapy.

Related Resource

Drug Brand Names

  • Divalproex sodium • Depakote
  • Levothyroxine • Levoxyl, Synthroid
  • Risperidone • Risperdal

Disclosure

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

References

1. Stern TA, Rosenbaum JF, Fava M, et al. eds. Massachusetts General Hospital comprehensive clinical psychiatry. Philadelphia, PA: Mosby; 2008;257-277.

2. Andrade L, Caraveo-Anduaga JJ, Berglund P, et al. The epidemiology of major depressive episodes: results from the International Consortium of Psychiatric Epidemiology (ICPE) Surveys. Int J Methods Psychiatr Res. 2003;12(1):3-21.

3. Merikangas KR, Low NC. The epidemiology of mood disorders. Curr Psychiatry Rep. 2004;6:411-421.

4. Brown GM. Psychiatric and neurologic aspects of endocrine disease. In Krieger DT Hughes JC, eds. Neuroendocrinology. Sunderland, MA: Sinauer Associates; 1980;185-194.

5. Maguire GA. Comprehensive understanding of schizophrenia and its treatment. Am J Health Syst Pharm. 2002;59(17 suppl 5):S4-S11.

6. Dreifuss FE, Langer DH. Side effects of valproate. Am J Med. 1988;84(1A):34-41.

7. Perry S, Jacobsen P. Neuropsychiatric manifestations of AIDS-spectrum disorders. Hosp Community Psychiatry. 1986;37:135-142.

8. Hildebrand J, Aoun M. Chronic meningitis: still a diagnostic challenge. J Neurol. 2003;250(6):653-660.

9. Bacon RM, Kugeler KJ, Mead PS. Centers for Disease Control and Prevention (CDC). Surveillance for Lyme disease—United States 1992-2006. MMWR Surveill Summ. 2008;57(10):1-9.

10. Hájek T, Pasková B, Janovská D, et al. Higher prevalence of antibodies to Borrelia burgdorferi in psychiatric patients than in healthy subjects. Am J Psychiatry. 2002;159:297-301.

11. Wormser GP, Dattwyler RJ, Shapiro ED, et al. The clinical assessment, treatment, and prevention of lyme disease, human granulocytic anaplasmosis, and babesiosis: clinical practice guidelines by the Infectious Diseases Society of America. Clin Infect Dis. 2006;43(9):1089-1134.

12. Krause PJ, Telford SR 3rd, Spielman A, et al. Concurrent Lyme disease and babesiosis. Evidence for increased severity and duration of illness. JAMA. 1996;275(21):1657-1660.

13. Hildenbrand P, Craven DE, Jones R, et al. Lyme neuroborreliosis: manifestations of a rapidly emerging zoonosis. AJNR Am J Neuroradiol. 2009;30:1079-1087.

14. Fallon BA, Nields JA. Lyme disease: a neuropsychiatric illness. Am J Psychiatry. 1994;151:1571-1583.

15. Fallon BA, Nields JA, Burrascano JJ, et al. The neuropsychiatric manifestations of Lyme borreliosis. Psychiatr Q. 1992;63(1):95-117.

16. Stricker RB, Johnson L. Lyme wars: let’s tackle the testing. BMJ. 2007;335:1008.-

17. Hodzic E, Feng S, Holden K, et al. Persistence of Borrelia burgdorferi following antibiotic treatment in mice. Antimicrob Agents Chemother. 2008;52(5):1728-1736.

18. Fallon BA, Keilp JG, Corbera KM, et al. A randomized, placebo-controlled trial of repeated IV antibiotic therapy for Lyme encephalopathy. Neurology. 2008;70(13):992-1003.

19. Sheridan JF, Dobbs C, Brown D, et al. Psychoneuroimmunology: stress effects on pathogenesis and immunity during infection. Clin Microbiol Rev. 1994;7(2):200-212.

20. Yssel H, Shanafelt MC, Soderberg C, et al. Borrelia burgdorferi activates a T helper 1-like T cell subset in Lyme arthritis. J Exp Med. 1991;174:593-601.

21. Benvenga S, Guarneri F, Vaccaro M, et al. Homologies between proteins of Borrelia burgdorferi and thyroid autoantigens. Thyroid. 2004;14(11):964-966.

References

1. Stern TA, Rosenbaum JF, Fava M, et al. eds. Massachusetts General Hospital comprehensive clinical psychiatry. Philadelphia, PA: Mosby; 2008;257-277.

2. Andrade L, Caraveo-Anduaga JJ, Berglund P, et al. The epidemiology of major depressive episodes: results from the International Consortium of Psychiatric Epidemiology (ICPE) Surveys. Int J Methods Psychiatr Res. 2003;12(1):3-21.

3. Merikangas KR, Low NC. The epidemiology of mood disorders. Curr Psychiatry Rep. 2004;6:411-421.

4. Brown GM. Psychiatric and neurologic aspects of endocrine disease. In Krieger DT Hughes JC, eds. Neuroendocrinology. Sunderland, MA: Sinauer Associates; 1980;185-194.

5. Maguire GA. Comprehensive understanding of schizophrenia and its treatment. Am J Health Syst Pharm. 2002;59(17 suppl 5):S4-S11.

6. Dreifuss FE, Langer DH. Side effects of valproate. Am J Med. 1988;84(1A):34-41.

7. Perry S, Jacobsen P. Neuropsychiatric manifestations of AIDS-spectrum disorders. Hosp Community Psychiatry. 1986;37:135-142.

8. Hildebrand J, Aoun M. Chronic meningitis: still a diagnostic challenge. J Neurol. 2003;250(6):653-660.

9. Bacon RM, Kugeler KJ, Mead PS. Centers for Disease Control and Prevention (CDC). Surveillance for Lyme disease—United States 1992-2006. MMWR Surveill Summ. 2008;57(10):1-9.

10. Hájek T, Pasková B, Janovská D, et al. Higher prevalence of antibodies to Borrelia burgdorferi in psychiatric patients than in healthy subjects. Am J Psychiatry. 2002;159:297-301.

11. Wormser GP, Dattwyler RJ, Shapiro ED, et al. The clinical assessment, treatment, and prevention of lyme disease, human granulocytic anaplasmosis, and babesiosis: clinical practice guidelines by the Infectious Diseases Society of America. Clin Infect Dis. 2006;43(9):1089-1134.

12. Krause PJ, Telford SR 3rd, Spielman A, et al. Concurrent Lyme disease and babesiosis. Evidence for increased severity and duration of illness. JAMA. 1996;275(21):1657-1660.

13. Hildenbrand P, Craven DE, Jones R, et al. Lyme neuroborreliosis: manifestations of a rapidly emerging zoonosis. AJNR Am J Neuroradiol. 2009;30:1079-1087.

14. Fallon BA, Nields JA. Lyme disease: a neuropsychiatric illness. Am J Psychiatry. 1994;151:1571-1583.

15. Fallon BA, Nields JA, Burrascano JJ, et al. The neuropsychiatric manifestations of Lyme borreliosis. Psychiatr Q. 1992;63(1):95-117.

16. Stricker RB, Johnson L. Lyme wars: let’s tackle the testing. BMJ. 2007;335:1008.-

17. Hodzic E, Feng S, Holden K, et al. Persistence of Borrelia burgdorferi following antibiotic treatment in mice. Antimicrob Agents Chemother. 2008;52(5):1728-1736.

18. Fallon BA, Keilp JG, Corbera KM, et al. A randomized, placebo-controlled trial of repeated IV antibiotic therapy for Lyme encephalopathy. Neurology. 2008;70(13):992-1003.

19. Sheridan JF, Dobbs C, Brown D, et al. Psychoneuroimmunology: stress effects on pathogenesis and immunity during infection. Clin Microbiol Rev. 1994;7(2):200-212.

20. Yssel H, Shanafelt MC, Soderberg C, et al. Borrelia burgdorferi activates a T helper 1-like T cell subset in Lyme arthritis. J Exp Med. 1991;174:593-601.

21. Benvenga S, Guarneri F, Vaccaro M, et al. Homologies between proteins of Borrelia burgdorferi and thyroid autoantigens. Thyroid. 2004;14(11):964-966.

Issue
Current Psychiatry - 10(10)
Issue
Current Psychiatry - 10(10)
Page Number
53-58
Page Number
53-58
Publications
Publications
Topics
Article Type
Display Headline
A curious case of depression
Display Headline
A curious case of depression
Legacy Keywords
Cases that test your skills; depression; Pavle Joksovic; Pieter Joost van Wattum; Catherine Chiles; bipolar I disorder; B; depression; fatigue; thrombocytopenia; rash; Lyme disease
Legacy Keywords
Cases that test your skills; depression; Pavle Joksovic; Pieter Joost van Wattum; Catherine Chiles; bipolar I disorder; B; depression; fatigue; thrombocytopenia; rash; Lyme disease
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Beyond lithium: Using psychotherapy to reduce suicide risk in bipolar disorder

Article Type
Changed
Tue, 12/11/2018 - 15:04
Display Headline
Beyond lithium: Using psychotherapy to reduce suicide risk in bipolar disorder

Discuss this article at www.facebook.com/CurrentPsychiatry

Patients with bipolar disorder (BD) have a high risk for suicidal ideation, suicide attempts, and suicide.1-3 Approximately 25% to 50% of BD patients attempt suicide at least once, and their attempts often are lethal—the ratio of attempts to completed suicides in BD patients is 3:1, compared with 30:1 in the general population.4 Lithium has been shown to effectively stabilize BD patients’ mood and significantly reduce the rates of suicide attempts and completed suicides,5-9 but does not reduce BD patients’ long-term suicide risk to that of the general population.

Literature on psychotherapeutic treatments for patients with BD primarily focuses on improving patients’ adherence to pharmacotherapy and achieving faster recovery and remission.10 Nonpharmacologic treatments for patients with BD include psychoeducation, family-focused psychoeducation, cognitive therapy, and interpersonal and social rhythm therapy (Table 1).11 Literature on nonpharmacologic treatments to address suicidality in BD patients is limited,12,13 and additional psychotherapeutic interventions to reduce suicide risk in BD patients are needed.14

In this article, I describe a novel psychotherapeutic intervention I use that integrates cognitive therapy principles with ideas derived from the psychosynthesis model.15,16 It consists of teaching patients to “disidentify” from suicidal thoughts, followed by a guided-imagery exercise in which patients experience a future positive life event with all 5 senses and internalize this experience. This creates a “hook into the future” that changes the present to match the future event and acts as an antidote to suicidal thoughts. I have used this strategy successfully in many patients as an adjunct to pharmacotherapy.

Table 1

Nonpharmacologic interventions for bipolar disorder

GoalsTechniques
Psychoeducation
  • Increase illness awareness
  • Improve medication compliance
  • Early detection of relapses
  • Establish lifestyle regularity
  • Education based on books, pamphlets, and Web sites regarding the symptoms, course, treatment, and self-management of BD
  • Self-monitoring of symptoms and behaviors
  • Discussion
Family-focused psychoeducation
  • Accept notion of vulnerability for future episodes
  • Accept need for mood-stabilizing medications
  • Educate to distinguish between patient’s personality traits and BD symptoms
  • Reestablish functional relationships after a mood episode
  • Education based on books, pamphlets, and Web sites regarding the symptoms, course, treatment, and self-management of BD
  • Enhance communication skills in the family
  • Education about problem-solving skills
Cognitive therapy
  • Challenge the patient’s dysfunctional thoughts and beliefs regarding self and the world as influenced by BD
  • Self-monitoring of dysfunctional thoughts and behaviors
  • Monitor moods and early signs of relapse
  • Develop a plan of action to deal with early signs of relapse
  • Emphasize the need of combined pharmacotherapy and psychotherapy
  • Promote the importance of regular sleep and healthy lifestyle
Interpersonal and social rhythm therapy
  • Stabilize daily routines and sleep/wake cycles
  • Gain insight into relationship between moods and interpersonal events
  • Relieve stress and interpersonal problems
  • Review history of illness
  • Track and identify connections between sleep patterns, activities, and mood
  • Develop a plan to stabilize social and circadian rhythms by maintaining consistent sleep/wake times and reducing excessive social stimulations
  • Explore and resolve interpersonal problems
BD: bipolar disorder
Source: Reference 11

A theoretical model

Roberto Assagioli, who established the approach to psychology called psychosynthesis, formulated a fundamental psychological principle in controlling one’s behavior: “We are dominated by everything with which our self becomes identified. We can dominate and control everything from which we disidentify ourselves.”15 According to the psychosynthesis model, it is easier to change thoughts we identify as foreign to “the self” (ego-dystonic) than thoughts we identify as being part of “the self” (ego-syntonic).

Patients whose suicidal thoughts are ego-syntonic identify with the thoughts as representing themselves and take ownership of these thoughts. Such patients are at a greater risk of acting on suicidal thoughts.

Patients whose suicidal thoughts are ego-dystonic consider the suicidal thoughts foreign to their core self and do not believe such thoughts represent them. In essence, they “disown” the thoughts and typically want to control and eliminate them. Examples of patients’ ego-syntonic vs ego-dystonic suicidal thoughts are listed in Table 2.

This construct calls for an intervention to help patients who have ego-syntonic suicidal thoughts restructure them as a manifestation of BD, rather then the patient’s core self belief. The intervention emphasizes the patient is not “a suicidal patient” but suffers from an illness that may manifest with suicidal ideation. Many BD patients overly identify with their disease, stating, “I am bipolar” or “I am suicidal.” The “I am” statement originates from the verb “to be,” which implies the disease is part of the patient’s identity. The goal of this intervention is to help the patient learn to disidentify from the disease and decide that suicidal thoughts do not represent their core self, but are a manifestation of the underlying disease.

 

 

The psychosynthesis model of helping patients disidentify and therefore disown suicidal thoughts is compatible with interventions that use mindfulness-based cognitive therapy training to teach patients to experience their thoughts as just passing through their consciousness without taking ownership of them.17

Table 2

Examples of ego-syntonic vs ego-dystonic suicidal thoughts

Ego-syntonicEgo-dystonic
‘I want to be dead. I found a simple and sure way to do it’‘I am having suicidal thoughts again and I don’t like it’
‘I know my family will be better off without me’‘I’m afraid the illness is coming back. I can’t stop these images’
‘Life is too hard, too much pain. I just want to end it all’‘I see my body in a coffin. It scares the hell out of me’
‘I’ve come to the end, life for me is over and done’‘I don’t want to die. Please help me get well again’
‘I know my life is over. I just have to find the right way to do it’‘It is as if a part of me wants to die but the rest of me wants to live’
‘Nobody cares about me. It is as if I am already dead’‘I know my family needs me. I want to be there for them’
‘I have nothing to live for’‘I have so much to live for, why am I having such crazy thoughts?’

The intervention

Assessment of suicidality is a fundamental skill for every mental health clinician.18 The psychotherapeutic intervention I use integrates the cognitive therapy principles of reframing, relabeling, and restructuring patients’ thoughts with disidentification from dysfunctional thoughts, feelings, and desires, based on psychosynthesis principles.

First, I conduct a comprehensive mental status examination that includes an in-depth exploration of the patient’s suicidal thoughts to determine if they are ego-syntonic or ego-dystonic. I begin by asking patients to clarify and elaborate on their statements referring to suicide, asking questions such as “Is there a part of you that objects to these thoughts?” and “Is there a part of you that wants to live?” If a patient indicates that he or she does experience inner conflict regarding such thoughts, these thoughts are classified as ego-dystonic. If a patient does not have any counter thoughts regarding the suicidal thoughts and fully identifies with them, the thoughts are classified as ego-syntonic.

I follow this with a treatment plan that helps patients change their view of their suicidal thoughts. I ask the patient to change these suicidal thoughts to ego-dystonic by focusing on the following statement: “I, (patient’s name), am a human being and like all human beings, I have thoughts; however, I am not my thoughts, I am much more than that.” I ask my patient to read this out loud and to mindfully meditate on this statement several times a day to reinforce the new understanding that these suicidal thoughts are a manifestation of the chemical imbalance of the mood disorder, and do not represent the patient as a person.

This intervention is paired with a future-focused internalized imagery experience I have described in previous articles.19,20 In this part of the treatment, the patient and I discuss a specific expected life milestone that is positive and for which he or she would want to be present (eg, children graduating from high school or college, a wedding, birth of a child/grandchild, etc.). Using guided imagery, the patient experiences this event with all 5 senses during the session. I instruct the patient to internalize the experience and bring it back from the future to the present. This creates a “hook into the future” that is coupled with this desired milestone event in the patient’s life.

The following 3 case studies provide examples of the application of this treatment intervention.

CASE 1: Disidentifying family history

Mrs. G, a 42-year-old mother of 2, suffers from bipolar II disorder with recurrent episodes of depression associated with ego-syntonic suicidal thoughts. She states that at times she feels she is a burden to her husband and children and believes they may be better off without her. She says she believes “ending it all” must be her destiny. After further investigation, I learn Mrs. G has a family history of BD and 3 relatives have committed suicide. This family history may partially explain her belief that suicide must be “in her genes.”

I discuss with Mrs. G the strategy of changing her thoughts. I tell her to write in her journal—which she brings to her sessions—the following statements: “I am a human being. I am an adult woman and mother of 2 children. I know I have thoughts but I am not my thoughts, I am much more than that. I know I have genes but I am not my genes, I am much more than that. I know I have feelings, but I am not my feelings, I am much more than that. I know I have cousins, uncles, aunts, and other relatives but I am not my relatives. I am uniquely myself, different from the others.”

 

 

I ask Mrs. G to read these statements out loud and repeat them several times a day to reinforce this new way of perceiving the suicidal thoughts and to disidentify from the thoughts and her family history as it relates to suicide.

Mrs. G and I talk about the future and expected family milestones. When I ask if her son would want her to be present at his college graduation, she says yes. We then discuss in detail the date, time of day, and location of this event, followed by a guided imagery exercise focused on the graduation. She is guided to experience this event with all 5 senses and describes the event in detail, including the expression on the faces of her husband and children, their voices, and the scent of their aftershave lotion. She hears her son saying, “Mom, I love you. Thank you for being there with me all these years. I could not have done it without you.” I ask Mrs. G to internalize these experiences and carve them into her memory. She is instructed to come back from this future-focused guided imagery experience. When her eyes open, she looks at me and describes her experience in great detail, at times using the past tense, which confirms that the future-focused event was internalized.

In her next session, Mrs. G reports an improvement in her sleep and a change in her suicidal thoughts, which now are only fleeting.

CASE 2: Experiencing graduation

Ms. J, age 17, was diagnosed with bipolar I disorder when she was 15. She has a family history of BD in her mother, 2 maternal aunts, her grandmother, and an older sister. All these women have a history of suicidal thoughts and suicide attempts requiring hospital treatment, but no completed suicides.

Ms. J has been taking an adequate combination of mood stabilizers. She has recovered from 2 previous depressive episodes and is experiencing a third relapse with suicidal thoughts. At times, she experiences these thoughts as ego-syntonic; at other times, they are ego-dystonic.

I first educate her about the nature of BD, explaining that her suicidal thoughts are a manifestation of a chemical imbalance in her brain as a result of the depressive relapse. I teach her to use guided imagery to focus on her favorite place of peace and serenity, the beach, which produces immediate relief of the intense anxiety she felt.

After we complete the disidentification exercise, I ask her to focus on her high school graduation ceremony, which is scheduled to take place in 1 year. In a state of guided imagery, she experiences her graduation from high school with all 5 senses. As she returns to a state of full alertness with her eyes open, she describes the graduation ceremony experience in detail using the past tense, as if it had already occurred, thereby creating her own hook into the future. I instruct her to write about this experience in her journal and bring it with her to the next session.

The following session, Ms. J reports that her suicide ideations have “disappeared.” She says this was accompanied by improvements in her overall mood and sleep.

CASE 3: Internalizing the future

Mr. C, a 38-year-old married father of 4 children, has bipolar II disorder and is in a depressed state. He has been treated with optimal doses of mood stabilizers and atypical antipsychotics but continues to have suicidal thoughts. These thoughts are at times ego-syntonic; he says, “My family would be better off without me.” When Mr. C’s mood improves, however, the suicidal thoughts become more ego-dystonic; he expresses fear that he might act out on the thoughts and states that he does not want to die, he really wants to live and get better. He has no history of suicide attempts.

During our session, I ask Mr. C to focus on a new perspective to understand his thoughts by repeating the following statements: “I, JC, am a human being. I know I have a bipolar mood disorder; however, I must remember I am not bipolar. I have suicidal thoughts; however, I am not my thoughts, I am much more than that. I know I want to live, to heal, and to get better. I want to be alive and well so I can see and participate in my children’s graduation from high school and be there when they get married and when my grandchildren are born.”

 

 

I teach Mr. C to use guided imagery, during which he experiences such future positive images and milestones in his life in all 5 senses and internalizes them by using the “back from the future” technique.17 By the end of the session, he reports feeling better, more hopeful, and confident in his abilities to control his suicidal thoughts. I instruct him to write in his diary about his experiences with the future-focused positive milestones and to bring this assignment to his next appointment.

At his next appointment, Mr. C reports that his suicidal thoughts have become more fleeting, lasting for 10 to 30 seconds, and then spontaneously change to focus on issues of the “here and now.” When I ask him to read what he’s written, what stands out is the use of past tense verbs to describe future-focused experiences. For me, this confirms that Mr. C has internalized the future, creating the desirable “future hook” that acts as an antidote to the suicidal thoughts.

Related Resources

  • Rouget BW, Aubry JM. Efficacy of psychoeducational approaches on bipolar disorders: a review of the literature. J Affect Disord. 2007;98:11-27.
  • Weinberg I, Ronningstam E, Goldblatt MJ, et al. Strategies in treatment of suicidality: identification of common and treatment-specific interventions in empirically supported treatment manuals. J Clin Psychiatry. 2010;71:699-706.

Drug Brand Name

  • Lithium • Eskalith, Lithobid

Disclosure

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

References

1. McIntyre RS, Konarski JZ. Bipolar disorder: a national health concern. CNS Spectr. 2004;9(11 suppl 12):6-15.

2. Tsai SY, Lee CH, Kuo CJ, et al. A retrospective analysis of risk and protective factors for natural death in bipolar disorder. J Clin Psychiatry. 2005;66(12):1586-1591.

3. Osby U, Brandt L, Correia N, et al. Excess mortality in bipolar and unipolar disorder in Sweden. Arch Gen Psychiatry. 2001;58(9):844-850.

4. Baldessarini RJ, Pompili M, Tondo L. Suicide in bipolar disorder: risks and management. CNS Spectr. 2006;11(6):465-471.

5. Gelenberg AJ. Can lithium help to prevent suicide? Acta Psychiatr Scand. 2001;104(3):161-162.

6. Schou M. Suicidal behavior and prophylactic lithium treatment of major mood disorders: a review of reviews. Suicide Life Threat Behav. 2001;30(3):289-293.

7. Burgess S, Geddes J, Hawton K, et al. Lithium for maintenance treatment of mood disorders. Cochrane Database Syst Rev. 2001;(3):CD003013.-

8. Baldessarini RJ, Tondo L, Davis P, et al. Decreased risk of suicides and attempts during long-term lithium treatment: a meta-analytic review. Bipolar Disord. 2006;8(5 Pt 2):625-639.

9. Tondo L, Baldessarini RJ. Long-term lithium treatment in the prevention of suicidal behavior in bipolar disorder patients. Epidemiol Psichiatr Soc. 2009;18(3):179-183.

10. Miklowitz DJ. Adjunctive psychotherapy for bipolar disorder: state of the evidence. Am J Psychiatry. 2008;165(11):1408-1419.

11. Hirschfeld RMA, Harris TH, Davis HK. Making efficacious choices: the integration of pharmacotherapy and nonpharmacologic approaches to the treatment of patients with bipolar disorder. Current Psychiatry. 2009;8(10 suppl):S6-S11.

12. Rucci P, Frank E, Kostelnik B, et al. Suicide attempts in patients with bipolar I disorder during acute and maintenance phases of intensive treatment with pharmacotherapy and adjunctive psychotherapy. Am J Psychiatry. 2002;159(7):1160-1164.

13. Fountoulakis KN, Gonda X, Siamouli M, et al. Psychotherapeutic intervention and suicide risk reduction in bipolar disorder: a review of the evidence. J Affect Disord. 2009;113(1-2):21-29.

14. Pompili M, Rihmer Z, Innamorati M, et al. Assessment and treatment of suicide risk in bipolar disorders. Expert Rev Neurother. 2009;9(1):109-136.

15. Assagioli R. Psychosynthesis: a collection of basic writings. New York NY: The Viking Press Inc.; 1965.

16. Assagioli R. The act of will. New York NY: The Viking Press Inc.; 1973.

17. Williams JM, Alatiq Y, Crane C, et al. Mindfulness-based cognitive therapy (MBCT) in bipolar disorder: preliminary evaluation of immediate effects on between-episode functioning. J Affect Disord. 2008;107(1-3):275-279.

18. Shea SC. The delicate art of eliciting suicidal ideation. Psychiatric Annals. 2004;34:385-400.

19. Torem MS. “Back from the future”: a powerful age-progression technique. Am J Clin Hypn. 1992;35(2):81-88.

20. Torem MS. Treating depression: a remedy from the future. In: Yapko MD ed. Hypnosis and treating depression: applications in clinical practice. New York, NY: Routledge; 2006:97–119.

Article PDF
Author and Disclosure Information

Moshe S. Torem, MD, DLFAPA
Professor of Psychiatry, Northeast Ohio Medical University, Rootstown, OH, Chief, Integrative Medicine, Akron General Medical Center, Medical Director, Center for Mind-Body Medicine, Akron, OH

Issue
Current Psychiatry - 10(10)
Publications
Topics
Page Number
39-45
Legacy Keywords
lithium; psychotherapy; suicide risk; reduce; bipolar disorder; BD; disown thoughts; internalize future; suicide ideation; suicide attempts; suicide; psychotheraputic treatments; nonpharmacologic interventions; Moshe Torem
Sections
Author and Disclosure Information

Moshe S. Torem, MD, DLFAPA
Professor of Psychiatry, Northeast Ohio Medical University, Rootstown, OH, Chief, Integrative Medicine, Akron General Medical Center, Medical Director, Center for Mind-Body Medicine, Akron, OH

Author and Disclosure Information

Moshe S. Torem, MD, DLFAPA
Professor of Psychiatry, Northeast Ohio Medical University, Rootstown, OH, Chief, Integrative Medicine, Akron General Medical Center, Medical Director, Center for Mind-Body Medicine, Akron, OH

Article PDF
Article PDF

Discuss this article at www.facebook.com/CurrentPsychiatry

Patients with bipolar disorder (BD) have a high risk for suicidal ideation, suicide attempts, and suicide.1-3 Approximately 25% to 50% of BD patients attempt suicide at least once, and their attempts often are lethal—the ratio of attempts to completed suicides in BD patients is 3:1, compared with 30:1 in the general population.4 Lithium has been shown to effectively stabilize BD patients’ mood and significantly reduce the rates of suicide attempts and completed suicides,5-9 but does not reduce BD patients’ long-term suicide risk to that of the general population.

Literature on psychotherapeutic treatments for patients with BD primarily focuses on improving patients’ adherence to pharmacotherapy and achieving faster recovery and remission.10 Nonpharmacologic treatments for patients with BD include psychoeducation, family-focused psychoeducation, cognitive therapy, and interpersonal and social rhythm therapy (Table 1).11 Literature on nonpharmacologic treatments to address suicidality in BD patients is limited,12,13 and additional psychotherapeutic interventions to reduce suicide risk in BD patients are needed.14

In this article, I describe a novel psychotherapeutic intervention I use that integrates cognitive therapy principles with ideas derived from the psychosynthesis model.15,16 It consists of teaching patients to “disidentify” from suicidal thoughts, followed by a guided-imagery exercise in which patients experience a future positive life event with all 5 senses and internalize this experience. This creates a “hook into the future” that changes the present to match the future event and acts as an antidote to suicidal thoughts. I have used this strategy successfully in many patients as an adjunct to pharmacotherapy.

Table 1

Nonpharmacologic interventions for bipolar disorder

GoalsTechniques
Psychoeducation
  • Increase illness awareness
  • Improve medication compliance
  • Early detection of relapses
  • Establish lifestyle regularity
  • Education based on books, pamphlets, and Web sites regarding the symptoms, course, treatment, and self-management of BD
  • Self-monitoring of symptoms and behaviors
  • Discussion
Family-focused psychoeducation
  • Accept notion of vulnerability for future episodes
  • Accept need for mood-stabilizing medications
  • Educate to distinguish between patient’s personality traits and BD symptoms
  • Reestablish functional relationships after a mood episode
  • Education based on books, pamphlets, and Web sites regarding the symptoms, course, treatment, and self-management of BD
  • Enhance communication skills in the family
  • Education about problem-solving skills
Cognitive therapy
  • Challenge the patient’s dysfunctional thoughts and beliefs regarding self and the world as influenced by BD
  • Self-monitoring of dysfunctional thoughts and behaviors
  • Monitor moods and early signs of relapse
  • Develop a plan of action to deal with early signs of relapse
  • Emphasize the need of combined pharmacotherapy and psychotherapy
  • Promote the importance of regular sleep and healthy lifestyle
Interpersonal and social rhythm therapy
  • Stabilize daily routines and sleep/wake cycles
  • Gain insight into relationship between moods and interpersonal events
  • Relieve stress and interpersonal problems
  • Review history of illness
  • Track and identify connections between sleep patterns, activities, and mood
  • Develop a plan to stabilize social and circadian rhythms by maintaining consistent sleep/wake times and reducing excessive social stimulations
  • Explore and resolve interpersonal problems
BD: bipolar disorder
Source: Reference 11

A theoretical model

Roberto Assagioli, who established the approach to psychology called psychosynthesis, formulated a fundamental psychological principle in controlling one’s behavior: “We are dominated by everything with which our self becomes identified. We can dominate and control everything from which we disidentify ourselves.”15 According to the psychosynthesis model, it is easier to change thoughts we identify as foreign to “the self” (ego-dystonic) than thoughts we identify as being part of “the self” (ego-syntonic).

Patients whose suicidal thoughts are ego-syntonic identify with the thoughts as representing themselves and take ownership of these thoughts. Such patients are at a greater risk of acting on suicidal thoughts.

Patients whose suicidal thoughts are ego-dystonic consider the suicidal thoughts foreign to their core self and do not believe such thoughts represent them. In essence, they “disown” the thoughts and typically want to control and eliminate them. Examples of patients’ ego-syntonic vs ego-dystonic suicidal thoughts are listed in Table 2.

This construct calls for an intervention to help patients who have ego-syntonic suicidal thoughts restructure them as a manifestation of BD, rather then the patient’s core self belief. The intervention emphasizes the patient is not “a suicidal patient” but suffers from an illness that may manifest with suicidal ideation. Many BD patients overly identify with their disease, stating, “I am bipolar” or “I am suicidal.” The “I am” statement originates from the verb “to be,” which implies the disease is part of the patient’s identity. The goal of this intervention is to help the patient learn to disidentify from the disease and decide that suicidal thoughts do not represent their core self, but are a manifestation of the underlying disease.

 

 

The psychosynthesis model of helping patients disidentify and therefore disown suicidal thoughts is compatible with interventions that use mindfulness-based cognitive therapy training to teach patients to experience their thoughts as just passing through their consciousness without taking ownership of them.17

Table 2

Examples of ego-syntonic vs ego-dystonic suicidal thoughts

Ego-syntonicEgo-dystonic
‘I want to be dead. I found a simple and sure way to do it’‘I am having suicidal thoughts again and I don’t like it’
‘I know my family will be better off without me’‘I’m afraid the illness is coming back. I can’t stop these images’
‘Life is too hard, too much pain. I just want to end it all’‘I see my body in a coffin. It scares the hell out of me’
‘I’ve come to the end, life for me is over and done’‘I don’t want to die. Please help me get well again’
‘I know my life is over. I just have to find the right way to do it’‘It is as if a part of me wants to die but the rest of me wants to live’
‘Nobody cares about me. It is as if I am already dead’‘I know my family needs me. I want to be there for them’
‘I have nothing to live for’‘I have so much to live for, why am I having such crazy thoughts?’

The intervention

Assessment of suicidality is a fundamental skill for every mental health clinician.18 The psychotherapeutic intervention I use integrates the cognitive therapy principles of reframing, relabeling, and restructuring patients’ thoughts with disidentification from dysfunctional thoughts, feelings, and desires, based on psychosynthesis principles.

First, I conduct a comprehensive mental status examination that includes an in-depth exploration of the patient’s suicidal thoughts to determine if they are ego-syntonic or ego-dystonic. I begin by asking patients to clarify and elaborate on their statements referring to suicide, asking questions such as “Is there a part of you that objects to these thoughts?” and “Is there a part of you that wants to live?” If a patient indicates that he or she does experience inner conflict regarding such thoughts, these thoughts are classified as ego-dystonic. If a patient does not have any counter thoughts regarding the suicidal thoughts and fully identifies with them, the thoughts are classified as ego-syntonic.

I follow this with a treatment plan that helps patients change their view of their suicidal thoughts. I ask the patient to change these suicidal thoughts to ego-dystonic by focusing on the following statement: “I, (patient’s name), am a human being and like all human beings, I have thoughts; however, I am not my thoughts, I am much more than that.” I ask my patient to read this out loud and to mindfully meditate on this statement several times a day to reinforce the new understanding that these suicidal thoughts are a manifestation of the chemical imbalance of the mood disorder, and do not represent the patient as a person.

This intervention is paired with a future-focused internalized imagery experience I have described in previous articles.19,20 In this part of the treatment, the patient and I discuss a specific expected life milestone that is positive and for which he or she would want to be present (eg, children graduating from high school or college, a wedding, birth of a child/grandchild, etc.). Using guided imagery, the patient experiences this event with all 5 senses during the session. I instruct the patient to internalize the experience and bring it back from the future to the present. This creates a “hook into the future” that is coupled with this desired milestone event in the patient’s life.

The following 3 case studies provide examples of the application of this treatment intervention.

CASE 1: Disidentifying family history

Mrs. G, a 42-year-old mother of 2, suffers from bipolar II disorder with recurrent episodes of depression associated with ego-syntonic suicidal thoughts. She states that at times she feels she is a burden to her husband and children and believes they may be better off without her. She says she believes “ending it all” must be her destiny. After further investigation, I learn Mrs. G has a family history of BD and 3 relatives have committed suicide. This family history may partially explain her belief that suicide must be “in her genes.”

I discuss with Mrs. G the strategy of changing her thoughts. I tell her to write in her journal—which she brings to her sessions—the following statements: “I am a human being. I am an adult woman and mother of 2 children. I know I have thoughts but I am not my thoughts, I am much more than that. I know I have genes but I am not my genes, I am much more than that. I know I have feelings, but I am not my feelings, I am much more than that. I know I have cousins, uncles, aunts, and other relatives but I am not my relatives. I am uniquely myself, different from the others.”

 

 

I ask Mrs. G to read these statements out loud and repeat them several times a day to reinforce this new way of perceiving the suicidal thoughts and to disidentify from the thoughts and her family history as it relates to suicide.

Mrs. G and I talk about the future and expected family milestones. When I ask if her son would want her to be present at his college graduation, she says yes. We then discuss in detail the date, time of day, and location of this event, followed by a guided imagery exercise focused on the graduation. She is guided to experience this event with all 5 senses and describes the event in detail, including the expression on the faces of her husband and children, their voices, and the scent of their aftershave lotion. She hears her son saying, “Mom, I love you. Thank you for being there with me all these years. I could not have done it without you.” I ask Mrs. G to internalize these experiences and carve them into her memory. She is instructed to come back from this future-focused guided imagery experience. When her eyes open, she looks at me and describes her experience in great detail, at times using the past tense, which confirms that the future-focused event was internalized.

In her next session, Mrs. G reports an improvement in her sleep and a change in her suicidal thoughts, which now are only fleeting.

CASE 2: Experiencing graduation

Ms. J, age 17, was diagnosed with bipolar I disorder when she was 15. She has a family history of BD in her mother, 2 maternal aunts, her grandmother, and an older sister. All these women have a history of suicidal thoughts and suicide attempts requiring hospital treatment, but no completed suicides.

Ms. J has been taking an adequate combination of mood stabilizers. She has recovered from 2 previous depressive episodes and is experiencing a third relapse with suicidal thoughts. At times, she experiences these thoughts as ego-syntonic; at other times, they are ego-dystonic.

I first educate her about the nature of BD, explaining that her suicidal thoughts are a manifestation of a chemical imbalance in her brain as a result of the depressive relapse. I teach her to use guided imagery to focus on her favorite place of peace and serenity, the beach, which produces immediate relief of the intense anxiety she felt.

After we complete the disidentification exercise, I ask her to focus on her high school graduation ceremony, which is scheduled to take place in 1 year. In a state of guided imagery, she experiences her graduation from high school with all 5 senses. As she returns to a state of full alertness with her eyes open, she describes the graduation ceremony experience in detail using the past tense, as if it had already occurred, thereby creating her own hook into the future. I instruct her to write about this experience in her journal and bring it with her to the next session.

The following session, Ms. J reports that her suicide ideations have “disappeared.” She says this was accompanied by improvements in her overall mood and sleep.

CASE 3: Internalizing the future

Mr. C, a 38-year-old married father of 4 children, has bipolar II disorder and is in a depressed state. He has been treated with optimal doses of mood stabilizers and atypical antipsychotics but continues to have suicidal thoughts. These thoughts are at times ego-syntonic; he says, “My family would be better off without me.” When Mr. C’s mood improves, however, the suicidal thoughts become more ego-dystonic; he expresses fear that he might act out on the thoughts and states that he does not want to die, he really wants to live and get better. He has no history of suicide attempts.

During our session, I ask Mr. C to focus on a new perspective to understand his thoughts by repeating the following statements: “I, JC, am a human being. I know I have a bipolar mood disorder; however, I must remember I am not bipolar. I have suicidal thoughts; however, I am not my thoughts, I am much more than that. I know I want to live, to heal, and to get better. I want to be alive and well so I can see and participate in my children’s graduation from high school and be there when they get married and when my grandchildren are born.”

 

 

I teach Mr. C to use guided imagery, during which he experiences such future positive images and milestones in his life in all 5 senses and internalizes them by using the “back from the future” technique.17 By the end of the session, he reports feeling better, more hopeful, and confident in his abilities to control his suicidal thoughts. I instruct him to write in his diary about his experiences with the future-focused positive milestones and to bring this assignment to his next appointment.

At his next appointment, Mr. C reports that his suicidal thoughts have become more fleeting, lasting for 10 to 30 seconds, and then spontaneously change to focus on issues of the “here and now.” When I ask him to read what he’s written, what stands out is the use of past tense verbs to describe future-focused experiences. For me, this confirms that Mr. C has internalized the future, creating the desirable “future hook” that acts as an antidote to the suicidal thoughts.

Related Resources

  • Rouget BW, Aubry JM. Efficacy of psychoeducational approaches on bipolar disorders: a review of the literature. J Affect Disord. 2007;98:11-27.
  • Weinberg I, Ronningstam E, Goldblatt MJ, et al. Strategies in treatment of suicidality: identification of common and treatment-specific interventions in empirically supported treatment manuals. J Clin Psychiatry. 2010;71:699-706.

Drug Brand Name

  • Lithium • Eskalith, Lithobid

Disclosure

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

Discuss this article at www.facebook.com/CurrentPsychiatry

Patients with bipolar disorder (BD) have a high risk for suicidal ideation, suicide attempts, and suicide.1-3 Approximately 25% to 50% of BD patients attempt suicide at least once, and their attempts often are lethal—the ratio of attempts to completed suicides in BD patients is 3:1, compared with 30:1 in the general population.4 Lithium has been shown to effectively stabilize BD patients’ mood and significantly reduce the rates of suicide attempts and completed suicides,5-9 but does not reduce BD patients’ long-term suicide risk to that of the general population.

Literature on psychotherapeutic treatments for patients with BD primarily focuses on improving patients’ adherence to pharmacotherapy and achieving faster recovery and remission.10 Nonpharmacologic treatments for patients with BD include psychoeducation, family-focused psychoeducation, cognitive therapy, and interpersonal and social rhythm therapy (Table 1).11 Literature on nonpharmacologic treatments to address suicidality in BD patients is limited,12,13 and additional psychotherapeutic interventions to reduce suicide risk in BD patients are needed.14

In this article, I describe a novel psychotherapeutic intervention I use that integrates cognitive therapy principles with ideas derived from the psychosynthesis model.15,16 It consists of teaching patients to “disidentify” from suicidal thoughts, followed by a guided-imagery exercise in which patients experience a future positive life event with all 5 senses and internalize this experience. This creates a “hook into the future” that changes the present to match the future event and acts as an antidote to suicidal thoughts. I have used this strategy successfully in many patients as an adjunct to pharmacotherapy.

Table 1

Nonpharmacologic interventions for bipolar disorder

GoalsTechniques
Psychoeducation
  • Increase illness awareness
  • Improve medication compliance
  • Early detection of relapses
  • Establish lifestyle regularity
  • Education based on books, pamphlets, and Web sites regarding the symptoms, course, treatment, and self-management of BD
  • Self-monitoring of symptoms and behaviors
  • Discussion
Family-focused psychoeducation
  • Accept notion of vulnerability for future episodes
  • Accept need for mood-stabilizing medications
  • Educate to distinguish between patient’s personality traits and BD symptoms
  • Reestablish functional relationships after a mood episode
  • Education based on books, pamphlets, and Web sites regarding the symptoms, course, treatment, and self-management of BD
  • Enhance communication skills in the family
  • Education about problem-solving skills
Cognitive therapy
  • Challenge the patient’s dysfunctional thoughts and beliefs regarding self and the world as influenced by BD
  • Self-monitoring of dysfunctional thoughts and behaviors
  • Monitor moods and early signs of relapse
  • Develop a plan of action to deal with early signs of relapse
  • Emphasize the need of combined pharmacotherapy and psychotherapy
  • Promote the importance of regular sleep and healthy lifestyle
Interpersonal and social rhythm therapy
  • Stabilize daily routines and sleep/wake cycles
  • Gain insight into relationship between moods and interpersonal events
  • Relieve stress and interpersonal problems
  • Review history of illness
  • Track and identify connections between sleep patterns, activities, and mood
  • Develop a plan to stabilize social and circadian rhythms by maintaining consistent sleep/wake times and reducing excessive social stimulations
  • Explore and resolve interpersonal problems
BD: bipolar disorder
Source: Reference 11

A theoretical model

Roberto Assagioli, who established the approach to psychology called psychosynthesis, formulated a fundamental psychological principle in controlling one’s behavior: “We are dominated by everything with which our self becomes identified. We can dominate and control everything from which we disidentify ourselves.”15 According to the psychosynthesis model, it is easier to change thoughts we identify as foreign to “the self” (ego-dystonic) than thoughts we identify as being part of “the self” (ego-syntonic).

Patients whose suicidal thoughts are ego-syntonic identify with the thoughts as representing themselves and take ownership of these thoughts. Such patients are at a greater risk of acting on suicidal thoughts.

Patients whose suicidal thoughts are ego-dystonic consider the suicidal thoughts foreign to their core self and do not believe such thoughts represent them. In essence, they “disown” the thoughts and typically want to control and eliminate them. Examples of patients’ ego-syntonic vs ego-dystonic suicidal thoughts are listed in Table 2.

This construct calls for an intervention to help patients who have ego-syntonic suicidal thoughts restructure them as a manifestation of BD, rather then the patient’s core self belief. The intervention emphasizes the patient is not “a suicidal patient” but suffers from an illness that may manifest with suicidal ideation. Many BD patients overly identify with their disease, stating, “I am bipolar” or “I am suicidal.” The “I am” statement originates from the verb “to be,” which implies the disease is part of the patient’s identity. The goal of this intervention is to help the patient learn to disidentify from the disease and decide that suicidal thoughts do not represent their core self, but are a manifestation of the underlying disease.

 

 

The psychosynthesis model of helping patients disidentify and therefore disown suicidal thoughts is compatible with interventions that use mindfulness-based cognitive therapy training to teach patients to experience their thoughts as just passing through their consciousness without taking ownership of them.17

Table 2

Examples of ego-syntonic vs ego-dystonic suicidal thoughts

Ego-syntonicEgo-dystonic
‘I want to be dead. I found a simple and sure way to do it’‘I am having suicidal thoughts again and I don’t like it’
‘I know my family will be better off without me’‘I’m afraid the illness is coming back. I can’t stop these images’
‘Life is too hard, too much pain. I just want to end it all’‘I see my body in a coffin. It scares the hell out of me’
‘I’ve come to the end, life for me is over and done’‘I don’t want to die. Please help me get well again’
‘I know my life is over. I just have to find the right way to do it’‘It is as if a part of me wants to die but the rest of me wants to live’
‘Nobody cares about me. It is as if I am already dead’‘I know my family needs me. I want to be there for them’
‘I have nothing to live for’‘I have so much to live for, why am I having such crazy thoughts?’

The intervention

Assessment of suicidality is a fundamental skill for every mental health clinician.18 The psychotherapeutic intervention I use integrates the cognitive therapy principles of reframing, relabeling, and restructuring patients’ thoughts with disidentification from dysfunctional thoughts, feelings, and desires, based on psychosynthesis principles.

First, I conduct a comprehensive mental status examination that includes an in-depth exploration of the patient’s suicidal thoughts to determine if they are ego-syntonic or ego-dystonic. I begin by asking patients to clarify and elaborate on their statements referring to suicide, asking questions such as “Is there a part of you that objects to these thoughts?” and “Is there a part of you that wants to live?” If a patient indicates that he or she does experience inner conflict regarding such thoughts, these thoughts are classified as ego-dystonic. If a patient does not have any counter thoughts regarding the suicidal thoughts and fully identifies with them, the thoughts are classified as ego-syntonic.

I follow this with a treatment plan that helps patients change their view of their suicidal thoughts. I ask the patient to change these suicidal thoughts to ego-dystonic by focusing on the following statement: “I, (patient’s name), am a human being and like all human beings, I have thoughts; however, I am not my thoughts, I am much more than that.” I ask my patient to read this out loud and to mindfully meditate on this statement several times a day to reinforce the new understanding that these suicidal thoughts are a manifestation of the chemical imbalance of the mood disorder, and do not represent the patient as a person.

This intervention is paired with a future-focused internalized imagery experience I have described in previous articles.19,20 In this part of the treatment, the patient and I discuss a specific expected life milestone that is positive and for which he or she would want to be present (eg, children graduating from high school or college, a wedding, birth of a child/grandchild, etc.). Using guided imagery, the patient experiences this event with all 5 senses during the session. I instruct the patient to internalize the experience and bring it back from the future to the present. This creates a “hook into the future” that is coupled with this desired milestone event in the patient’s life.

The following 3 case studies provide examples of the application of this treatment intervention.

CASE 1: Disidentifying family history

Mrs. G, a 42-year-old mother of 2, suffers from bipolar II disorder with recurrent episodes of depression associated with ego-syntonic suicidal thoughts. She states that at times she feels she is a burden to her husband and children and believes they may be better off without her. She says she believes “ending it all” must be her destiny. After further investigation, I learn Mrs. G has a family history of BD and 3 relatives have committed suicide. This family history may partially explain her belief that suicide must be “in her genes.”

I discuss with Mrs. G the strategy of changing her thoughts. I tell her to write in her journal—which she brings to her sessions—the following statements: “I am a human being. I am an adult woman and mother of 2 children. I know I have thoughts but I am not my thoughts, I am much more than that. I know I have genes but I am not my genes, I am much more than that. I know I have feelings, but I am not my feelings, I am much more than that. I know I have cousins, uncles, aunts, and other relatives but I am not my relatives. I am uniquely myself, different from the others.”

 

 

I ask Mrs. G to read these statements out loud and repeat them several times a day to reinforce this new way of perceiving the suicidal thoughts and to disidentify from the thoughts and her family history as it relates to suicide.

Mrs. G and I talk about the future and expected family milestones. When I ask if her son would want her to be present at his college graduation, she says yes. We then discuss in detail the date, time of day, and location of this event, followed by a guided imagery exercise focused on the graduation. She is guided to experience this event with all 5 senses and describes the event in detail, including the expression on the faces of her husband and children, their voices, and the scent of their aftershave lotion. She hears her son saying, “Mom, I love you. Thank you for being there with me all these years. I could not have done it without you.” I ask Mrs. G to internalize these experiences and carve them into her memory. She is instructed to come back from this future-focused guided imagery experience. When her eyes open, she looks at me and describes her experience in great detail, at times using the past tense, which confirms that the future-focused event was internalized.

In her next session, Mrs. G reports an improvement in her sleep and a change in her suicidal thoughts, which now are only fleeting.

CASE 2: Experiencing graduation

Ms. J, age 17, was diagnosed with bipolar I disorder when she was 15. She has a family history of BD in her mother, 2 maternal aunts, her grandmother, and an older sister. All these women have a history of suicidal thoughts and suicide attempts requiring hospital treatment, but no completed suicides.

Ms. J has been taking an adequate combination of mood stabilizers. She has recovered from 2 previous depressive episodes and is experiencing a third relapse with suicidal thoughts. At times, she experiences these thoughts as ego-syntonic; at other times, they are ego-dystonic.

I first educate her about the nature of BD, explaining that her suicidal thoughts are a manifestation of a chemical imbalance in her brain as a result of the depressive relapse. I teach her to use guided imagery to focus on her favorite place of peace and serenity, the beach, which produces immediate relief of the intense anxiety she felt.

After we complete the disidentification exercise, I ask her to focus on her high school graduation ceremony, which is scheduled to take place in 1 year. In a state of guided imagery, she experiences her graduation from high school with all 5 senses. As she returns to a state of full alertness with her eyes open, she describes the graduation ceremony experience in detail using the past tense, as if it had already occurred, thereby creating her own hook into the future. I instruct her to write about this experience in her journal and bring it with her to the next session.

The following session, Ms. J reports that her suicide ideations have “disappeared.” She says this was accompanied by improvements in her overall mood and sleep.

CASE 3: Internalizing the future

Mr. C, a 38-year-old married father of 4 children, has bipolar II disorder and is in a depressed state. He has been treated with optimal doses of mood stabilizers and atypical antipsychotics but continues to have suicidal thoughts. These thoughts are at times ego-syntonic; he says, “My family would be better off without me.” When Mr. C’s mood improves, however, the suicidal thoughts become more ego-dystonic; he expresses fear that he might act out on the thoughts and states that he does not want to die, he really wants to live and get better. He has no history of suicide attempts.

During our session, I ask Mr. C to focus on a new perspective to understand his thoughts by repeating the following statements: “I, JC, am a human being. I know I have a bipolar mood disorder; however, I must remember I am not bipolar. I have suicidal thoughts; however, I am not my thoughts, I am much more than that. I know I want to live, to heal, and to get better. I want to be alive and well so I can see and participate in my children’s graduation from high school and be there when they get married and when my grandchildren are born.”

 

 

I teach Mr. C to use guided imagery, during which he experiences such future positive images and milestones in his life in all 5 senses and internalizes them by using the “back from the future” technique.17 By the end of the session, he reports feeling better, more hopeful, and confident in his abilities to control his suicidal thoughts. I instruct him to write in his diary about his experiences with the future-focused positive milestones and to bring this assignment to his next appointment.

At his next appointment, Mr. C reports that his suicidal thoughts have become more fleeting, lasting for 10 to 30 seconds, and then spontaneously change to focus on issues of the “here and now.” When I ask him to read what he’s written, what stands out is the use of past tense verbs to describe future-focused experiences. For me, this confirms that Mr. C has internalized the future, creating the desirable “future hook” that acts as an antidote to the suicidal thoughts.

Related Resources

  • Rouget BW, Aubry JM. Efficacy of psychoeducational approaches on bipolar disorders: a review of the literature. J Affect Disord. 2007;98:11-27.
  • Weinberg I, Ronningstam E, Goldblatt MJ, et al. Strategies in treatment of suicidality: identification of common and treatment-specific interventions in empirically supported treatment manuals. J Clin Psychiatry. 2010;71:699-706.

Drug Brand Name

  • Lithium • Eskalith, Lithobid

Disclosure

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

References

1. McIntyre RS, Konarski JZ. Bipolar disorder: a national health concern. CNS Spectr. 2004;9(11 suppl 12):6-15.

2. Tsai SY, Lee CH, Kuo CJ, et al. A retrospective analysis of risk and protective factors for natural death in bipolar disorder. J Clin Psychiatry. 2005;66(12):1586-1591.

3. Osby U, Brandt L, Correia N, et al. Excess mortality in bipolar and unipolar disorder in Sweden. Arch Gen Psychiatry. 2001;58(9):844-850.

4. Baldessarini RJ, Pompili M, Tondo L. Suicide in bipolar disorder: risks and management. CNS Spectr. 2006;11(6):465-471.

5. Gelenberg AJ. Can lithium help to prevent suicide? Acta Psychiatr Scand. 2001;104(3):161-162.

6. Schou M. Suicidal behavior and prophylactic lithium treatment of major mood disorders: a review of reviews. Suicide Life Threat Behav. 2001;30(3):289-293.

7. Burgess S, Geddes J, Hawton K, et al. Lithium for maintenance treatment of mood disorders. Cochrane Database Syst Rev. 2001;(3):CD003013.-

8. Baldessarini RJ, Tondo L, Davis P, et al. Decreased risk of suicides and attempts during long-term lithium treatment: a meta-analytic review. Bipolar Disord. 2006;8(5 Pt 2):625-639.

9. Tondo L, Baldessarini RJ. Long-term lithium treatment in the prevention of suicidal behavior in bipolar disorder patients. Epidemiol Psichiatr Soc. 2009;18(3):179-183.

10. Miklowitz DJ. Adjunctive psychotherapy for bipolar disorder: state of the evidence. Am J Psychiatry. 2008;165(11):1408-1419.

11. Hirschfeld RMA, Harris TH, Davis HK. Making efficacious choices: the integration of pharmacotherapy and nonpharmacologic approaches to the treatment of patients with bipolar disorder. Current Psychiatry. 2009;8(10 suppl):S6-S11.

12. Rucci P, Frank E, Kostelnik B, et al. Suicide attempts in patients with bipolar I disorder during acute and maintenance phases of intensive treatment with pharmacotherapy and adjunctive psychotherapy. Am J Psychiatry. 2002;159(7):1160-1164.

13. Fountoulakis KN, Gonda X, Siamouli M, et al. Psychotherapeutic intervention and suicide risk reduction in bipolar disorder: a review of the evidence. J Affect Disord. 2009;113(1-2):21-29.

14. Pompili M, Rihmer Z, Innamorati M, et al. Assessment and treatment of suicide risk in bipolar disorders. Expert Rev Neurother. 2009;9(1):109-136.

15. Assagioli R. Psychosynthesis: a collection of basic writings. New York NY: The Viking Press Inc.; 1965.

16. Assagioli R. The act of will. New York NY: The Viking Press Inc.; 1973.

17. Williams JM, Alatiq Y, Crane C, et al. Mindfulness-based cognitive therapy (MBCT) in bipolar disorder: preliminary evaluation of immediate effects on between-episode functioning. J Affect Disord. 2008;107(1-3):275-279.

18. Shea SC. The delicate art of eliciting suicidal ideation. Psychiatric Annals. 2004;34:385-400.

19. Torem MS. “Back from the future”: a powerful age-progression technique. Am J Clin Hypn. 1992;35(2):81-88.

20. Torem MS. Treating depression: a remedy from the future. In: Yapko MD ed. Hypnosis and treating depression: applications in clinical practice. New York, NY: Routledge; 2006:97–119.

References

1. McIntyre RS, Konarski JZ. Bipolar disorder: a national health concern. CNS Spectr. 2004;9(11 suppl 12):6-15.

2. Tsai SY, Lee CH, Kuo CJ, et al. A retrospective analysis of risk and protective factors for natural death in bipolar disorder. J Clin Psychiatry. 2005;66(12):1586-1591.

3. Osby U, Brandt L, Correia N, et al. Excess mortality in bipolar and unipolar disorder in Sweden. Arch Gen Psychiatry. 2001;58(9):844-850.

4. Baldessarini RJ, Pompili M, Tondo L. Suicide in bipolar disorder: risks and management. CNS Spectr. 2006;11(6):465-471.

5. Gelenberg AJ. Can lithium help to prevent suicide? Acta Psychiatr Scand. 2001;104(3):161-162.

6. Schou M. Suicidal behavior and prophylactic lithium treatment of major mood disorders: a review of reviews. Suicide Life Threat Behav. 2001;30(3):289-293.

7. Burgess S, Geddes J, Hawton K, et al. Lithium for maintenance treatment of mood disorders. Cochrane Database Syst Rev. 2001;(3):CD003013.-

8. Baldessarini RJ, Tondo L, Davis P, et al. Decreased risk of suicides and attempts during long-term lithium treatment: a meta-analytic review. Bipolar Disord. 2006;8(5 Pt 2):625-639.

9. Tondo L, Baldessarini RJ. Long-term lithium treatment in the prevention of suicidal behavior in bipolar disorder patients. Epidemiol Psichiatr Soc. 2009;18(3):179-183.

10. Miklowitz DJ. Adjunctive psychotherapy for bipolar disorder: state of the evidence. Am J Psychiatry. 2008;165(11):1408-1419.

11. Hirschfeld RMA, Harris TH, Davis HK. Making efficacious choices: the integration of pharmacotherapy and nonpharmacologic approaches to the treatment of patients with bipolar disorder. Current Psychiatry. 2009;8(10 suppl):S6-S11.

12. Rucci P, Frank E, Kostelnik B, et al. Suicide attempts in patients with bipolar I disorder during acute and maintenance phases of intensive treatment with pharmacotherapy and adjunctive psychotherapy. Am J Psychiatry. 2002;159(7):1160-1164.

13. Fountoulakis KN, Gonda X, Siamouli M, et al. Psychotherapeutic intervention and suicide risk reduction in bipolar disorder: a review of the evidence. J Affect Disord. 2009;113(1-2):21-29.

14. Pompili M, Rihmer Z, Innamorati M, et al. Assessment and treatment of suicide risk in bipolar disorders. Expert Rev Neurother. 2009;9(1):109-136.

15. Assagioli R. Psychosynthesis: a collection of basic writings. New York NY: The Viking Press Inc.; 1965.

16. Assagioli R. The act of will. New York NY: The Viking Press Inc.; 1973.

17. Williams JM, Alatiq Y, Crane C, et al. Mindfulness-based cognitive therapy (MBCT) in bipolar disorder: preliminary evaluation of immediate effects on between-episode functioning. J Affect Disord. 2008;107(1-3):275-279.

18. Shea SC. The delicate art of eliciting suicidal ideation. Psychiatric Annals. 2004;34:385-400.

19. Torem MS. “Back from the future”: a powerful age-progression technique. Am J Clin Hypn. 1992;35(2):81-88.

20. Torem MS. Treating depression: a remedy from the future. In: Yapko MD ed. Hypnosis and treating depression: applications in clinical practice. New York, NY: Routledge; 2006:97–119.

Issue
Current Psychiatry - 10(10)
Issue
Current Psychiatry - 10(10)
Page Number
39-45
Page Number
39-45
Publications
Publications
Topics
Article Type
Display Headline
Beyond lithium: Using psychotherapy to reduce suicide risk in bipolar disorder
Display Headline
Beyond lithium: Using psychotherapy to reduce suicide risk in bipolar disorder
Legacy Keywords
lithium; psychotherapy; suicide risk; reduce; bipolar disorder; BD; disown thoughts; internalize future; suicide ideation; suicide attempts; suicide; psychotheraputic treatments; nonpharmacologic interventions; Moshe Torem
Legacy Keywords
lithium; psychotherapy; suicide risk; reduce; bipolar disorder; BD; disown thoughts; internalize future; suicide ideation; suicide attempts; suicide; psychotheraputic treatments; nonpharmacologic interventions; Moshe Torem
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Is there a link between aripiprazole and treatment-emergent psychosis?

Article Type
Changed
Tue, 12/11/2018 - 14:48
Display Headline
Is there a link between aripiprazole and treatment-emergent psychosis?

Discuss this article at www.facebook.com/CurrentPsychiatry

 

Practice Points

Aripiprazole may interact preferentially with distinct conformations of the D2 receptor, leading to a spectrum of pharmacologic effects, including acting as a full agonist, partial agonist, or antagonist.

Clinical predictors of aripiprazole-associated worsening of psychosis include low baseline level of psychopathology and previous treatment with high-dose antipsychotics.

• Rapid transition from a medication with significant anticholinergic properties to 1 without these properties may result in symptoms of activation, including restlessness, insomnia, and anxiety, which can be mistaken for worsening psychosis.

Akathisia, a common adverse effect of aripiprazole, may masquerade as treatment-emergent worsening of psychotic symptoms.

Mr. N, age 29, presents to the emergency department at the urging of his family because of poor self-care, bizarre behavior, and disturbed sleep. He first experienced psychiatric symptoms 10 years ago after his mother died. He became dysphoric and paranoid, displaying bizarre responses and behaviors with poor self-care and a gradual functional decline. He has been taking sertraline, 100 mg/d, for 10 years.

Upon arrival at the hospital’s inpatient unit, Mr. N is unkempt, oddly related, and paranoid. His affect is constricted. Mr. N displays thought blocking and possibly is responding to internal stimuli. Sertraline is continued and haloperidol, 1 mg/d, is initiated. For the next 2 weeks, Mr. N continues to be oddly related, irritable, and paranoid, and experiences disturbed sleep and thought blocking. After an episode of impulsive aggression, the treatment team initiates aripiprazole, which is titrated to 30 mg/d for 1 week. Mr. N’s clinical status worsens; he is menacing toward other patients and his thinking is more disorganized, with loose associations and ideas of reference. He requires 4 injections of IM haloperidol, 5 mg, and several visits to the seclusion room over the next week. Haloperidol is increased to 30 mg/d over the next 10 days, then aripiprazole is discontinued because of a putative drug interaction with haloperidol. Following the medication changes Mr. N demonstrates better behavioral control, but still is grossly psychotic. While awaiting transfer to a state hospital, Mr. N receives a trial of olanzapine, 20 to 40 mg/d, for 2 weeks without significant benefit.

Several clinical trials demonstrate a significant reduction in intensity of psychotic symptoms with aripiprazole, which has a unique mechanism of action.1 However, since its FDA approval in 2002, several case reports have described treatment-emergent psychotic symptoms associated with aripiprazole initiation. Over the past 40 years, reports of worsening psychosis associated with antipsychotics have been limited to patients with schizophrenia who were taking high dosages or who had high plasma concentrations, when anticholinergic delirium may have explained increased psychotic symptoms.2-4

How can a drug effectively treat psychotic symptoms and occasionally worsen them? In this article, we discuss the relevant pharmacology and clinical literature on aripiprazole and try to make sense of this apparent paradox.

Unique pharmacologic profile

Antipsychotics have been reported to be either neutral antagonists or inverse agonists at the D2 receptor, based on in vitro data.5 Aripiprazole and its main metabolite, dehydroaripiprazole, originally were described as partial agonists at D2 dopamine receptors.6,7 However, it appears aripiprazole’s pharmacologic action is better explained by the concept of functional selectivity. Aripiprazole may interact preferentially with distinct conformations of the D2 receptor, leading to a spectrum of pharmacologic effects, including acting as a full agonist, partial agonist, or antagonistic.5

Researchers have hypothesized that the pathophysiology of schizophrenia may, in part, be caused by dysfunction of mesocorticolimbic dopaminergic neurons characterized by an enhanced sensitivity of postsynaptic D2 receptors and increased sensitivity to dopaminergic drugs.8,9 In addition, chronic treatment with a D2 receptor antagonist is associated with increases in postsynaptic dopamine receptor density (ie, an increase in receptor reserve).10,11 Upregulation of D2 receptors may explain several features seen in patients chronically treated with antipsychotics, including tardive dyskinesia12 and rapid psychotic relapse after discontinuing an antipsychotic (supersensitivity psychosis).13 Because chronic antipsychotic treatment leads to high postsynaptic receptor reserve, aripiprazole initiation may produce overactivation of D2 receptors, which might worsen a patient’s condition.14 In vitro data15-18 and clinical observations indicate that aripiprazole has intrinsic efficacy at D2 receptors, as do clinical observations, such as:

 

 

  • its propensity to reduce serum prolactin19
  • a decreased likelihood of producing extrapyramidal side effects despite >80% occupancy of D2 receptors6
  • case reports documenting aripiprazole-associated mania,20 improvement of risperidone-associated cognitive impairment,21 and pathologic gambling.22

Emergence or worsening of psychotic symptoms or a marginal antipsychotic effect may occur if aripiprazole is indeed a postsynaptic D2 receptor agonist. An individual patient’s outcome likely would depend on his or her sensitivity to psychosis and concurrent or previous exposure to a D2 receptor antagonist. For example, stimulation of postsynaptic D2 receptors may be further augmented if the dosage of the previous antipsychotic was reduced or withdrawn before initiating aripiprazole because additional receptors would be available for interaction with aripiprazole.

 

 

 

Case reports

A literature review revealed 23 reports of treatment-emergent psychosis associated with aripiprazole initiation (Table). The mean age of the patients was 47 (range: 17 to 69) and 57% were men. Most patients (87%) were diagnosed with a schizophrenia-spectrum illness before aripiprazole initiation. Most (57%) had mild, stable, or no psychotic symptoms before aripiprazole initiation. Most were receiving relatively high doses of antipsychotics (average chlorpromazine equivalents [CPZE]: 648 mg/d) before aripiprazole initiation. This medication was either decreased or discontinued in 70% of patients.

Emergence or worsening of psychotic symptoms included agitation, aggressive behavior, and increased psychomotor activity. However, akathisia evaluation was described in only 2 reports: 1 author identified akathisia symptoms, but attributed them to a concomitant antipsychotic (fluphenazine)23 and the other report specifically excluded the possibility of akathisia.24 Two systematic studies have attempted to establish risk factors for aripiprazole-associated worsening psychosis (Box).14,25

In our literature review, the mean final dose of aripiprazole was 21.5 mg/d (range: 2 to 60 mg/d). In the cases describing subsequent treatment, all but 1 patient were switched to another antipsychotic, including 2 whose psychotic symptoms stabilized with continuation of aripiprazole and addition of a second antipsychotic. Interestingly, in the case reported by Adan-Manes et al,26 initial treatment with aripiprazole monotherapy was efficacious, but a subsequent trial of adjunctive aripiprazole resulted in worsening psychosis.

Table

Case reports: Treatment-emergent psychosis associated with aripiprazole

 

StudyAge, sexDiagnosisBefore aripiprazole initiationPre-aripiprazole treatmentAripiprazole doseConcomitant psychotropic treatmentSubsequent treatment
Chiu et al, 2011a39, MSchizophreniaPsychiatrically stable, tardive dystoniaClozapine, 300 mg/d10 mg/dValproic acid, 1,000 mg/d, clonazepam, 2 mg/d, mephenoxalone, 800 mg/dClozapine
Ekinci et al, 2010b17, MADHDInattention and impulsive aggressionTapered and discontinued risperidone, 2.5 mg/d5 mg/dMethylphenidate, 54 mg/dRisperidone, 2 mg/d, methylphenidate, 36 mg/d
Selvaraj et al, 2010c49, FChronic depressionDepressive symptoms, suicidal ideationNone stated2 mg/dDuloxetine, 80 mg/d, clonazepam, 2 mg/dDuloxetine, 120 mg/d
Adan-Manes et al, 2009d23, MSchizophreniaNo psychotic symptomsAbrupt decrease of amisulpride dose from 800 mg/d to 400 mg/d20 mg/dBiperiden, 4 mg/dAmisulpride, 800 mg/d
Cho et al, 2009e45, FSchizophreniaPersistent psychotic symptoms, new onset diabetes with acute ketoacidosisHaloperidol, 20 mg/d, abrupt clozapine discontinuation15 mg/dValproic acid, nortriptylineMolindone, 150 mg/d
Ahuja et al, 2007f35, FSchizoaffective disorderStable before medication changeTapered amisulpride, 400 mg/d, over 6 weeks15 mg/dNoneAmisulpride, 600 mg/d
Lea et al, 2007g57, MSchizophreniaPersistent psychotic symptoms, treatment resistance, recent recovery from NMSDiscontinued ziprasidone, 200 mg/d30 mg/dLorazepam, 2 mg/d, amantadine, 100 mg, sertraline, 50 mg/dClozapine
Lea et al, 2007g49, MSchizoaffective disorderDelusions, verbal aggression, substance abuse, HCVDecreased quetiapine dose from 800 mg/d to 400 mg/d15 mg/dDivalproex, 1,000 mg/d, fluvoxamine, 200 mg/d, clonazepam, 2 mg/dLithium, quetiapine, 500 mg/d, haloperidol, 2 mg/d
Lea et al, 2007g60, MSchizophreniaDelusions, labile mood, aggressionRisperidone, 3 mg/d, interruption of fluphenazine, 75 mg/d20 mg/dDivalproex, 4,500 mg/d, benztropine, 3 mg/dNot discussed
Raja, 2007h30, MSchizoaffective disorderNegative symptoms, otherwise stable, recent citalopram discontinuationDiscontinued amisulpride, 800 mg/d over 2 weeks30 mg/dLithiumAmisulpride, 500 mg/d
Raja, 2007h69, FBipolar disorderHistory of multiple relapses; presented with tremor, akathisia, weight gainDiscontinued risperidone, 2 mg/d, over 2 weeks15 mg/dLithiumRisperidone, 4 mg
Raja, 2007h59, FSchizophreniaNegative symptoms, otherwise stableReduced risperidone dosage from 5 mg/d to 4 mg/d7.5 mg/dNoneRisperidone, 5 mg/d
Thone, 2007i31, MSchizophreniaConfusion, agitation, delusions worsened with aripiprazole dose increaseNone60 mg/dNoneAripiprazole dose reduction to 15 mg/d, olanzapine, 10 mg/d
Glick et al, 2006j55, FSchizophreniaStable before medication changeTapered and discontinued thioridazine, 600 mg/d, over 3 months30 mg/dNoneChlorpromazine, 200 mg/d, aripiprazole, 30 mg/d
Glick et al, 2006j52, MSchizophreniaNegative symptomsDecreased olanzapine dose from 30 mg/d to 20 mg/d30 mg/dNoneOlanzapine, 30 mg/d
Barnas et al, 2005k57, FSchizoaffective disorderStable before medication changeDiscontinued perphenazine, 8 mg/d30 mg/dNoneQuetiapine, 350 mg/d
DeQuardo, 2004l54, MSchizophreniaHistory of aggression, residual paranoia, severe EPSHaloperidol, 200 mg/d15 mg/dBenztropineHaloperidol
DeQuardo, 2004l51, MSchizophreniaHistory of aggression, persistent psychotic symptoms, treatment resistanceOlanzapine, 60 mg/d10 mg/dNoneOlanzapine
Ramaswamy et al, 2004m43, FSchizoaffective disorderPsychiatrically stable, multiple medication changes, including substituting carbamazepine for valproic acidDiscontinued ziprasidone, 160 mg/d, discontinued quetiapine, 400 mg/d, over 2 weeks30 mg/dPropranolol, 30 mg/d, l-thyroxine, .05 mg/d, carbamazepine, 600 mg/dNot available
Ramaswawamy et al, 2004m57, FSchizoaffective disorderHistory of multiple hospitalizations, but stable before medication changeDecreased olanzapine dose from 20 mg/d to 15 mg/d30 mg/dValproic acid, 2,000 mg/dZiprasidone
Ramaswawamy et al, 2004m67, FSchizophreniaRemote hospitalizations, recent worsened psychosisDecreased ziprasidone dose from 200 mg/d to 160 mg/d 2 months previously30 mg/dCarbamazepine, 200 mg/dNot discussed
Ramaswamy et al, 2004m46, MSchizophreniaPersistent delusions while receiving risperidone, TDRisperidone, 3 mg/d15 mg/dValproic acid, 1,500 mg/dRisperidone, 3 mg/d
Reeves et al, 2004n50, MSchizoaffective disorderRelatively stable with nonthreatening delusions, hallucinationsQuetiapine, 800 mg/d30 mg/dDivalproex, 2,000 mg/dOlanzapine, 20 mg/d
ADHD: attention-deficit/hyperactivity disorder; EPS: extrapyramidal symptoms; HCV: hepatitis C virus; NMS: neuroleptic malignant syndrome; TD: tardive dyskinesia Source:
References
a. Chiu YH, Chen CH, Lu ML. Worsening psychosis after adding aripiprazole to clozapine. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35(1):291-292.
b. Ekinci O, Sabuncuoglu O. Psychosis associated with switching from risperidone to aripiprazole in an adolescent on methylphenidate treatment. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35(2):648-649.
c. Selvaraj V, Ramaswamy S, Sharma A, et al. New-onset psychosis and emergence of suicidal ideation with aripiprazole. Am J Psychiatry. 2010;167(12):1535-1536.
d. Adan-Manes J, Garcia-Parajua P. Aripiprazole in combination with other antipsychotic drugs may worsen psychosis. J Clin Pharm Ther. 2009;34(2):245-246.
e. Cho DY, Lindenmayer JP. Aripiprazole-induced agitation after clozapine discontinuation: a case report. J Clin Psychiatry. 2009;70(1):141-143.
f. Ahuja N, Lloyd AJ. Aripiprazole and worsening of psychosis: a case report. J Clin Psychiatry. 2007;68(5):805-806.
g. Lea JW, Stoner SC, Lafollette J. Agitation associated with aripiprazole initiation. Pharmacotherapy. 2007;27(9):1339-1342.
h. Raja M. Improvement or worsening of psychotic symptoms after treatment with low doses of aripiprazole. Int J Neuropsychopharmacol. 2007;10(1):107-110.
i. Thone J. Worsened agitation and confusion in schizophrenia subsequent to high-dose aripiprazole. J Neuropsychiatry Clin Neurosci. 2007;19(4):481-482.
j. Glick ID, Duggal V, Hodulik C. Aripiprazole as a dopamine partial agonist: positive and negative effects. J Clin Psychopharmacol. 2006;26(1):101-103.
k. Barnas ME, Hussain N, Petrides G. Treatment-emergent psychosis with aripiprazole. J Clin Psychiatry. 2005;66(10):1339.
l. DeQuardo JR. Worsened agitation with aripiprazole: adverse effect of dopamine partial agonism? J Clin Psychiatry. 2004;65(1):132-133.
m. Ramaswamy S, Vijay D, William M, et al. Aripiprazole possibly worsens psychosis. Int Clin Psychopharmacol. 2004;19(1):45-48.
n. Reeves RR, Mack JE. Worsening schizoaffective disorder with aripiprazole. Am J Psychiatry. 2004;161(7):1308.
 

 

 

 

Clinical predictors of aripiprazole-associated psychotic symptoms

Takeuchi et al14 aimed to establish predictors of worsening psychosis in a naturalistic setting where patients slowly transitioned to aripiprazole from previous antipsychotic treatment. Patients were required to be on a stable dose of an antipsychotic before participating in the study. Aripiprazole was started at 12 mg/d for 2 weeks with flexible dosing from weeks 2 to 52. Previous antipsychotic therapy was reduced biweekly by 25%. The incidence of worsening psychopathology after aripiprazole initiation was higher in the group of patients who had previously received high-dose antipsychotic therapy (average chlorpromazine equivalents [CPZE]: 727 mg/d) compared with the group on low dosages (average CPZE: 382 mg/d). It is possible that previous high-dose antipsychotic therapy was indicative of more significant baseline psychopathology; however, the worsened group and stabilized group had similar baseline Clinical Global Impressions-Severity scores.

Pae et al25 aimed to find predictors of worsening psychosis with aripiprazole in patients whose previous antipsychotic therapy was immediately discontinued. They found lower baseline disease severity was associated with significant worsening during the first month of aripiprazole treatment.

Other potential explanations

 

Aripiprazole’s manufacturer reported the incidence of psychosis-related adverse events in an analysis of 9 randomized schizophrenia trials.27 The rates of psychosis-related adverse events ranged from 0.6% to 18%, but there was no apparent relationship to study design or method of transitioning to aripiprazole. Rates of psychosis-related adverse events were similar between aripiprazole and the control group (placebo in 3 studies, another antipsychotic in 2 studies).

Emergence or worsening of psychotic symptoms temporally associated with aripiprazole initiation does not necessarily imply causation. As in Mr. N’s case, it is not always possible to determine whether worsening psychosis is the natural disease course or a treatment effect. In addition, it is not possible to differentiate lack of efficacy from a true propensity for aripiprazole to worsen psychosis.

 

It also is conceivable discontinuation or dosage reduction of a previous antipsychotic would worsen psychotic symptoms or cause side effects. When significant changes in psychopathology or side effects develop during the transition from 1 antipsychotic to another, it is difficult to determine etiology. Specifically, rapid transition from a medication with significant anticholinergic and antihistaminic properties—such as quetiapine or olanzapine—to 1 without these properties—such as aripiprazole—may result in symptoms of activation, including restlessness, insomnia, and anxiety. Consequently, these symptoms could be mistaken for worsening psychosis.28 Only 1 patient in this series was reported to abruptly discontinue an antipsychotic with significant anticholinergic properties (clozapine) before initiating aripiprazole.24 Studies by Takeuchi et al14 and Pae et al25 did not report the relative baseline use of antipsychotic medication with anticholinergic properties.

In a pooled analysis of treatment-emergent adverse events in 5 randomized clinical trials of patients receiving aripiprazole for acute relapse of schizophrenia, the incidence of akathisia was 10%, although it is not clear if this is a dose-related adverse effect.29 Because akathisia may be confused for worsening psychosis,30 it is possible akathisia was mistakenly identified as worsening psychotic symptoms in Mr. N’s case, as well as several reports from our literature review.

Covert akathisia is unlikely to explain worsening psychopathology observed in all patients in our literature review because confusion of akathisia and worsening psychosis is not a widespread phenomenon. In a post hoc analysis of pooled safety data from aripiprazole trials, Kane et al31 did not find a correlation between presence of akathisia and aripiprazole efficacy as measured by the Positive and Negative Syndrome Scale (PANSS) total, PANSS positive, PANSS negative, Clinical Global Impressions-Severity, Clinical Global Impressions-Improvement, and percentage of responders. Pae et al25 also noted there was no correlation between scores on the Barnes Akathisia Rating Scale and worsening psychopathology in patients switched to aripiprazole.

 

An antagonist always is an antagonist and clinicians have appreciated this concept since the days of chlorpromazine. The activity of aripiprazole, however, is on a pharmacologic continuum between a neutral antagonist and full agonist and currently there is no way to precisely determine the level of D2 receptor agonist action in a patient.

Although it is interesting to speculate that aripiprazole’s D2 receptor agonist action may contribute to worsening psychosis,32-34 there are other plausible explanations to consider. Rapid transition from a drug with significant anticholinergic properties and aripiprazole-associated akathisia may contribute to worsening psychopathology in patients starting aripiprazole. Because covert side effects may be incorrectly identified as psychotic agitation, we cannot exclude this as a possible etiologic factor in Mr. N’s case as well as the cases in our literature review.

 

 

Related Resource

 

  • Abilify [package insert]. Princeton, NJ: Bristol-Myers Squibb; 2011.

Drug Brand Names

 

  • Amantadine • Symmetrel
  • Aripiprazole • Abilify
  • Benztropine • Cogentin
  • Biperiden • Akineton
  • Carbamazepine • Tegretol
  • Chlorpromazine • Thorazine
  • Clonazepam • Klonopin
  • Clozapine • Clozaril
  • Divalproex • Depakote
  • Duloxetine • Cymbalta
  • Fluphenazine • Permitil, Prolixin
  • Fluvoxamine • Luvox
  • Haloperidol • Haldol
  • Lithium • Eskalith, Lithobid
  • Lorazepam • Ativan
  • Nortriptyline • Aventyl, Pamelor
  • Methylphenidate • Concerta
  • Molindone • Moban
  • Olanzapine • Zyprexa
  • Perphenazine • Trilafon
  • Propranolol • Inderal
  • Quetiapine • Seroquel
  • Risperidone • Risperdal
  • Sertraline • Zoloft
  • Thioridazine • Mellaril
  • Thyroxine • Synthroid
  • Valproic acid • Depakene
  • Ziprasidone • Geodon

Disclosure

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

References

 

1. Citrome L. A review of aripiprazole in the treatment of patients with schizophrenia or bipolar I disorder. Neuropsychiatr Dis Treat. 2006;2(4):427-443.

2. Chong SA, Tan CH, Lee HS. Worsening of psychosis with clozapine and selective serotonin reuptake inhibitor combination: two case reports. J Clin Psychopharmacol. 1997;17(1):68-69.

3. Bowers MB Jr, Swigar ME. Psychotic patients who become worse on neuroleptics. J Clin Psychopharmacol. 1988;8(6):417-421.

4. Tornatore FL, Lee D, Sramek JJ. Psychotic exacerbation with haloperidol. Drug Intell Clin Pharm. 1981;15(3):209-213.

5. Beaulieu JM, Gainetdinov RR. The physiology signaling, and pharmacology of dopamine receptors. Pharmacol Rev. 2011;63(1):182-217.

6. Grunder G, Carlsson A, Wong DF. Mechanism of new antipsychotic medications: occupancy is not just antagonism. Arch Gen Psychiatry. 2003;60(10):974-977.

7. Wood MD, Scott C, Clarke K, et al. Aripiprazole and its human metabolite are partial agonists at the human dopamine D2 receptor, but the rodent metabolite displays antagonist properties. Eur J Pharmacol. 2006;546(1-3):88-94.

8. Seeman P, Weinshenker D, Quirion R, et al. Dopamine supersensitivity correlates with D2High states, implying many paths to psychosis. Proc Natl Acad Sci U S A. 2005;102(9):3513-3518.

9. Seeman P, Ko F, Jack E, et al. Consistent with dopamine supersensitivity, RGS9 expression is diminished in the amphetamine-treated animal model of schizophrenia and in postmortem schizophrenia brain. Synapse. 2007;61(5):303-309.

10. Burt DR, Creese I, Snyder SH. Antischizophrenic drugs: chronic treatment elevates dopamine receptor binding in brain. Science. 1977;196(4287):326-328.

11. Silvestri S, Seeman MV, Negrete JC, et al. Increased dopamine D2 receptor binding after long-term treatment with antipsychotics in humans: a clinical PET study. Psychopharmacology (Berl). 2000;152(2):174-180.

12. Sayers AC, Bürki HR, Ruch W, et al. Neuroleptic-induced hypersensitivity of striatal dopamine receptors in the rat as a model of tardive dyskinesias. Effects of clozapine, haloperidol, loxapine and chlorpromazine. Psychopharmacologia. 1975;41(2):97-104.

13. Moncrieff J. Does antipsychotic withdrawal provoke psychosis? Review of the literature on rapid onset psychosis (supersensitivity psychosis) and withdrawal-related relapse. Acta Psychiatr Scand. 2006;114(1):3-13.

14. Takeuchi H, Uchida H, Suzuki T, et al. Predictors of clinical worsening after a switch to aripiprazole in patients with schizophrenia: a 1-year naturalistic follow-up study. J Clin Psychopharmacol. 2009;29(4):394-395.

15. Shapiro DA, Renock S, Arrington E, et al. Aripiprazole, a novel atypical antipsychotic drug with a unique and robust pharmacology. Neuropsychopharmacology. 2003;28(8):1400-1411.

16. Urban JD, Vargas GA, von Zastrow M, et al. Aripiprazole has functionally selective actions at dopamine D2 receptor-mediated signaling pathways. Neuropsychopharmacology. 2007;32(1):67-77.

17. Klewe IV, Nielsen SM, Tarpo L, et al. Recruitment of beta-arrestin2 to the dopamine D2 receptor: Insights into anti-psychotic and anti-parkinsonian drug receptor signaling. Neuropharmacology. 2008;54(8):1215-1222.

18. Masri B, Salahpour A, Didriksen M, et al. Antagonism of dopamine D2 receptor/beta-arrestin 2 interaction is a common property of clinically effective antipsychotics. Proc Natl Acad Sci U S A. 2008;105(36):13656-13661.

19. Shim JC, Shin JG, Kelly DL, et al. Adjunctive treatment with a dopamine partial agonist, aripiprazole, for antipsychotic-induced hyperprolactinemia: a placebo-controlled trial. Am J Psychiatry. 2007;164(9):1404-1410.

20. Padala PR, Wengel SP, Petty F. Manic episode during treatment with aripiprazole. Am J Psychiatry. 2007;164(1):172-173.

21. Hu CH, Pai N, Huang XF, et al. Potential control of risperidone-related cognitive deficits by adjunctive aripiprazole treatment. J Clin Psychopharmacol. 2011;31(1):135-136;author reply 136–137.

22. Cohen J, Magalon D, Boyer L, et al. Aripiprazole-induced pathological gambling: a report of 3 cases. Curr Drug Saf. 2011;6(1):51-53.

23. Lea JW, Stoner SC, Lafollette J. Agitation associated with aripiprazole initiation. Pharmacotherapy. 2007;27(9):1339-1342.

24. Cho DY, Lindenmayer JP. Aripiprazole-induced agitation after clozapine discontinuation: a case report. J Clin Psychiatry. 2009;70(1):141-143.

25. Pae CU, Chiesa A, Mandelli L, et al. Predictors of early worsening after switch to aripiprazole: a randomized, controlled, open-label study. Clin Drug Investig. 2010;30(3):187-193.

26. Adan-Manes J, Garcia-Parajua P. Aripiprazole in combination with other antipsychotic drugs may worsen psychosis. J Clin Pharm Ther. 2009;34(2):245-246.

27. Cognata-Smith C, Baker RA, Pikalov A, et al. Analysis of nine aripiprazole trials to evaluate strategies for switching patients with schizophrenia to aripiprazole. Paper presented at: 162nd Annual Meeting American Psychiatric Association; May 16-21, 2009; San Francisco, CA.

28. Lieberman J. Cholinergic rebound in neuroleptic withdrawal syndromes. Psychosomatics. 1981;22(3):253-254.

29. Marder SR, McQuade RD, Stock E, et al. Aripiprazole in the treatment of schizophrenia: Safety and tolerability in short-term, placebo-controlled trials. Schizophr Res. 2003;61(2-3):123-136.

30. Kane JM, Fleischhacker WW, Hansen L, et al. Akathisia: an updated review focusing on second-generation antipsychotics. J Clin Psychiatry. 2009;70(5):627-643.

31. Kane JM, Barnes TR, Correll CU, et al. Evaluation of akathisia in patients with schizophrenia, schizoaffective disorder, or bipolar I disorder: A post hoc analysis of pooled data from short- and long-term aripiprazole trials. J Psychopharmacol. 2010;24(7):1019-1029.

32. Fleischhacker WW, McQuade RD, Marcus RN, et al. A double-blind, randomized comparative study of aripiprazole and olanzapine in patients with schizophrenia. Biol Psychiatry. 2009;65(6):510-517.

33. Kane JM, Osuntokun O, Kryzhanovskaya LA, et al. A 28-week, randomized, double-blind study of olanzapine versus aripiprazole in the treatment of schizophrenia. J Clin Psychiatry. 2009;70(4):572-581.

34. Kane JM, Correll CU, Goff DC, et al. A multicenter, randomized, double-blind, placebo-controlled, 16-week study of adjunctive aripiprazole for schizophrenia or schizoaffective disorder inadequately treated with quetiapine or risperidone monotherapy. J Clin Psychiatry. 2009;70(10):1348-1357.

Article PDF
Author and Disclosure Information

 

James J. Gugger, PharmD, BCPP
Dr. Gugger is Assistant Clinical Professor, St. John’s University, College of Pharmacy and Allied Health Professions, Queens, NY
Courtney L. Tam, PharmD
Dr. Tam is Pharmacy Practice Resident, St. John’s University, College of Pharmacy and Allied Health Professions, Queens, NY
Charles R. Ashby, Jr, PhD
Dr. Ashby is Professor, St. John’s University, College of Pharmacy and Allied Health Professions, Queens, NY

Vicki L. Ellingrod, PharmD, BCPP, FCCP
Series Editor

Issue
Current Psychiatry - 10(10)
Publications
Topics
Page Number
53-60
Legacy Keywords
aripiprazole; treatment-emergent psychosis; James Gugger; Courtney Tam; Charles Ashby; Vicki Ellingrod; akathisia; D2 receptor; pharmacologic effects; worsening psychosis
Sections
Author and Disclosure Information

 

James J. Gugger, PharmD, BCPP
Dr. Gugger is Assistant Clinical Professor, St. John’s University, College of Pharmacy and Allied Health Professions, Queens, NY
Courtney L. Tam, PharmD
Dr. Tam is Pharmacy Practice Resident, St. John’s University, College of Pharmacy and Allied Health Professions, Queens, NY
Charles R. Ashby, Jr, PhD
Dr. Ashby is Professor, St. John’s University, College of Pharmacy and Allied Health Professions, Queens, NY

Vicki L. Ellingrod, PharmD, BCPP, FCCP
Series Editor

Author and Disclosure Information

 

James J. Gugger, PharmD, BCPP
Dr. Gugger is Assistant Clinical Professor, St. John’s University, College of Pharmacy and Allied Health Professions, Queens, NY
Courtney L. Tam, PharmD
Dr. Tam is Pharmacy Practice Resident, St. John’s University, College of Pharmacy and Allied Health Professions, Queens, NY
Charles R. Ashby, Jr, PhD
Dr. Ashby is Professor, St. John’s University, College of Pharmacy and Allied Health Professions, Queens, NY

Vicki L. Ellingrod, PharmD, BCPP, FCCP
Series Editor

Article PDF
Article PDF

Discuss this article at www.facebook.com/CurrentPsychiatry

 

Practice Points

Aripiprazole may interact preferentially with distinct conformations of the D2 receptor, leading to a spectrum of pharmacologic effects, including acting as a full agonist, partial agonist, or antagonist.

Clinical predictors of aripiprazole-associated worsening of psychosis include low baseline level of psychopathology and previous treatment with high-dose antipsychotics.

• Rapid transition from a medication with significant anticholinergic properties to 1 without these properties may result in symptoms of activation, including restlessness, insomnia, and anxiety, which can be mistaken for worsening psychosis.

Akathisia, a common adverse effect of aripiprazole, may masquerade as treatment-emergent worsening of psychotic symptoms.

Mr. N, age 29, presents to the emergency department at the urging of his family because of poor self-care, bizarre behavior, and disturbed sleep. He first experienced psychiatric symptoms 10 years ago after his mother died. He became dysphoric and paranoid, displaying bizarre responses and behaviors with poor self-care and a gradual functional decline. He has been taking sertraline, 100 mg/d, for 10 years.

Upon arrival at the hospital’s inpatient unit, Mr. N is unkempt, oddly related, and paranoid. His affect is constricted. Mr. N displays thought blocking and possibly is responding to internal stimuli. Sertraline is continued and haloperidol, 1 mg/d, is initiated. For the next 2 weeks, Mr. N continues to be oddly related, irritable, and paranoid, and experiences disturbed sleep and thought blocking. After an episode of impulsive aggression, the treatment team initiates aripiprazole, which is titrated to 30 mg/d for 1 week. Mr. N’s clinical status worsens; he is menacing toward other patients and his thinking is more disorganized, with loose associations and ideas of reference. He requires 4 injections of IM haloperidol, 5 mg, and several visits to the seclusion room over the next week. Haloperidol is increased to 30 mg/d over the next 10 days, then aripiprazole is discontinued because of a putative drug interaction with haloperidol. Following the medication changes Mr. N demonstrates better behavioral control, but still is grossly psychotic. While awaiting transfer to a state hospital, Mr. N receives a trial of olanzapine, 20 to 40 mg/d, for 2 weeks without significant benefit.

Several clinical trials demonstrate a significant reduction in intensity of psychotic symptoms with aripiprazole, which has a unique mechanism of action.1 However, since its FDA approval in 2002, several case reports have described treatment-emergent psychotic symptoms associated with aripiprazole initiation. Over the past 40 years, reports of worsening psychosis associated with antipsychotics have been limited to patients with schizophrenia who were taking high dosages or who had high plasma concentrations, when anticholinergic delirium may have explained increased psychotic symptoms.2-4

How can a drug effectively treat psychotic symptoms and occasionally worsen them? In this article, we discuss the relevant pharmacology and clinical literature on aripiprazole and try to make sense of this apparent paradox.

Unique pharmacologic profile

Antipsychotics have been reported to be either neutral antagonists or inverse agonists at the D2 receptor, based on in vitro data.5 Aripiprazole and its main metabolite, dehydroaripiprazole, originally were described as partial agonists at D2 dopamine receptors.6,7 However, it appears aripiprazole’s pharmacologic action is better explained by the concept of functional selectivity. Aripiprazole may interact preferentially with distinct conformations of the D2 receptor, leading to a spectrum of pharmacologic effects, including acting as a full agonist, partial agonist, or antagonistic.5

Researchers have hypothesized that the pathophysiology of schizophrenia may, in part, be caused by dysfunction of mesocorticolimbic dopaminergic neurons characterized by an enhanced sensitivity of postsynaptic D2 receptors and increased sensitivity to dopaminergic drugs.8,9 In addition, chronic treatment with a D2 receptor antagonist is associated with increases in postsynaptic dopamine receptor density (ie, an increase in receptor reserve).10,11 Upregulation of D2 receptors may explain several features seen in patients chronically treated with antipsychotics, including tardive dyskinesia12 and rapid psychotic relapse after discontinuing an antipsychotic (supersensitivity psychosis).13 Because chronic antipsychotic treatment leads to high postsynaptic receptor reserve, aripiprazole initiation may produce overactivation of D2 receptors, which might worsen a patient’s condition.14 In vitro data15-18 and clinical observations indicate that aripiprazole has intrinsic efficacy at D2 receptors, as do clinical observations, such as:

 

 

  • its propensity to reduce serum prolactin19
  • a decreased likelihood of producing extrapyramidal side effects despite >80% occupancy of D2 receptors6
  • case reports documenting aripiprazole-associated mania,20 improvement of risperidone-associated cognitive impairment,21 and pathologic gambling.22

Emergence or worsening of psychotic symptoms or a marginal antipsychotic effect may occur if aripiprazole is indeed a postsynaptic D2 receptor agonist. An individual patient’s outcome likely would depend on his or her sensitivity to psychosis and concurrent or previous exposure to a D2 receptor antagonist. For example, stimulation of postsynaptic D2 receptors may be further augmented if the dosage of the previous antipsychotic was reduced or withdrawn before initiating aripiprazole because additional receptors would be available for interaction with aripiprazole.

 

 

 

Case reports

A literature review revealed 23 reports of treatment-emergent psychosis associated with aripiprazole initiation (Table). The mean age of the patients was 47 (range: 17 to 69) and 57% were men. Most patients (87%) were diagnosed with a schizophrenia-spectrum illness before aripiprazole initiation. Most (57%) had mild, stable, or no psychotic symptoms before aripiprazole initiation. Most were receiving relatively high doses of antipsychotics (average chlorpromazine equivalents [CPZE]: 648 mg/d) before aripiprazole initiation. This medication was either decreased or discontinued in 70% of patients.

Emergence or worsening of psychotic symptoms included agitation, aggressive behavior, and increased psychomotor activity. However, akathisia evaluation was described in only 2 reports: 1 author identified akathisia symptoms, but attributed them to a concomitant antipsychotic (fluphenazine)23 and the other report specifically excluded the possibility of akathisia.24 Two systematic studies have attempted to establish risk factors for aripiprazole-associated worsening psychosis (Box).14,25

In our literature review, the mean final dose of aripiprazole was 21.5 mg/d (range: 2 to 60 mg/d). In the cases describing subsequent treatment, all but 1 patient were switched to another antipsychotic, including 2 whose psychotic symptoms stabilized with continuation of aripiprazole and addition of a second antipsychotic. Interestingly, in the case reported by Adan-Manes et al,26 initial treatment with aripiprazole monotherapy was efficacious, but a subsequent trial of adjunctive aripiprazole resulted in worsening psychosis.

Table

Case reports: Treatment-emergent psychosis associated with aripiprazole

 

StudyAge, sexDiagnosisBefore aripiprazole initiationPre-aripiprazole treatmentAripiprazole doseConcomitant psychotropic treatmentSubsequent treatment
Chiu et al, 2011a39, MSchizophreniaPsychiatrically stable, tardive dystoniaClozapine, 300 mg/d10 mg/dValproic acid, 1,000 mg/d, clonazepam, 2 mg/d, mephenoxalone, 800 mg/dClozapine
Ekinci et al, 2010b17, MADHDInattention and impulsive aggressionTapered and discontinued risperidone, 2.5 mg/d5 mg/dMethylphenidate, 54 mg/dRisperidone, 2 mg/d, methylphenidate, 36 mg/d
Selvaraj et al, 2010c49, FChronic depressionDepressive symptoms, suicidal ideationNone stated2 mg/dDuloxetine, 80 mg/d, clonazepam, 2 mg/dDuloxetine, 120 mg/d
Adan-Manes et al, 2009d23, MSchizophreniaNo psychotic symptomsAbrupt decrease of amisulpride dose from 800 mg/d to 400 mg/d20 mg/dBiperiden, 4 mg/dAmisulpride, 800 mg/d
Cho et al, 2009e45, FSchizophreniaPersistent psychotic symptoms, new onset diabetes with acute ketoacidosisHaloperidol, 20 mg/d, abrupt clozapine discontinuation15 mg/dValproic acid, nortriptylineMolindone, 150 mg/d
Ahuja et al, 2007f35, FSchizoaffective disorderStable before medication changeTapered amisulpride, 400 mg/d, over 6 weeks15 mg/dNoneAmisulpride, 600 mg/d
Lea et al, 2007g57, MSchizophreniaPersistent psychotic symptoms, treatment resistance, recent recovery from NMSDiscontinued ziprasidone, 200 mg/d30 mg/dLorazepam, 2 mg/d, amantadine, 100 mg, sertraline, 50 mg/dClozapine
Lea et al, 2007g49, MSchizoaffective disorderDelusions, verbal aggression, substance abuse, HCVDecreased quetiapine dose from 800 mg/d to 400 mg/d15 mg/dDivalproex, 1,000 mg/d, fluvoxamine, 200 mg/d, clonazepam, 2 mg/dLithium, quetiapine, 500 mg/d, haloperidol, 2 mg/d
Lea et al, 2007g60, MSchizophreniaDelusions, labile mood, aggressionRisperidone, 3 mg/d, interruption of fluphenazine, 75 mg/d20 mg/dDivalproex, 4,500 mg/d, benztropine, 3 mg/dNot discussed
Raja, 2007h30, MSchizoaffective disorderNegative symptoms, otherwise stable, recent citalopram discontinuationDiscontinued amisulpride, 800 mg/d over 2 weeks30 mg/dLithiumAmisulpride, 500 mg/d
Raja, 2007h69, FBipolar disorderHistory of multiple relapses; presented with tremor, akathisia, weight gainDiscontinued risperidone, 2 mg/d, over 2 weeks15 mg/dLithiumRisperidone, 4 mg
Raja, 2007h59, FSchizophreniaNegative symptoms, otherwise stableReduced risperidone dosage from 5 mg/d to 4 mg/d7.5 mg/dNoneRisperidone, 5 mg/d
Thone, 2007i31, MSchizophreniaConfusion, agitation, delusions worsened with aripiprazole dose increaseNone60 mg/dNoneAripiprazole dose reduction to 15 mg/d, olanzapine, 10 mg/d
Glick et al, 2006j55, FSchizophreniaStable before medication changeTapered and discontinued thioridazine, 600 mg/d, over 3 months30 mg/dNoneChlorpromazine, 200 mg/d, aripiprazole, 30 mg/d
Glick et al, 2006j52, MSchizophreniaNegative symptomsDecreased olanzapine dose from 30 mg/d to 20 mg/d30 mg/dNoneOlanzapine, 30 mg/d
Barnas et al, 2005k57, FSchizoaffective disorderStable before medication changeDiscontinued perphenazine, 8 mg/d30 mg/dNoneQuetiapine, 350 mg/d
DeQuardo, 2004l54, MSchizophreniaHistory of aggression, residual paranoia, severe EPSHaloperidol, 200 mg/d15 mg/dBenztropineHaloperidol
DeQuardo, 2004l51, MSchizophreniaHistory of aggression, persistent psychotic symptoms, treatment resistanceOlanzapine, 60 mg/d10 mg/dNoneOlanzapine
Ramaswamy et al, 2004m43, FSchizoaffective disorderPsychiatrically stable, multiple medication changes, including substituting carbamazepine for valproic acidDiscontinued ziprasidone, 160 mg/d, discontinued quetiapine, 400 mg/d, over 2 weeks30 mg/dPropranolol, 30 mg/d, l-thyroxine, .05 mg/d, carbamazepine, 600 mg/dNot available
Ramaswawamy et al, 2004m57, FSchizoaffective disorderHistory of multiple hospitalizations, but stable before medication changeDecreased olanzapine dose from 20 mg/d to 15 mg/d30 mg/dValproic acid, 2,000 mg/dZiprasidone
Ramaswawamy et al, 2004m67, FSchizophreniaRemote hospitalizations, recent worsened psychosisDecreased ziprasidone dose from 200 mg/d to 160 mg/d 2 months previously30 mg/dCarbamazepine, 200 mg/dNot discussed
Ramaswamy et al, 2004m46, MSchizophreniaPersistent delusions while receiving risperidone, TDRisperidone, 3 mg/d15 mg/dValproic acid, 1,500 mg/dRisperidone, 3 mg/d
Reeves et al, 2004n50, MSchizoaffective disorderRelatively stable with nonthreatening delusions, hallucinationsQuetiapine, 800 mg/d30 mg/dDivalproex, 2,000 mg/dOlanzapine, 20 mg/d
ADHD: attention-deficit/hyperactivity disorder; EPS: extrapyramidal symptoms; HCV: hepatitis C virus; NMS: neuroleptic malignant syndrome; TD: tardive dyskinesia Source:
References
a. Chiu YH, Chen CH, Lu ML. Worsening psychosis after adding aripiprazole to clozapine. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35(1):291-292.
b. Ekinci O, Sabuncuoglu O. Psychosis associated with switching from risperidone to aripiprazole in an adolescent on methylphenidate treatment. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35(2):648-649.
c. Selvaraj V, Ramaswamy S, Sharma A, et al. New-onset psychosis and emergence of suicidal ideation with aripiprazole. Am J Psychiatry. 2010;167(12):1535-1536.
d. Adan-Manes J, Garcia-Parajua P. Aripiprazole in combination with other antipsychotic drugs may worsen psychosis. J Clin Pharm Ther. 2009;34(2):245-246.
e. Cho DY, Lindenmayer JP. Aripiprazole-induced agitation after clozapine discontinuation: a case report. J Clin Psychiatry. 2009;70(1):141-143.
f. Ahuja N, Lloyd AJ. Aripiprazole and worsening of psychosis: a case report. J Clin Psychiatry. 2007;68(5):805-806.
g. Lea JW, Stoner SC, Lafollette J. Agitation associated with aripiprazole initiation. Pharmacotherapy. 2007;27(9):1339-1342.
h. Raja M. Improvement or worsening of psychotic symptoms after treatment with low doses of aripiprazole. Int J Neuropsychopharmacol. 2007;10(1):107-110.
i. Thone J. Worsened agitation and confusion in schizophrenia subsequent to high-dose aripiprazole. J Neuropsychiatry Clin Neurosci. 2007;19(4):481-482.
j. Glick ID, Duggal V, Hodulik C. Aripiprazole as a dopamine partial agonist: positive and negative effects. J Clin Psychopharmacol. 2006;26(1):101-103.
k. Barnas ME, Hussain N, Petrides G. Treatment-emergent psychosis with aripiprazole. J Clin Psychiatry. 2005;66(10):1339.
l. DeQuardo JR. Worsened agitation with aripiprazole: adverse effect of dopamine partial agonism? J Clin Psychiatry. 2004;65(1):132-133.
m. Ramaswamy S, Vijay D, William M, et al. Aripiprazole possibly worsens psychosis. Int Clin Psychopharmacol. 2004;19(1):45-48.
n. Reeves RR, Mack JE. Worsening schizoaffective disorder with aripiprazole. Am J Psychiatry. 2004;161(7):1308.
 

 

 

 

Clinical predictors of aripiprazole-associated psychotic symptoms

Takeuchi et al14 aimed to establish predictors of worsening psychosis in a naturalistic setting where patients slowly transitioned to aripiprazole from previous antipsychotic treatment. Patients were required to be on a stable dose of an antipsychotic before participating in the study. Aripiprazole was started at 12 mg/d for 2 weeks with flexible dosing from weeks 2 to 52. Previous antipsychotic therapy was reduced biweekly by 25%. The incidence of worsening psychopathology after aripiprazole initiation was higher in the group of patients who had previously received high-dose antipsychotic therapy (average chlorpromazine equivalents [CPZE]: 727 mg/d) compared with the group on low dosages (average CPZE: 382 mg/d). It is possible that previous high-dose antipsychotic therapy was indicative of more significant baseline psychopathology; however, the worsened group and stabilized group had similar baseline Clinical Global Impressions-Severity scores.

Pae et al25 aimed to find predictors of worsening psychosis with aripiprazole in patients whose previous antipsychotic therapy was immediately discontinued. They found lower baseline disease severity was associated with significant worsening during the first month of aripiprazole treatment.

Other potential explanations

 

Aripiprazole’s manufacturer reported the incidence of psychosis-related adverse events in an analysis of 9 randomized schizophrenia trials.27 The rates of psychosis-related adverse events ranged from 0.6% to 18%, but there was no apparent relationship to study design or method of transitioning to aripiprazole. Rates of psychosis-related adverse events were similar between aripiprazole and the control group (placebo in 3 studies, another antipsychotic in 2 studies).

Emergence or worsening of psychotic symptoms temporally associated with aripiprazole initiation does not necessarily imply causation. As in Mr. N’s case, it is not always possible to determine whether worsening psychosis is the natural disease course or a treatment effect. In addition, it is not possible to differentiate lack of efficacy from a true propensity for aripiprazole to worsen psychosis.

 

It also is conceivable discontinuation or dosage reduction of a previous antipsychotic would worsen psychotic symptoms or cause side effects. When significant changes in psychopathology or side effects develop during the transition from 1 antipsychotic to another, it is difficult to determine etiology. Specifically, rapid transition from a medication with significant anticholinergic and antihistaminic properties—such as quetiapine or olanzapine—to 1 without these properties—such as aripiprazole—may result in symptoms of activation, including restlessness, insomnia, and anxiety. Consequently, these symptoms could be mistaken for worsening psychosis.28 Only 1 patient in this series was reported to abruptly discontinue an antipsychotic with significant anticholinergic properties (clozapine) before initiating aripiprazole.24 Studies by Takeuchi et al14 and Pae et al25 did not report the relative baseline use of antipsychotic medication with anticholinergic properties.

In a pooled analysis of treatment-emergent adverse events in 5 randomized clinical trials of patients receiving aripiprazole for acute relapse of schizophrenia, the incidence of akathisia was 10%, although it is not clear if this is a dose-related adverse effect.29 Because akathisia may be confused for worsening psychosis,30 it is possible akathisia was mistakenly identified as worsening psychotic symptoms in Mr. N’s case, as well as several reports from our literature review.

Covert akathisia is unlikely to explain worsening psychopathology observed in all patients in our literature review because confusion of akathisia and worsening psychosis is not a widespread phenomenon. In a post hoc analysis of pooled safety data from aripiprazole trials, Kane et al31 did not find a correlation between presence of akathisia and aripiprazole efficacy as measured by the Positive and Negative Syndrome Scale (PANSS) total, PANSS positive, PANSS negative, Clinical Global Impressions-Severity, Clinical Global Impressions-Improvement, and percentage of responders. Pae et al25 also noted there was no correlation between scores on the Barnes Akathisia Rating Scale and worsening psychopathology in patients switched to aripiprazole.

 

An antagonist always is an antagonist and clinicians have appreciated this concept since the days of chlorpromazine. The activity of aripiprazole, however, is on a pharmacologic continuum between a neutral antagonist and full agonist and currently there is no way to precisely determine the level of D2 receptor agonist action in a patient.

Although it is interesting to speculate that aripiprazole’s D2 receptor agonist action may contribute to worsening psychosis,32-34 there are other plausible explanations to consider. Rapid transition from a drug with significant anticholinergic properties and aripiprazole-associated akathisia may contribute to worsening psychopathology in patients starting aripiprazole. Because covert side effects may be incorrectly identified as psychotic agitation, we cannot exclude this as a possible etiologic factor in Mr. N’s case as well as the cases in our literature review.

 

 

Related Resource

 

  • Abilify [package insert]. Princeton, NJ: Bristol-Myers Squibb; 2011.

Drug Brand Names

 

  • Amantadine • Symmetrel
  • Aripiprazole • Abilify
  • Benztropine • Cogentin
  • Biperiden • Akineton
  • Carbamazepine • Tegretol
  • Chlorpromazine • Thorazine
  • Clonazepam • Klonopin
  • Clozapine • Clozaril
  • Divalproex • Depakote
  • Duloxetine • Cymbalta
  • Fluphenazine • Permitil, Prolixin
  • Fluvoxamine • Luvox
  • Haloperidol • Haldol
  • Lithium • Eskalith, Lithobid
  • Lorazepam • Ativan
  • Nortriptyline • Aventyl, Pamelor
  • Methylphenidate • Concerta
  • Molindone • Moban
  • Olanzapine • Zyprexa
  • Perphenazine • Trilafon
  • Propranolol • Inderal
  • Quetiapine • Seroquel
  • Risperidone • Risperdal
  • Sertraline • Zoloft
  • Thioridazine • Mellaril
  • Thyroxine • Synthroid
  • Valproic acid • Depakene
  • Ziprasidone • Geodon

Disclosure

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

Discuss this article at www.facebook.com/CurrentPsychiatry

 

Practice Points

Aripiprazole may interact preferentially with distinct conformations of the D2 receptor, leading to a spectrum of pharmacologic effects, including acting as a full agonist, partial agonist, or antagonist.

Clinical predictors of aripiprazole-associated worsening of psychosis include low baseline level of psychopathology and previous treatment with high-dose antipsychotics.

• Rapid transition from a medication with significant anticholinergic properties to 1 without these properties may result in symptoms of activation, including restlessness, insomnia, and anxiety, which can be mistaken for worsening psychosis.

Akathisia, a common adverse effect of aripiprazole, may masquerade as treatment-emergent worsening of psychotic symptoms.

Mr. N, age 29, presents to the emergency department at the urging of his family because of poor self-care, bizarre behavior, and disturbed sleep. He first experienced psychiatric symptoms 10 years ago after his mother died. He became dysphoric and paranoid, displaying bizarre responses and behaviors with poor self-care and a gradual functional decline. He has been taking sertraline, 100 mg/d, for 10 years.

Upon arrival at the hospital’s inpatient unit, Mr. N is unkempt, oddly related, and paranoid. His affect is constricted. Mr. N displays thought blocking and possibly is responding to internal stimuli. Sertraline is continued and haloperidol, 1 mg/d, is initiated. For the next 2 weeks, Mr. N continues to be oddly related, irritable, and paranoid, and experiences disturbed sleep and thought blocking. After an episode of impulsive aggression, the treatment team initiates aripiprazole, which is titrated to 30 mg/d for 1 week. Mr. N’s clinical status worsens; he is menacing toward other patients and his thinking is more disorganized, with loose associations and ideas of reference. He requires 4 injections of IM haloperidol, 5 mg, and several visits to the seclusion room over the next week. Haloperidol is increased to 30 mg/d over the next 10 days, then aripiprazole is discontinued because of a putative drug interaction with haloperidol. Following the medication changes Mr. N demonstrates better behavioral control, but still is grossly psychotic. While awaiting transfer to a state hospital, Mr. N receives a trial of olanzapine, 20 to 40 mg/d, for 2 weeks without significant benefit.

Several clinical trials demonstrate a significant reduction in intensity of psychotic symptoms with aripiprazole, which has a unique mechanism of action.1 However, since its FDA approval in 2002, several case reports have described treatment-emergent psychotic symptoms associated with aripiprazole initiation. Over the past 40 years, reports of worsening psychosis associated with antipsychotics have been limited to patients with schizophrenia who were taking high dosages or who had high plasma concentrations, when anticholinergic delirium may have explained increased psychotic symptoms.2-4

How can a drug effectively treat psychotic symptoms and occasionally worsen them? In this article, we discuss the relevant pharmacology and clinical literature on aripiprazole and try to make sense of this apparent paradox.

Unique pharmacologic profile

Antipsychotics have been reported to be either neutral antagonists or inverse agonists at the D2 receptor, based on in vitro data.5 Aripiprazole and its main metabolite, dehydroaripiprazole, originally were described as partial agonists at D2 dopamine receptors.6,7 However, it appears aripiprazole’s pharmacologic action is better explained by the concept of functional selectivity. Aripiprazole may interact preferentially with distinct conformations of the D2 receptor, leading to a spectrum of pharmacologic effects, including acting as a full agonist, partial agonist, or antagonistic.5

Researchers have hypothesized that the pathophysiology of schizophrenia may, in part, be caused by dysfunction of mesocorticolimbic dopaminergic neurons characterized by an enhanced sensitivity of postsynaptic D2 receptors and increased sensitivity to dopaminergic drugs.8,9 In addition, chronic treatment with a D2 receptor antagonist is associated with increases in postsynaptic dopamine receptor density (ie, an increase in receptor reserve).10,11 Upregulation of D2 receptors may explain several features seen in patients chronically treated with antipsychotics, including tardive dyskinesia12 and rapid psychotic relapse after discontinuing an antipsychotic (supersensitivity psychosis).13 Because chronic antipsychotic treatment leads to high postsynaptic receptor reserve, aripiprazole initiation may produce overactivation of D2 receptors, which might worsen a patient’s condition.14 In vitro data15-18 and clinical observations indicate that aripiprazole has intrinsic efficacy at D2 receptors, as do clinical observations, such as:

 

 

  • its propensity to reduce serum prolactin19
  • a decreased likelihood of producing extrapyramidal side effects despite >80% occupancy of D2 receptors6
  • case reports documenting aripiprazole-associated mania,20 improvement of risperidone-associated cognitive impairment,21 and pathologic gambling.22

Emergence or worsening of psychotic symptoms or a marginal antipsychotic effect may occur if aripiprazole is indeed a postsynaptic D2 receptor agonist. An individual patient’s outcome likely would depend on his or her sensitivity to psychosis and concurrent or previous exposure to a D2 receptor antagonist. For example, stimulation of postsynaptic D2 receptors may be further augmented if the dosage of the previous antipsychotic was reduced or withdrawn before initiating aripiprazole because additional receptors would be available for interaction with aripiprazole.

 

 

 

Case reports

A literature review revealed 23 reports of treatment-emergent psychosis associated with aripiprazole initiation (Table). The mean age of the patients was 47 (range: 17 to 69) and 57% were men. Most patients (87%) were diagnosed with a schizophrenia-spectrum illness before aripiprazole initiation. Most (57%) had mild, stable, or no psychotic symptoms before aripiprazole initiation. Most were receiving relatively high doses of antipsychotics (average chlorpromazine equivalents [CPZE]: 648 mg/d) before aripiprazole initiation. This medication was either decreased or discontinued in 70% of patients.

Emergence or worsening of psychotic symptoms included agitation, aggressive behavior, and increased psychomotor activity. However, akathisia evaluation was described in only 2 reports: 1 author identified akathisia symptoms, but attributed them to a concomitant antipsychotic (fluphenazine)23 and the other report specifically excluded the possibility of akathisia.24 Two systematic studies have attempted to establish risk factors for aripiprazole-associated worsening psychosis (Box).14,25

In our literature review, the mean final dose of aripiprazole was 21.5 mg/d (range: 2 to 60 mg/d). In the cases describing subsequent treatment, all but 1 patient were switched to another antipsychotic, including 2 whose psychotic symptoms stabilized with continuation of aripiprazole and addition of a second antipsychotic. Interestingly, in the case reported by Adan-Manes et al,26 initial treatment with aripiprazole monotherapy was efficacious, but a subsequent trial of adjunctive aripiprazole resulted in worsening psychosis.

Table

Case reports: Treatment-emergent psychosis associated with aripiprazole

 

StudyAge, sexDiagnosisBefore aripiprazole initiationPre-aripiprazole treatmentAripiprazole doseConcomitant psychotropic treatmentSubsequent treatment
Chiu et al, 2011a39, MSchizophreniaPsychiatrically stable, tardive dystoniaClozapine, 300 mg/d10 mg/dValproic acid, 1,000 mg/d, clonazepam, 2 mg/d, mephenoxalone, 800 mg/dClozapine
Ekinci et al, 2010b17, MADHDInattention and impulsive aggressionTapered and discontinued risperidone, 2.5 mg/d5 mg/dMethylphenidate, 54 mg/dRisperidone, 2 mg/d, methylphenidate, 36 mg/d
Selvaraj et al, 2010c49, FChronic depressionDepressive symptoms, suicidal ideationNone stated2 mg/dDuloxetine, 80 mg/d, clonazepam, 2 mg/dDuloxetine, 120 mg/d
Adan-Manes et al, 2009d23, MSchizophreniaNo psychotic symptomsAbrupt decrease of amisulpride dose from 800 mg/d to 400 mg/d20 mg/dBiperiden, 4 mg/dAmisulpride, 800 mg/d
Cho et al, 2009e45, FSchizophreniaPersistent psychotic symptoms, new onset diabetes with acute ketoacidosisHaloperidol, 20 mg/d, abrupt clozapine discontinuation15 mg/dValproic acid, nortriptylineMolindone, 150 mg/d
Ahuja et al, 2007f35, FSchizoaffective disorderStable before medication changeTapered amisulpride, 400 mg/d, over 6 weeks15 mg/dNoneAmisulpride, 600 mg/d
Lea et al, 2007g57, MSchizophreniaPersistent psychotic symptoms, treatment resistance, recent recovery from NMSDiscontinued ziprasidone, 200 mg/d30 mg/dLorazepam, 2 mg/d, amantadine, 100 mg, sertraline, 50 mg/dClozapine
Lea et al, 2007g49, MSchizoaffective disorderDelusions, verbal aggression, substance abuse, HCVDecreased quetiapine dose from 800 mg/d to 400 mg/d15 mg/dDivalproex, 1,000 mg/d, fluvoxamine, 200 mg/d, clonazepam, 2 mg/dLithium, quetiapine, 500 mg/d, haloperidol, 2 mg/d
Lea et al, 2007g60, MSchizophreniaDelusions, labile mood, aggressionRisperidone, 3 mg/d, interruption of fluphenazine, 75 mg/d20 mg/dDivalproex, 4,500 mg/d, benztropine, 3 mg/dNot discussed
Raja, 2007h30, MSchizoaffective disorderNegative symptoms, otherwise stable, recent citalopram discontinuationDiscontinued amisulpride, 800 mg/d over 2 weeks30 mg/dLithiumAmisulpride, 500 mg/d
Raja, 2007h69, FBipolar disorderHistory of multiple relapses; presented with tremor, akathisia, weight gainDiscontinued risperidone, 2 mg/d, over 2 weeks15 mg/dLithiumRisperidone, 4 mg
Raja, 2007h59, FSchizophreniaNegative symptoms, otherwise stableReduced risperidone dosage from 5 mg/d to 4 mg/d7.5 mg/dNoneRisperidone, 5 mg/d
Thone, 2007i31, MSchizophreniaConfusion, agitation, delusions worsened with aripiprazole dose increaseNone60 mg/dNoneAripiprazole dose reduction to 15 mg/d, olanzapine, 10 mg/d
Glick et al, 2006j55, FSchizophreniaStable before medication changeTapered and discontinued thioridazine, 600 mg/d, over 3 months30 mg/dNoneChlorpromazine, 200 mg/d, aripiprazole, 30 mg/d
Glick et al, 2006j52, MSchizophreniaNegative symptomsDecreased olanzapine dose from 30 mg/d to 20 mg/d30 mg/dNoneOlanzapine, 30 mg/d
Barnas et al, 2005k57, FSchizoaffective disorderStable before medication changeDiscontinued perphenazine, 8 mg/d30 mg/dNoneQuetiapine, 350 mg/d
DeQuardo, 2004l54, MSchizophreniaHistory of aggression, residual paranoia, severe EPSHaloperidol, 200 mg/d15 mg/dBenztropineHaloperidol
DeQuardo, 2004l51, MSchizophreniaHistory of aggression, persistent psychotic symptoms, treatment resistanceOlanzapine, 60 mg/d10 mg/dNoneOlanzapine
Ramaswamy et al, 2004m43, FSchizoaffective disorderPsychiatrically stable, multiple medication changes, including substituting carbamazepine for valproic acidDiscontinued ziprasidone, 160 mg/d, discontinued quetiapine, 400 mg/d, over 2 weeks30 mg/dPropranolol, 30 mg/d, l-thyroxine, .05 mg/d, carbamazepine, 600 mg/dNot available
Ramaswawamy et al, 2004m57, FSchizoaffective disorderHistory of multiple hospitalizations, but stable before medication changeDecreased olanzapine dose from 20 mg/d to 15 mg/d30 mg/dValproic acid, 2,000 mg/dZiprasidone
Ramaswawamy et al, 2004m67, FSchizophreniaRemote hospitalizations, recent worsened psychosisDecreased ziprasidone dose from 200 mg/d to 160 mg/d 2 months previously30 mg/dCarbamazepine, 200 mg/dNot discussed
Ramaswamy et al, 2004m46, MSchizophreniaPersistent delusions while receiving risperidone, TDRisperidone, 3 mg/d15 mg/dValproic acid, 1,500 mg/dRisperidone, 3 mg/d
Reeves et al, 2004n50, MSchizoaffective disorderRelatively stable with nonthreatening delusions, hallucinationsQuetiapine, 800 mg/d30 mg/dDivalproex, 2,000 mg/dOlanzapine, 20 mg/d
ADHD: attention-deficit/hyperactivity disorder; EPS: extrapyramidal symptoms; HCV: hepatitis C virus; NMS: neuroleptic malignant syndrome; TD: tardive dyskinesia Source:
References
a. Chiu YH, Chen CH, Lu ML. Worsening psychosis after adding aripiprazole to clozapine. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35(1):291-292.
b. Ekinci O, Sabuncuoglu O. Psychosis associated with switching from risperidone to aripiprazole in an adolescent on methylphenidate treatment. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35(2):648-649.
c. Selvaraj V, Ramaswamy S, Sharma A, et al. New-onset psychosis and emergence of suicidal ideation with aripiprazole. Am J Psychiatry. 2010;167(12):1535-1536.
d. Adan-Manes J, Garcia-Parajua P. Aripiprazole in combination with other antipsychotic drugs may worsen psychosis. J Clin Pharm Ther. 2009;34(2):245-246.
e. Cho DY, Lindenmayer JP. Aripiprazole-induced agitation after clozapine discontinuation: a case report. J Clin Psychiatry. 2009;70(1):141-143.
f. Ahuja N, Lloyd AJ. Aripiprazole and worsening of psychosis: a case report. J Clin Psychiatry. 2007;68(5):805-806.
g. Lea JW, Stoner SC, Lafollette J. Agitation associated with aripiprazole initiation. Pharmacotherapy. 2007;27(9):1339-1342.
h. Raja M. Improvement or worsening of psychotic symptoms after treatment with low doses of aripiprazole. Int J Neuropsychopharmacol. 2007;10(1):107-110.
i. Thone J. Worsened agitation and confusion in schizophrenia subsequent to high-dose aripiprazole. J Neuropsychiatry Clin Neurosci. 2007;19(4):481-482.
j. Glick ID, Duggal V, Hodulik C. Aripiprazole as a dopamine partial agonist: positive and negative effects. J Clin Psychopharmacol. 2006;26(1):101-103.
k. Barnas ME, Hussain N, Petrides G. Treatment-emergent psychosis with aripiprazole. J Clin Psychiatry. 2005;66(10):1339.
l. DeQuardo JR. Worsened agitation with aripiprazole: adverse effect of dopamine partial agonism? J Clin Psychiatry. 2004;65(1):132-133.
m. Ramaswamy S, Vijay D, William M, et al. Aripiprazole possibly worsens psychosis. Int Clin Psychopharmacol. 2004;19(1):45-48.
n. Reeves RR, Mack JE. Worsening schizoaffective disorder with aripiprazole. Am J Psychiatry. 2004;161(7):1308.
 

 

 

 

Clinical predictors of aripiprazole-associated psychotic symptoms

Takeuchi et al14 aimed to establish predictors of worsening psychosis in a naturalistic setting where patients slowly transitioned to aripiprazole from previous antipsychotic treatment. Patients were required to be on a stable dose of an antipsychotic before participating in the study. Aripiprazole was started at 12 mg/d for 2 weeks with flexible dosing from weeks 2 to 52. Previous antipsychotic therapy was reduced biweekly by 25%. The incidence of worsening psychopathology after aripiprazole initiation was higher in the group of patients who had previously received high-dose antipsychotic therapy (average chlorpromazine equivalents [CPZE]: 727 mg/d) compared with the group on low dosages (average CPZE: 382 mg/d). It is possible that previous high-dose antipsychotic therapy was indicative of more significant baseline psychopathology; however, the worsened group and stabilized group had similar baseline Clinical Global Impressions-Severity scores.

Pae et al25 aimed to find predictors of worsening psychosis with aripiprazole in patients whose previous antipsychotic therapy was immediately discontinued. They found lower baseline disease severity was associated with significant worsening during the first month of aripiprazole treatment.

Other potential explanations

 

Aripiprazole’s manufacturer reported the incidence of psychosis-related adverse events in an analysis of 9 randomized schizophrenia trials.27 The rates of psychosis-related adverse events ranged from 0.6% to 18%, but there was no apparent relationship to study design or method of transitioning to aripiprazole. Rates of psychosis-related adverse events were similar between aripiprazole and the control group (placebo in 3 studies, another antipsychotic in 2 studies).

Emergence or worsening of psychotic symptoms temporally associated with aripiprazole initiation does not necessarily imply causation. As in Mr. N’s case, it is not always possible to determine whether worsening psychosis is the natural disease course or a treatment effect. In addition, it is not possible to differentiate lack of efficacy from a true propensity for aripiprazole to worsen psychosis.

 

It also is conceivable discontinuation or dosage reduction of a previous antipsychotic would worsen psychotic symptoms or cause side effects. When significant changes in psychopathology or side effects develop during the transition from 1 antipsychotic to another, it is difficult to determine etiology. Specifically, rapid transition from a medication with significant anticholinergic and antihistaminic properties—such as quetiapine or olanzapine—to 1 without these properties—such as aripiprazole—may result in symptoms of activation, including restlessness, insomnia, and anxiety. Consequently, these symptoms could be mistaken for worsening psychosis.28 Only 1 patient in this series was reported to abruptly discontinue an antipsychotic with significant anticholinergic properties (clozapine) before initiating aripiprazole.24 Studies by Takeuchi et al14 and Pae et al25 did not report the relative baseline use of antipsychotic medication with anticholinergic properties.

In a pooled analysis of treatment-emergent adverse events in 5 randomized clinical trials of patients receiving aripiprazole for acute relapse of schizophrenia, the incidence of akathisia was 10%, although it is not clear if this is a dose-related adverse effect.29 Because akathisia may be confused for worsening psychosis,30 it is possible akathisia was mistakenly identified as worsening psychotic symptoms in Mr. N’s case, as well as several reports from our literature review.

Covert akathisia is unlikely to explain worsening psychopathology observed in all patients in our literature review because confusion of akathisia and worsening psychosis is not a widespread phenomenon. In a post hoc analysis of pooled safety data from aripiprazole trials, Kane et al31 did not find a correlation between presence of akathisia and aripiprazole efficacy as measured by the Positive and Negative Syndrome Scale (PANSS) total, PANSS positive, PANSS negative, Clinical Global Impressions-Severity, Clinical Global Impressions-Improvement, and percentage of responders. Pae et al25 also noted there was no correlation between scores on the Barnes Akathisia Rating Scale and worsening psychopathology in patients switched to aripiprazole.

 

An antagonist always is an antagonist and clinicians have appreciated this concept since the days of chlorpromazine. The activity of aripiprazole, however, is on a pharmacologic continuum between a neutral antagonist and full agonist and currently there is no way to precisely determine the level of D2 receptor agonist action in a patient.

Although it is interesting to speculate that aripiprazole’s D2 receptor agonist action may contribute to worsening psychosis,32-34 there are other plausible explanations to consider. Rapid transition from a drug with significant anticholinergic properties and aripiprazole-associated akathisia may contribute to worsening psychopathology in patients starting aripiprazole. Because covert side effects may be incorrectly identified as psychotic agitation, we cannot exclude this as a possible etiologic factor in Mr. N’s case as well as the cases in our literature review.

 

 

Related Resource

 

  • Abilify [package insert]. Princeton, NJ: Bristol-Myers Squibb; 2011.

Drug Brand Names

 

  • Amantadine • Symmetrel
  • Aripiprazole • Abilify
  • Benztropine • Cogentin
  • Biperiden • Akineton
  • Carbamazepine • Tegretol
  • Chlorpromazine • Thorazine
  • Clonazepam • Klonopin
  • Clozapine • Clozaril
  • Divalproex • Depakote
  • Duloxetine • Cymbalta
  • Fluphenazine • Permitil, Prolixin
  • Fluvoxamine • Luvox
  • Haloperidol • Haldol
  • Lithium • Eskalith, Lithobid
  • Lorazepam • Ativan
  • Nortriptyline • Aventyl, Pamelor
  • Methylphenidate • Concerta
  • Molindone • Moban
  • Olanzapine • Zyprexa
  • Perphenazine • Trilafon
  • Propranolol • Inderal
  • Quetiapine • Seroquel
  • Risperidone • Risperdal
  • Sertraline • Zoloft
  • Thioridazine • Mellaril
  • Thyroxine • Synthroid
  • Valproic acid • Depakene
  • Ziprasidone • Geodon

Disclosure

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

References

 

1. Citrome L. A review of aripiprazole in the treatment of patients with schizophrenia or bipolar I disorder. Neuropsychiatr Dis Treat. 2006;2(4):427-443.

2. Chong SA, Tan CH, Lee HS. Worsening of psychosis with clozapine and selective serotonin reuptake inhibitor combination: two case reports. J Clin Psychopharmacol. 1997;17(1):68-69.

3. Bowers MB Jr, Swigar ME. Psychotic patients who become worse on neuroleptics. J Clin Psychopharmacol. 1988;8(6):417-421.

4. Tornatore FL, Lee D, Sramek JJ. Psychotic exacerbation with haloperidol. Drug Intell Clin Pharm. 1981;15(3):209-213.

5. Beaulieu JM, Gainetdinov RR. The physiology signaling, and pharmacology of dopamine receptors. Pharmacol Rev. 2011;63(1):182-217.

6. Grunder G, Carlsson A, Wong DF. Mechanism of new antipsychotic medications: occupancy is not just antagonism. Arch Gen Psychiatry. 2003;60(10):974-977.

7. Wood MD, Scott C, Clarke K, et al. Aripiprazole and its human metabolite are partial agonists at the human dopamine D2 receptor, but the rodent metabolite displays antagonist properties. Eur J Pharmacol. 2006;546(1-3):88-94.

8. Seeman P, Weinshenker D, Quirion R, et al. Dopamine supersensitivity correlates with D2High states, implying many paths to psychosis. Proc Natl Acad Sci U S A. 2005;102(9):3513-3518.

9. Seeman P, Ko F, Jack E, et al. Consistent with dopamine supersensitivity, RGS9 expression is diminished in the amphetamine-treated animal model of schizophrenia and in postmortem schizophrenia brain. Synapse. 2007;61(5):303-309.

10. Burt DR, Creese I, Snyder SH. Antischizophrenic drugs: chronic treatment elevates dopamine receptor binding in brain. Science. 1977;196(4287):326-328.

11. Silvestri S, Seeman MV, Negrete JC, et al. Increased dopamine D2 receptor binding after long-term treatment with antipsychotics in humans: a clinical PET study. Psychopharmacology (Berl). 2000;152(2):174-180.

12. Sayers AC, Bürki HR, Ruch W, et al. Neuroleptic-induced hypersensitivity of striatal dopamine receptors in the rat as a model of tardive dyskinesias. Effects of clozapine, haloperidol, loxapine and chlorpromazine. Psychopharmacologia. 1975;41(2):97-104.

13. Moncrieff J. Does antipsychotic withdrawal provoke psychosis? Review of the literature on rapid onset psychosis (supersensitivity psychosis) and withdrawal-related relapse. Acta Psychiatr Scand. 2006;114(1):3-13.

14. Takeuchi H, Uchida H, Suzuki T, et al. Predictors of clinical worsening after a switch to aripiprazole in patients with schizophrenia: a 1-year naturalistic follow-up study. J Clin Psychopharmacol. 2009;29(4):394-395.

15. Shapiro DA, Renock S, Arrington E, et al. Aripiprazole, a novel atypical antipsychotic drug with a unique and robust pharmacology. Neuropsychopharmacology. 2003;28(8):1400-1411.

16. Urban JD, Vargas GA, von Zastrow M, et al. Aripiprazole has functionally selective actions at dopamine D2 receptor-mediated signaling pathways. Neuropsychopharmacology. 2007;32(1):67-77.

17. Klewe IV, Nielsen SM, Tarpo L, et al. Recruitment of beta-arrestin2 to the dopamine D2 receptor: Insights into anti-psychotic and anti-parkinsonian drug receptor signaling. Neuropharmacology. 2008;54(8):1215-1222.

18. Masri B, Salahpour A, Didriksen M, et al. Antagonism of dopamine D2 receptor/beta-arrestin 2 interaction is a common property of clinically effective antipsychotics. Proc Natl Acad Sci U S A. 2008;105(36):13656-13661.

19. Shim JC, Shin JG, Kelly DL, et al. Adjunctive treatment with a dopamine partial agonist, aripiprazole, for antipsychotic-induced hyperprolactinemia: a placebo-controlled trial. Am J Psychiatry. 2007;164(9):1404-1410.

20. Padala PR, Wengel SP, Petty F. Manic episode during treatment with aripiprazole. Am J Psychiatry. 2007;164(1):172-173.

21. Hu CH, Pai N, Huang XF, et al. Potential control of risperidone-related cognitive deficits by adjunctive aripiprazole treatment. J Clin Psychopharmacol. 2011;31(1):135-136;author reply 136–137.

22. Cohen J, Magalon D, Boyer L, et al. Aripiprazole-induced pathological gambling: a report of 3 cases. Curr Drug Saf. 2011;6(1):51-53.

23. Lea JW, Stoner SC, Lafollette J. Agitation associated with aripiprazole initiation. Pharmacotherapy. 2007;27(9):1339-1342.

24. Cho DY, Lindenmayer JP. Aripiprazole-induced agitation after clozapine discontinuation: a case report. J Clin Psychiatry. 2009;70(1):141-143.

25. Pae CU, Chiesa A, Mandelli L, et al. Predictors of early worsening after switch to aripiprazole: a randomized, controlled, open-label study. Clin Drug Investig. 2010;30(3):187-193.

26. Adan-Manes J, Garcia-Parajua P. Aripiprazole in combination with other antipsychotic drugs may worsen psychosis. J Clin Pharm Ther. 2009;34(2):245-246.

27. Cognata-Smith C, Baker RA, Pikalov A, et al. Analysis of nine aripiprazole trials to evaluate strategies for switching patients with schizophrenia to aripiprazole. Paper presented at: 162nd Annual Meeting American Psychiatric Association; May 16-21, 2009; San Francisco, CA.

28. Lieberman J. Cholinergic rebound in neuroleptic withdrawal syndromes. Psychosomatics. 1981;22(3):253-254.

29. Marder SR, McQuade RD, Stock E, et al. Aripiprazole in the treatment of schizophrenia: Safety and tolerability in short-term, placebo-controlled trials. Schizophr Res. 2003;61(2-3):123-136.

30. Kane JM, Fleischhacker WW, Hansen L, et al. Akathisia: an updated review focusing on second-generation antipsychotics. J Clin Psychiatry. 2009;70(5):627-643.

31. Kane JM, Barnes TR, Correll CU, et al. Evaluation of akathisia in patients with schizophrenia, schizoaffective disorder, or bipolar I disorder: A post hoc analysis of pooled data from short- and long-term aripiprazole trials. J Psychopharmacol. 2010;24(7):1019-1029.

32. Fleischhacker WW, McQuade RD, Marcus RN, et al. A double-blind, randomized comparative study of aripiprazole and olanzapine in patients with schizophrenia. Biol Psychiatry. 2009;65(6):510-517.

33. Kane JM, Osuntokun O, Kryzhanovskaya LA, et al. A 28-week, randomized, double-blind study of olanzapine versus aripiprazole in the treatment of schizophrenia. J Clin Psychiatry. 2009;70(4):572-581.

34. Kane JM, Correll CU, Goff DC, et al. A multicenter, randomized, double-blind, placebo-controlled, 16-week study of adjunctive aripiprazole for schizophrenia or schizoaffective disorder inadequately treated with quetiapine or risperidone monotherapy. J Clin Psychiatry. 2009;70(10):1348-1357.

References

 

1. Citrome L. A review of aripiprazole in the treatment of patients with schizophrenia or bipolar I disorder. Neuropsychiatr Dis Treat. 2006;2(4):427-443.

2. Chong SA, Tan CH, Lee HS. Worsening of psychosis with clozapine and selective serotonin reuptake inhibitor combination: two case reports. J Clin Psychopharmacol. 1997;17(1):68-69.

3. Bowers MB Jr, Swigar ME. Psychotic patients who become worse on neuroleptics. J Clin Psychopharmacol. 1988;8(6):417-421.

4. Tornatore FL, Lee D, Sramek JJ. Psychotic exacerbation with haloperidol. Drug Intell Clin Pharm. 1981;15(3):209-213.

5. Beaulieu JM, Gainetdinov RR. The physiology signaling, and pharmacology of dopamine receptors. Pharmacol Rev. 2011;63(1):182-217.

6. Grunder G, Carlsson A, Wong DF. Mechanism of new antipsychotic medications: occupancy is not just antagonism. Arch Gen Psychiatry. 2003;60(10):974-977.

7. Wood MD, Scott C, Clarke K, et al. Aripiprazole and its human metabolite are partial agonists at the human dopamine D2 receptor, but the rodent metabolite displays antagonist properties. Eur J Pharmacol. 2006;546(1-3):88-94.

8. Seeman P, Weinshenker D, Quirion R, et al. Dopamine supersensitivity correlates with D2High states, implying many paths to psychosis. Proc Natl Acad Sci U S A. 2005;102(9):3513-3518.

9. Seeman P, Ko F, Jack E, et al. Consistent with dopamine supersensitivity, RGS9 expression is diminished in the amphetamine-treated animal model of schizophrenia and in postmortem schizophrenia brain. Synapse. 2007;61(5):303-309.

10. Burt DR, Creese I, Snyder SH. Antischizophrenic drugs: chronic treatment elevates dopamine receptor binding in brain. Science. 1977;196(4287):326-328.

11. Silvestri S, Seeman MV, Negrete JC, et al. Increased dopamine D2 receptor binding after long-term treatment with antipsychotics in humans: a clinical PET study. Psychopharmacology (Berl). 2000;152(2):174-180.

12. Sayers AC, Bürki HR, Ruch W, et al. Neuroleptic-induced hypersensitivity of striatal dopamine receptors in the rat as a model of tardive dyskinesias. Effects of clozapine, haloperidol, loxapine and chlorpromazine. Psychopharmacologia. 1975;41(2):97-104.

13. Moncrieff J. Does antipsychotic withdrawal provoke psychosis? Review of the literature on rapid onset psychosis (supersensitivity psychosis) and withdrawal-related relapse. Acta Psychiatr Scand. 2006;114(1):3-13.

14. Takeuchi H, Uchida H, Suzuki T, et al. Predictors of clinical worsening after a switch to aripiprazole in patients with schizophrenia: a 1-year naturalistic follow-up study. J Clin Psychopharmacol. 2009;29(4):394-395.

15. Shapiro DA, Renock S, Arrington E, et al. Aripiprazole, a novel atypical antipsychotic drug with a unique and robust pharmacology. Neuropsychopharmacology. 2003;28(8):1400-1411.

16. Urban JD, Vargas GA, von Zastrow M, et al. Aripiprazole has functionally selective actions at dopamine D2 receptor-mediated signaling pathways. Neuropsychopharmacology. 2007;32(1):67-77.

17. Klewe IV, Nielsen SM, Tarpo L, et al. Recruitment of beta-arrestin2 to the dopamine D2 receptor: Insights into anti-psychotic and anti-parkinsonian drug receptor signaling. Neuropharmacology. 2008;54(8):1215-1222.

18. Masri B, Salahpour A, Didriksen M, et al. Antagonism of dopamine D2 receptor/beta-arrestin 2 interaction is a common property of clinically effective antipsychotics. Proc Natl Acad Sci U S A. 2008;105(36):13656-13661.

19. Shim JC, Shin JG, Kelly DL, et al. Adjunctive treatment with a dopamine partial agonist, aripiprazole, for antipsychotic-induced hyperprolactinemia: a placebo-controlled trial. Am J Psychiatry. 2007;164(9):1404-1410.

20. Padala PR, Wengel SP, Petty F. Manic episode during treatment with aripiprazole. Am J Psychiatry. 2007;164(1):172-173.

21. Hu CH, Pai N, Huang XF, et al. Potential control of risperidone-related cognitive deficits by adjunctive aripiprazole treatment. J Clin Psychopharmacol. 2011;31(1):135-136;author reply 136–137.

22. Cohen J, Magalon D, Boyer L, et al. Aripiprazole-induced pathological gambling: a report of 3 cases. Curr Drug Saf. 2011;6(1):51-53.

23. Lea JW, Stoner SC, Lafollette J. Agitation associated with aripiprazole initiation. Pharmacotherapy. 2007;27(9):1339-1342.

24. Cho DY, Lindenmayer JP. Aripiprazole-induced agitation after clozapine discontinuation: a case report. J Clin Psychiatry. 2009;70(1):141-143.

25. Pae CU, Chiesa A, Mandelli L, et al. Predictors of early worsening after switch to aripiprazole: a randomized, controlled, open-label study. Clin Drug Investig. 2010;30(3):187-193.

26. Adan-Manes J, Garcia-Parajua P. Aripiprazole in combination with other antipsychotic drugs may worsen psychosis. J Clin Pharm Ther. 2009;34(2):245-246.

27. Cognata-Smith C, Baker RA, Pikalov A, et al. Analysis of nine aripiprazole trials to evaluate strategies for switching patients with schizophrenia to aripiprazole. Paper presented at: 162nd Annual Meeting American Psychiatric Association; May 16-21, 2009; San Francisco, CA.

28. Lieberman J. Cholinergic rebound in neuroleptic withdrawal syndromes. Psychosomatics. 1981;22(3):253-254.

29. Marder SR, McQuade RD, Stock E, et al. Aripiprazole in the treatment of schizophrenia: Safety and tolerability in short-term, placebo-controlled trials. Schizophr Res. 2003;61(2-3):123-136.

30. Kane JM, Fleischhacker WW, Hansen L, et al. Akathisia: an updated review focusing on second-generation antipsychotics. J Clin Psychiatry. 2009;70(5):627-643.

31. Kane JM, Barnes TR, Correll CU, et al. Evaluation of akathisia in patients with schizophrenia, schizoaffective disorder, or bipolar I disorder: A post hoc analysis of pooled data from short- and long-term aripiprazole trials. J Psychopharmacol. 2010;24(7):1019-1029.

32. Fleischhacker WW, McQuade RD, Marcus RN, et al. A double-blind, randomized comparative study of aripiprazole and olanzapine in patients with schizophrenia. Biol Psychiatry. 2009;65(6):510-517.

33. Kane JM, Osuntokun O, Kryzhanovskaya LA, et al. A 28-week, randomized, double-blind study of olanzapine versus aripiprazole in the treatment of schizophrenia. J Clin Psychiatry. 2009;70(4):572-581.

34. Kane JM, Correll CU, Goff DC, et al. A multicenter, randomized, double-blind, placebo-controlled, 16-week study of adjunctive aripiprazole for schizophrenia or schizoaffective disorder inadequately treated with quetiapine or risperidone monotherapy. J Clin Psychiatry. 2009;70(10):1348-1357.

Issue
Current Psychiatry - 10(10)
Issue
Current Psychiatry - 10(10)
Page Number
53-60
Page Number
53-60
Publications
Publications
Topics
Article Type
Display Headline
Is there a link between aripiprazole and treatment-emergent psychosis?
Display Headline
Is there a link between aripiprazole and treatment-emergent psychosis?
Legacy Keywords
aripiprazole; treatment-emergent psychosis; James Gugger; Courtney Tam; Charles Ashby; Vicki Ellingrod; akathisia; D2 receptor; pharmacologic effects; worsening psychosis
Legacy Keywords
aripiprazole; treatment-emergent psychosis; James Gugger; Courtney Tam; Charles Ashby; Vicki Ellingrod; akathisia; D2 receptor; pharmacologic effects; worsening psychosis
Sections
Disallow All Ads
Alternative CME
Article PDF Media

How to use patient-centered language in documentation

Article Type
Changed
Tue, 12/11/2018 - 15:19
Display Headline
How to use patient-centered language in documentation

Discuss this article at www.facebook.com/CurrentPsychiatry

As psychiatric care transitions to using electronic medical records, providers should be aware that patients—and their legal representation— have increasing access to their medical records. Use of patient-centered, nonjudgmental language will better preserve the physician/patient alliance.

Consider the type of language you would find acceptable in documents describing the care provided to you or a loved one. Whenever possible, describe behavior by using objective and phenomenological terms. Nothing is sacrificed by replacing words that carry a negative connotation with less charged words. However, it is acceptable— and can add to the evaluation—to quote the patient’s own words.

The Table below lists alternative terms and phrases for use in psychiatric documentation.

Table

Nonjudgmental language for psychiatric documentation

Language with negative connotationPatient-centered language
PromiscuityImpulsive sexual behavior
Self-mutilationNonsuicidal self-injury
Manipulative, ‘gamey’The patient sought to meet their need of… (or describe specific behaviors)
Refused medicationDeclined or chose not to accept medication
Nasty, rude, meanThe patient used offensive language. The patient behaved in an aggressive manner by…
TriggerPrompt
DemandingMade repeated requests
NoncompliantDid not adhere to the treatment plan
Frantic, desperateUrgent, acute, demonstrated intense feelings of…
Disturbed, dysfunctionalDysregulated, difficult to manage
NeedySought reassurance
Failed medication trialTreatment with this medication was not associated with improvement
Shopping spreeImpulsive spending behavior
The patient complains of…The patient reported…
Drug bingeHeavy substance use over a short period

Disclosure

Dr. Nelson receives grant research/support from the Minnesota Medical Foundation.

Article PDF
Author and Disclosure Information

Katharine J. Nelson, MD
Dr. Nelson is Assistant Professor of Psychiatry and Medical Director of the Borderline Personality Disorder Program, Department of Psychiatry, University of Minnesota, Minneapolis, MN

Issue
Current Psychiatry - 10(10)
Publications
Topics
Page Number
70-70
Legacy Keywords
patient-centered language; documentation; electronic medical records; nonjudgmental language; objective terms; phenomenological terms
Sections
Author and Disclosure Information

Katharine J. Nelson, MD
Dr. Nelson is Assistant Professor of Psychiatry and Medical Director of the Borderline Personality Disorder Program, Department of Psychiatry, University of Minnesota, Minneapolis, MN

Author and Disclosure Information

Katharine J. Nelson, MD
Dr. Nelson is Assistant Professor of Psychiatry and Medical Director of the Borderline Personality Disorder Program, Department of Psychiatry, University of Minnesota, Minneapolis, MN

Article PDF
Article PDF

Discuss this article at www.facebook.com/CurrentPsychiatry

As psychiatric care transitions to using electronic medical records, providers should be aware that patients—and their legal representation— have increasing access to their medical records. Use of patient-centered, nonjudgmental language will better preserve the physician/patient alliance.

Consider the type of language you would find acceptable in documents describing the care provided to you or a loved one. Whenever possible, describe behavior by using objective and phenomenological terms. Nothing is sacrificed by replacing words that carry a negative connotation with less charged words. However, it is acceptable— and can add to the evaluation—to quote the patient’s own words.

The Table below lists alternative terms and phrases for use in psychiatric documentation.

Table

Nonjudgmental language for psychiatric documentation

Language with negative connotationPatient-centered language
PromiscuityImpulsive sexual behavior
Self-mutilationNonsuicidal self-injury
Manipulative, ‘gamey’The patient sought to meet their need of… (or describe specific behaviors)
Refused medicationDeclined or chose not to accept medication
Nasty, rude, meanThe patient used offensive language. The patient behaved in an aggressive manner by…
TriggerPrompt
DemandingMade repeated requests
NoncompliantDid not adhere to the treatment plan
Frantic, desperateUrgent, acute, demonstrated intense feelings of…
Disturbed, dysfunctionalDysregulated, difficult to manage
NeedySought reassurance
Failed medication trialTreatment with this medication was not associated with improvement
Shopping spreeImpulsive spending behavior
The patient complains of…The patient reported…
Drug bingeHeavy substance use over a short period

Disclosure

Dr. Nelson receives grant research/support from the Minnesota Medical Foundation.

Discuss this article at www.facebook.com/CurrentPsychiatry

As psychiatric care transitions to using electronic medical records, providers should be aware that patients—and their legal representation— have increasing access to their medical records. Use of patient-centered, nonjudgmental language will better preserve the physician/patient alliance.

Consider the type of language you would find acceptable in documents describing the care provided to you or a loved one. Whenever possible, describe behavior by using objective and phenomenological terms. Nothing is sacrificed by replacing words that carry a negative connotation with less charged words. However, it is acceptable— and can add to the evaluation—to quote the patient’s own words.

The Table below lists alternative terms and phrases for use in psychiatric documentation.

Table

Nonjudgmental language for psychiatric documentation

Language with negative connotationPatient-centered language
PromiscuityImpulsive sexual behavior
Self-mutilationNonsuicidal self-injury
Manipulative, ‘gamey’The patient sought to meet their need of… (or describe specific behaviors)
Refused medicationDeclined or chose not to accept medication
Nasty, rude, meanThe patient used offensive language. The patient behaved in an aggressive manner by…
TriggerPrompt
DemandingMade repeated requests
NoncompliantDid not adhere to the treatment plan
Frantic, desperateUrgent, acute, demonstrated intense feelings of…
Disturbed, dysfunctionalDysregulated, difficult to manage
NeedySought reassurance
Failed medication trialTreatment with this medication was not associated with improvement
Shopping spreeImpulsive spending behavior
The patient complains of…The patient reported…
Drug bingeHeavy substance use over a short period

Disclosure

Dr. Nelson receives grant research/support from the Minnesota Medical Foundation.

Issue
Current Psychiatry - 10(10)
Issue
Current Psychiatry - 10(10)
Page Number
70-70
Page Number
70-70
Publications
Publications
Topics
Article Type
Display Headline
How to use patient-centered language in documentation
Display Headline
How to use patient-centered language in documentation
Legacy Keywords
patient-centered language; documentation; electronic medical records; nonjudgmental language; objective terms; phenomenological terms
Legacy Keywords
patient-centered language; documentation; electronic medical records; nonjudgmental language; objective terms; phenomenological terms
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Reducing clozapine-induced hypersalivation

Article Type
Changed
Tue, 12/11/2018 - 15:19
Display Headline
Reducing clozapine-induced hypersalivation

Discuss this article at www.facebook.com/CurrentPsychiatry

Hypersalivation is a well-documented side effect of clozapine that may affect nearly 30% of patients who take this medication.1 Because clozapine has anticholinergic properties that are expected to reduce secretions, hypersalivation is considered a paradoxical effect. This paradox can be explained by examining clozapine’s actions at the molecular level.

Clozapine has antagonistic activity at many types of receptors, including D1 to D5 dopaminergic, α1 and α2 adrenergic, serotonergic, histaminergic, and M1, M2, M3, and M5 cholinergic.2 By contrast, clozapine’s activity at M4 cholinergic receptors is agonistic. The combination of clozapine’s antagonist activity at α2 adrenergic receptors and agonist activity at muscarinic M4 receptors results in hypersalivation.3 Clozapine also increases pooling of saliva in the mouth by diminishing esophageal motility, which gives the appearance of hypersalivation as a result of reduced saliva clearance through normal swallowing.4

Practical lifestyle changes

Excessive saliva secretion and pooling can be reduced by practical behavioral and/or pharmacologic interventions. Patients who chew gum during the day will increase their swallowing unconsciously5; recommend sugarless gum to help avoid tooth decay and gum disease. If hypersalivation is particularly bothersome at night, patients may find it useful to cover their pillows with an absorbent towel.

Pharmacologic treatments

Pharmacologic interventions rely on counteracting clozapine’s secretion-inducing effects by opposing M4 agonism, α2 antagonism, or both. Antimuscarinic medications such as benztropine, trihexyphenidyl, amitriptyline, or pirenzepine often are used to reduce hypersalivation,6 although 1 systematic review concluded further evidence is needed to support the effectiveness of this approach.7 The α2 antagonism can be opposed by using the α2 agonist clonidine, which can be administered as a weekly transdermal patch of 0.1 to 0.2 mg.8 In 1 retrospective study, the cholinergic and adrenergic mechanisms of hypersalivation were addressed by combining benztropine (2 mg/d in a divided dose) with the α1 antagonist terazosin (2 mg/d). This combination reduced hypersalivation significantly in all patients after 12 weeks, exceeding the efficacy of either benztropine or terazosin administered alone.9

Hypersalivation is an inconvenient, potentially embarrassing aspect of clozapine treatment that can cause avoidable distress. One or more of these suggestions may help control an adverse effect that could diminish patient satisfaction and undermine treatment compliance.

Disclosure

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

References

1. Safferman A, Lieberman JA, Kane JM, et al. Update on the clinical efficacy and side effects of clozapine. Schizophr Bull. 1991;17(2):247-261.

2. Zorn SH, Jones SB, Ward KM, et al. Clozapine is a potent and selective muscarinic M4 receptor agonist. Eur J Pharmacol. 1994;269(3):R1-R2.

3. Berlan M, Montastruc JL, Lafontan M. Pharmacological prospects for alpha 2-adrenoceptor antagonist therapy. Trends Pharmacol Sci. 1992;13(7):277-282.

4. Sadock BJ, Sadock VA. Kaplan and Sadock’s comprehensive textbook of psychiatry. 7th ed. Philadelphia PA: Lippincott Williams & Wilkins; 2000.

5. Bourgeois JA, Drexler KG, Hall MJ. Hypersalivation and clozapine. Hosp Community Psychiatry. 1991;42(11):1174.-

6. Cree A, Mir S, Fahy T. A review of the treatment options for clozapine-induced hypersalivation. The Psychiatrist. 2001;25:114-116.

7. Syed R, Au K, Cahill C, et al. Pharmacological interventions for clozapine-induced hypersalivation. Cochrane Database Syst Rev. 2008;(3):CD005579.-

8. Grabowski J. Clonidine treatment of clozapine-induced hypersalivation. J Clin Psychopharmacol. 1992;12(1):69-70.

9. Reinstein MJ, Sirotovskaya LA, Chasanov MA, et al. Comparative efficacy and tolerability of benztropine and terazosin in the treatment of hypersalivation secondary to clozapine. Clin Drug Investig. 1999;17(2):97-102.

Article PDF
Author and Disclosure Information

Gurprit Lamba, MD
Dr. Lamba is a Geriatric Psychiatry Fellow, Partners HealthCare Geriatric Psychiatry Fellowship, Boston, MA
James M. Ellison, MD, MPH
Dr. Ellison is Associate Professor of Psychiatry, Harvard Medical School, Boston, MA

Issue
Current Psychiatry - 10(10)
Publications
Topics
Page Number
77-78
Legacy Keywords
clozapine; hypersalivation; Pearls; anticholinergic; lifestyle changes; pharmacologic treatments; benztropine; trihexyphenidyl; amitriptyline; pirenzepine; Gurprit Lamba; James Ellison
Sections
Author and Disclosure Information

Gurprit Lamba, MD
Dr. Lamba is a Geriatric Psychiatry Fellow, Partners HealthCare Geriatric Psychiatry Fellowship, Boston, MA
James M. Ellison, MD, MPH
Dr. Ellison is Associate Professor of Psychiatry, Harvard Medical School, Boston, MA

Author and Disclosure Information

Gurprit Lamba, MD
Dr. Lamba is a Geriatric Psychiatry Fellow, Partners HealthCare Geriatric Psychiatry Fellowship, Boston, MA
James M. Ellison, MD, MPH
Dr. Ellison is Associate Professor of Psychiatry, Harvard Medical School, Boston, MA

Article PDF
Article PDF

Discuss this article at www.facebook.com/CurrentPsychiatry

Hypersalivation is a well-documented side effect of clozapine that may affect nearly 30% of patients who take this medication.1 Because clozapine has anticholinergic properties that are expected to reduce secretions, hypersalivation is considered a paradoxical effect. This paradox can be explained by examining clozapine’s actions at the molecular level.

Clozapine has antagonistic activity at many types of receptors, including D1 to D5 dopaminergic, α1 and α2 adrenergic, serotonergic, histaminergic, and M1, M2, M3, and M5 cholinergic.2 By contrast, clozapine’s activity at M4 cholinergic receptors is agonistic. The combination of clozapine’s antagonist activity at α2 adrenergic receptors and agonist activity at muscarinic M4 receptors results in hypersalivation.3 Clozapine also increases pooling of saliva in the mouth by diminishing esophageal motility, which gives the appearance of hypersalivation as a result of reduced saliva clearance through normal swallowing.4

Practical lifestyle changes

Excessive saliva secretion and pooling can be reduced by practical behavioral and/or pharmacologic interventions. Patients who chew gum during the day will increase their swallowing unconsciously5; recommend sugarless gum to help avoid tooth decay and gum disease. If hypersalivation is particularly bothersome at night, patients may find it useful to cover their pillows with an absorbent towel.

Pharmacologic treatments

Pharmacologic interventions rely on counteracting clozapine’s secretion-inducing effects by opposing M4 agonism, α2 antagonism, or both. Antimuscarinic medications such as benztropine, trihexyphenidyl, amitriptyline, or pirenzepine often are used to reduce hypersalivation,6 although 1 systematic review concluded further evidence is needed to support the effectiveness of this approach.7 The α2 antagonism can be opposed by using the α2 agonist clonidine, which can be administered as a weekly transdermal patch of 0.1 to 0.2 mg.8 In 1 retrospective study, the cholinergic and adrenergic mechanisms of hypersalivation were addressed by combining benztropine (2 mg/d in a divided dose) with the α1 antagonist terazosin (2 mg/d). This combination reduced hypersalivation significantly in all patients after 12 weeks, exceeding the efficacy of either benztropine or terazosin administered alone.9

Hypersalivation is an inconvenient, potentially embarrassing aspect of clozapine treatment that can cause avoidable distress. One or more of these suggestions may help control an adverse effect that could diminish patient satisfaction and undermine treatment compliance.

Disclosure

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

Discuss this article at www.facebook.com/CurrentPsychiatry

Hypersalivation is a well-documented side effect of clozapine that may affect nearly 30% of patients who take this medication.1 Because clozapine has anticholinergic properties that are expected to reduce secretions, hypersalivation is considered a paradoxical effect. This paradox can be explained by examining clozapine’s actions at the molecular level.

Clozapine has antagonistic activity at many types of receptors, including D1 to D5 dopaminergic, α1 and α2 adrenergic, serotonergic, histaminergic, and M1, M2, M3, and M5 cholinergic.2 By contrast, clozapine’s activity at M4 cholinergic receptors is agonistic. The combination of clozapine’s antagonist activity at α2 adrenergic receptors and agonist activity at muscarinic M4 receptors results in hypersalivation.3 Clozapine also increases pooling of saliva in the mouth by diminishing esophageal motility, which gives the appearance of hypersalivation as a result of reduced saliva clearance through normal swallowing.4

Practical lifestyle changes

Excessive saliva secretion and pooling can be reduced by practical behavioral and/or pharmacologic interventions. Patients who chew gum during the day will increase their swallowing unconsciously5; recommend sugarless gum to help avoid tooth decay and gum disease. If hypersalivation is particularly bothersome at night, patients may find it useful to cover their pillows with an absorbent towel.

Pharmacologic treatments

Pharmacologic interventions rely on counteracting clozapine’s secretion-inducing effects by opposing M4 agonism, α2 antagonism, or both. Antimuscarinic medications such as benztropine, trihexyphenidyl, amitriptyline, or pirenzepine often are used to reduce hypersalivation,6 although 1 systematic review concluded further evidence is needed to support the effectiveness of this approach.7 The α2 antagonism can be opposed by using the α2 agonist clonidine, which can be administered as a weekly transdermal patch of 0.1 to 0.2 mg.8 In 1 retrospective study, the cholinergic and adrenergic mechanisms of hypersalivation were addressed by combining benztropine (2 mg/d in a divided dose) with the α1 antagonist terazosin (2 mg/d). This combination reduced hypersalivation significantly in all patients after 12 weeks, exceeding the efficacy of either benztropine or terazosin administered alone.9

Hypersalivation is an inconvenient, potentially embarrassing aspect of clozapine treatment that can cause avoidable distress. One or more of these suggestions may help control an adverse effect that could diminish patient satisfaction and undermine treatment compliance.

Disclosure

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

References

1. Safferman A, Lieberman JA, Kane JM, et al. Update on the clinical efficacy and side effects of clozapine. Schizophr Bull. 1991;17(2):247-261.

2. Zorn SH, Jones SB, Ward KM, et al. Clozapine is a potent and selective muscarinic M4 receptor agonist. Eur J Pharmacol. 1994;269(3):R1-R2.

3. Berlan M, Montastruc JL, Lafontan M. Pharmacological prospects for alpha 2-adrenoceptor antagonist therapy. Trends Pharmacol Sci. 1992;13(7):277-282.

4. Sadock BJ, Sadock VA. Kaplan and Sadock’s comprehensive textbook of psychiatry. 7th ed. Philadelphia PA: Lippincott Williams & Wilkins; 2000.

5. Bourgeois JA, Drexler KG, Hall MJ. Hypersalivation and clozapine. Hosp Community Psychiatry. 1991;42(11):1174.-

6. Cree A, Mir S, Fahy T. A review of the treatment options for clozapine-induced hypersalivation. The Psychiatrist. 2001;25:114-116.

7. Syed R, Au K, Cahill C, et al. Pharmacological interventions for clozapine-induced hypersalivation. Cochrane Database Syst Rev. 2008;(3):CD005579.-

8. Grabowski J. Clonidine treatment of clozapine-induced hypersalivation. J Clin Psychopharmacol. 1992;12(1):69-70.

9. Reinstein MJ, Sirotovskaya LA, Chasanov MA, et al. Comparative efficacy and tolerability of benztropine and terazosin in the treatment of hypersalivation secondary to clozapine. Clin Drug Investig. 1999;17(2):97-102.

References

1. Safferman A, Lieberman JA, Kane JM, et al. Update on the clinical efficacy and side effects of clozapine. Schizophr Bull. 1991;17(2):247-261.

2. Zorn SH, Jones SB, Ward KM, et al. Clozapine is a potent and selective muscarinic M4 receptor agonist. Eur J Pharmacol. 1994;269(3):R1-R2.

3. Berlan M, Montastruc JL, Lafontan M. Pharmacological prospects for alpha 2-adrenoceptor antagonist therapy. Trends Pharmacol Sci. 1992;13(7):277-282.

4. Sadock BJ, Sadock VA. Kaplan and Sadock’s comprehensive textbook of psychiatry. 7th ed. Philadelphia PA: Lippincott Williams & Wilkins; 2000.

5. Bourgeois JA, Drexler KG, Hall MJ. Hypersalivation and clozapine. Hosp Community Psychiatry. 1991;42(11):1174.-

6. Cree A, Mir S, Fahy T. A review of the treatment options for clozapine-induced hypersalivation. The Psychiatrist. 2001;25:114-116.

7. Syed R, Au K, Cahill C, et al. Pharmacological interventions for clozapine-induced hypersalivation. Cochrane Database Syst Rev. 2008;(3):CD005579.-

8. Grabowski J. Clonidine treatment of clozapine-induced hypersalivation. J Clin Psychopharmacol. 1992;12(1):69-70.

9. Reinstein MJ, Sirotovskaya LA, Chasanov MA, et al. Comparative efficacy and tolerability of benztropine and terazosin in the treatment of hypersalivation secondary to clozapine. Clin Drug Investig. 1999;17(2):97-102.

Issue
Current Psychiatry - 10(10)
Issue
Current Psychiatry - 10(10)
Page Number
77-78
Page Number
77-78
Publications
Publications
Topics
Article Type
Display Headline
Reducing clozapine-induced hypersalivation
Display Headline
Reducing clozapine-induced hypersalivation
Legacy Keywords
clozapine; hypersalivation; Pearls; anticholinergic; lifestyle changes; pharmacologic treatments; benztropine; trihexyphenidyl; amitriptyline; pirenzepine; Gurprit Lamba; James Ellison
Legacy Keywords
clozapine; hypersalivation; Pearls; anticholinergic; lifestyle changes; pharmacologic treatments; benztropine; trihexyphenidyl; amitriptyline; pirenzepine; Gurprit Lamba; James Ellison
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Delirium and benzodiazepines

Article Type
Changed
Mon, 04/16/2018 - 14:26
Display Headline
Delirium and benzodiazepines

I read with concern the letter Steven Lippmann, MD, wrote (“Treating delirium,” Comments and Controversies, Current Psychiatry, July 2011, p. 53) in response to “Atypical antipsychotics for delirium: A reasonable alternative to haloperidol?” (Current Psychiatry, January 2011, p. 37-46). Although the original article acknowledged that haloperidol has been the “gold standard” in treating agitation during delirium, the authors wrote a review of evidence-based literature, summarizing the emerging literature on the use of other antipsychotics. I agree with Dr. Lippmann that diagnosing and treating the underlying cause of delirium should be the primary focus, but I strongly disagree that benzodiazepines are safer than antipsychotics in managing behavioral aspects of delirium.

The January 2011 article gives a good summary of the literature on using antipsychotics in delirium. I would like to focus on Dr. Lippmann’s assertion that benzodiazepines are safer than antipsychotics. No study has found evidence for benzodiazepines’ effectiveness in non-alcohol–related delirium.1 However, a number of studies have found benzodiazepines are an independent risk factor for delirium.

A randomized controlled trial comparing a benzodiazepine (lorazepam) with antipsychotics (haloperidol, chlorpromazine) to treat patients with delirium had to be interrupted prematurely.2 This study found improvement in delirium symptoms (measured by the Delirium Rating Scale) with both haloperidol and chlorpromazine. The researchers decided to discontinue the lorazepam treatment arm because of unacceptable adverse effects among lorazepam-treated participants, including excessive sedation, ataxia, disinhibition, and worsened confusion.

A study of surgical patients found a significant association between delirium and postoperative exposure to benzodiazepines.3 A study of delirium risk factors in an intensive care unit found lorazepam to be a consistent and significant predictor of delirium.4 Two other studies have found benzodiazepines can cause or worsen delirium.5,6 In conclusion, there is no evidence for use of benzodiazepines to manage behavioral symptoms in delirium. Benzodiazepines alone are not safer than antipsychotics and are not indicated for treating non-alcohol–related delirium.

Rashi Aggarwal, MD
Assistant Professor
Department of Psychiatry
New Jersey Medical School, UMDNJ
Newark, NJ

References

Reference

1. Lonergan E, Luxenberg J, Areosa Sastre A. Benzodiazepines for delirium. Cochrane Database Syst Rev. 2009;(4):CD006379.-

2. Breitbart W, Marotta R, Platt MM, et al. A double-blind trial of haloperidol, chlorpromazine, and lorazepam in the treatment of delirium in hospitalized AIDS patients. Am J Psychiatry. 1996;153(2):231-237.

3. Marcantonio ER, Juarez G, Goldman L, et al. The relationship of postoperative delirium with psychoactive medications. JAMA. 1994;272(19):1518-1522.

4. Pandharipande P, Shintani A, Peterson J, et al. Lorazepam is an independent risk factor for transitioning to delirium in intensive care unit patients. Anesthesiology. 2006;104(1):21-26.

5. Pandharipande PP, Pun BT, Herr DL, et al. Effect of sedation with dexmedetomidine vs lorazepam on acute brain dysfunction in mechanically ventilated patients: the MENDS randomized controlled trial. JAMA. 2007;298(22):2644-2653.

6. Dubois MJ, Bergeron N, Dumont M, et al. Delirium in an intensive care unit: a study of risk factors. Intensive Care Med. 2001;27(8):1297-1304.

Article PDF
Author and Disclosure Information

Issue
Current Psychiatry - 10(10)
Publications
Topics
Page Number
4-4
Legacy Keywords
Comments & controversies; delirium; benzodiazepines; Steven Lippmann; atypical antipsychotics; haloperidol; Rashi Aggarwal
Sections
Author and Disclosure Information

Author and Disclosure Information

Article PDF
Article PDF

I read with concern the letter Steven Lippmann, MD, wrote (“Treating delirium,” Comments and Controversies, Current Psychiatry, July 2011, p. 53) in response to “Atypical antipsychotics for delirium: A reasonable alternative to haloperidol?” (Current Psychiatry, January 2011, p. 37-46). Although the original article acknowledged that haloperidol has been the “gold standard” in treating agitation during delirium, the authors wrote a review of evidence-based literature, summarizing the emerging literature on the use of other antipsychotics. I agree with Dr. Lippmann that diagnosing and treating the underlying cause of delirium should be the primary focus, but I strongly disagree that benzodiazepines are safer than antipsychotics in managing behavioral aspects of delirium.

The January 2011 article gives a good summary of the literature on using antipsychotics in delirium. I would like to focus on Dr. Lippmann’s assertion that benzodiazepines are safer than antipsychotics. No study has found evidence for benzodiazepines’ effectiveness in non-alcohol–related delirium.1 However, a number of studies have found benzodiazepines are an independent risk factor for delirium.

A randomized controlled trial comparing a benzodiazepine (lorazepam) with antipsychotics (haloperidol, chlorpromazine) to treat patients with delirium had to be interrupted prematurely.2 This study found improvement in delirium symptoms (measured by the Delirium Rating Scale) with both haloperidol and chlorpromazine. The researchers decided to discontinue the lorazepam treatment arm because of unacceptable adverse effects among lorazepam-treated participants, including excessive sedation, ataxia, disinhibition, and worsened confusion.

A study of surgical patients found a significant association between delirium and postoperative exposure to benzodiazepines.3 A study of delirium risk factors in an intensive care unit found lorazepam to be a consistent and significant predictor of delirium.4 Two other studies have found benzodiazepines can cause or worsen delirium.5,6 In conclusion, there is no evidence for use of benzodiazepines to manage behavioral symptoms in delirium. Benzodiazepines alone are not safer than antipsychotics and are not indicated for treating non-alcohol–related delirium.

Rashi Aggarwal, MD
Assistant Professor
Department of Psychiatry
New Jersey Medical School, UMDNJ
Newark, NJ

I read with concern the letter Steven Lippmann, MD, wrote (“Treating delirium,” Comments and Controversies, Current Psychiatry, July 2011, p. 53) in response to “Atypical antipsychotics for delirium: A reasonable alternative to haloperidol?” (Current Psychiatry, January 2011, p. 37-46). Although the original article acknowledged that haloperidol has been the “gold standard” in treating agitation during delirium, the authors wrote a review of evidence-based literature, summarizing the emerging literature on the use of other antipsychotics. I agree with Dr. Lippmann that diagnosing and treating the underlying cause of delirium should be the primary focus, but I strongly disagree that benzodiazepines are safer than antipsychotics in managing behavioral aspects of delirium.

The January 2011 article gives a good summary of the literature on using antipsychotics in delirium. I would like to focus on Dr. Lippmann’s assertion that benzodiazepines are safer than antipsychotics. No study has found evidence for benzodiazepines’ effectiveness in non-alcohol–related delirium.1 However, a number of studies have found benzodiazepines are an independent risk factor for delirium.

A randomized controlled trial comparing a benzodiazepine (lorazepam) with antipsychotics (haloperidol, chlorpromazine) to treat patients with delirium had to be interrupted prematurely.2 This study found improvement in delirium symptoms (measured by the Delirium Rating Scale) with both haloperidol and chlorpromazine. The researchers decided to discontinue the lorazepam treatment arm because of unacceptable adverse effects among lorazepam-treated participants, including excessive sedation, ataxia, disinhibition, and worsened confusion.

A study of surgical patients found a significant association between delirium and postoperative exposure to benzodiazepines.3 A study of delirium risk factors in an intensive care unit found lorazepam to be a consistent and significant predictor of delirium.4 Two other studies have found benzodiazepines can cause or worsen delirium.5,6 In conclusion, there is no evidence for use of benzodiazepines to manage behavioral symptoms in delirium. Benzodiazepines alone are not safer than antipsychotics and are not indicated for treating non-alcohol–related delirium.

Rashi Aggarwal, MD
Assistant Professor
Department of Psychiatry
New Jersey Medical School, UMDNJ
Newark, NJ

References

Reference

1. Lonergan E, Luxenberg J, Areosa Sastre A. Benzodiazepines for delirium. Cochrane Database Syst Rev. 2009;(4):CD006379.-

2. Breitbart W, Marotta R, Platt MM, et al. A double-blind trial of haloperidol, chlorpromazine, and lorazepam in the treatment of delirium in hospitalized AIDS patients. Am J Psychiatry. 1996;153(2):231-237.

3. Marcantonio ER, Juarez G, Goldman L, et al. The relationship of postoperative delirium with psychoactive medications. JAMA. 1994;272(19):1518-1522.

4. Pandharipande P, Shintani A, Peterson J, et al. Lorazepam is an independent risk factor for transitioning to delirium in intensive care unit patients. Anesthesiology. 2006;104(1):21-26.

5. Pandharipande PP, Pun BT, Herr DL, et al. Effect of sedation with dexmedetomidine vs lorazepam on acute brain dysfunction in mechanically ventilated patients: the MENDS randomized controlled trial. JAMA. 2007;298(22):2644-2653.

6. Dubois MJ, Bergeron N, Dumont M, et al. Delirium in an intensive care unit: a study of risk factors. Intensive Care Med. 2001;27(8):1297-1304.

References

Reference

1. Lonergan E, Luxenberg J, Areosa Sastre A. Benzodiazepines for delirium. Cochrane Database Syst Rev. 2009;(4):CD006379.-

2. Breitbart W, Marotta R, Platt MM, et al. A double-blind trial of haloperidol, chlorpromazine, and lorazepam in the treatment of delirium in hospitalized AIDS patients. Am J Psychiatry. 1996;153(2):231-237.

3. Marcantonio ER, Juarez G, Goldman L, et al. The relationship of postoperative delirium with psychoactive medications. JAMA. 1994;272(19):1518-1522.

4. Pandharipande P, Shintani A, Peterson J, et al. Lorazepam is an independent risk factor for transitioning to delirium in intensive care unit patients. Anesthesiology. 2006;104(1):21-26.

5. Pandharipande PP, Pun BT, Herr DL, et al. Effect of sedation with dexmedetomidine vs lorazepam on acute brain dysfunction in mechanically ventilated patients: the MENDS randomized controlled trial. JAMA. 2007;298(22):2644-2653.

6. Dubois MJ, Bergeron N, Dumont M, et al. Delirium in an intensive care unit: a study of risk factors. Intensive Care Med. 2001;27(8):1297-1304.

Issue
Current Psychiatry - 10(10)
Issue
Current Psychiatry - 10(10)
Page Number
4-4
Page Number
4-4
Publications
Publications
Topics
Article Type
Display Headline
Delirium and benzodiazepines
Display Headline
Delirium and benzodiazepines
Legacy Keywords
Comments & controversies; delirium; benzodiazepines; Steven Lippmann; atypical antipsychotics; haloperidol; Rashi Aggarwal
Legacy Keywords
Comments & controversies; delirium; benzodiazepines; Steven Lippmann; atypical antipsychotics; haloperidol; Rashi Aggarwal
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media