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

COPD comorbid with mental illness: What psychiatrists can do

Article Type
Changed
Mon, 04/16/2018 - 14:37
Display Headline
COPD comorbid with mental illness: What psychiatrists can do

Chronic obstructive pulmonary disease (COPD) usually is not diagnosed until clinically apparent and moderately advanced. Patients might not notice chronic dyspnea and smoker’s cough, or might consider their symptoms “normal” and not seek medical care. Delayed diagnosis is particularly prevalent in the psychiatric population, in which co-existing medical problems tend to remain unrecognized and untreated.1

Life expectancy of people with serious mental illness (SMI) is 13 to 30 years less than that of the general population—a gap that has widened over time.2 Pulmonary disease is a leading cause of elevated mortality risk in SMI, along with cardiovascular and infectious disease, diabetes, and barriers to care. Having a comorbid mental illness triples the mortality risk of chronic lower respiratory disease (Table 1).3


This article describes how you can intervene and improve quality of life for your patients with COPD by:

 

  • asking all patients, especially smokers, if they are experiencing classic symptoms of COPD
  • advocating for and supporting smoking cessation efforts
  • avoiding drug interactions and off-target dosing related to COPD and nicotine replacement therapy
  • considering, if feasible, a switch from typical to atypical antipsychotic therapy, which could reduce smoking behavior.


What is COPD?
COPD is preventable and treatable. It is characterized by “persistent airflow limitation that is usually progressive and associated with an enhanced chronic inflammatory response in the airways and the lungs to inhaled noxious particles or gases.”4

Smoking tobacco is the greatest risk factor for developing COPD.5 An estimated 50% to 80% of people with schizophrenia are smokers, as are 55% of people with bipolar disorder.6 COPD is a leading cause of morbidity and mortality worldwide,7,8 and its prevalence is projected to increase as the global population and smoking rates grow.9

A simplified schema of the pathophysiology of COPD implicates 4 lung areas: parenchyma, pulmonary vasculature, central airways, and peripheral airways.10 Variation in the areas affected and severity of change contributes to the disease’s heterogeneous presentation, which can include pulmonary hypertension, hypersecretion of mucus, ciliary dysfunction, airway hyperinflation, and impaired gas exchange.11,12 Many of these features lead to systemic effects as well, particularly on cardiac function.


When to test a patient for COPD
Early diagnosis and treatment can substantially improve quality-of-life outcomes for patients with COPD. The clinical approach (Figure 1) begins with recognizing classic symptoms. Consider COPD in any patient with:

 

  • dyspnea (particularly if becoming worse, persistent, or associated with exercise)
  • chronic cough
  • chronic sputum production
  • history of risk-factor exposure (particularly tobacco smoke)
  • family history of COPD.4


If the history and physical exam suggest COPD (Table 2), spirometry is the most reliable test to quantify and characterize lung dysfunction. It is not indicated as a screening tool for healthy adults or appropriate when a patient is acutely ill. Forced expiratory volume in the first second of expiration divided by the measured forced vital capacity (FEV1/FVC) < 0.7 defines clinical COPD and determines the need for pharmacologic intervention. Laboratory studies could be useful in certain clinical scenarios, such as serum testing for alpha1-antitrypsin deficiency in patients age <45 with emphysema. Plain film imaging might be useful to support a COPD diagnosis or rule out alternate diagnoses.


Psychopharmacology issues with comorbid COPD
Pharmacotherapy for psychiatric disorders can exacerbate comorbid COPD. For example, long-term use of phenothiazine-related typical antipsychotics for schizophrenia has been linked to an increased incidence of COPD.13 Antipsychotic side effects such as acute laryngeal dystonia and tardive dyskinesia, most commonly seen with first-generation antipsychotic use, can aggravate dyspnea caused by COPD. Opioids and most hypnotics, sedatives, and anxiolytics suppress the respiratory drive, and therefore should be used with caution in patients with COPD.

Carefully monitor serum levels of medications before and during attempts at smoking cessation. Nicotine’s induction of the cytochrome P450 1A2 system increases the metabolism of antipsychotics such as clozapine, fluvoxamine, olanzapine, and haloperidol. As a result, potentially toxic drug levels can occur when a smoker tries to quit.14

Screen patients with COPD for co­morbid psychiatric conditions. New psychiatric symptoms can emerge after COPD has been diagnosed, even in patients without pre-existing psychopathology.

Anxiety is a particularly common COPD comorbidity that can be difficult to manage. Selective serotonin reuptake inhibitors, buspirone, cognitive-behavioral therapy, and pulmonary rehabilitation can be helpful, although the effect of antidepressants on respiration is controversial. Nortriptyline has been shown to be effective in treating both anxiety and depressive symptoms in patients with COPD.15 Avoid using hypnotics to manage sleep problems related to COPD; instead, focus on minimizing sleep disturbance by limiting cough and dyspnea.


Antipsychotics and nicotine metabolism
Multiple studies have focused on the interplay among nicotine, dopamine, and antipsychotic agents. Nicotine receptors are present in the ventral tegmental dopaminergic cell bodies, which induce the release of dopamine and other neurotransmitters when stimulated. Smoking has been noted to increase in patients administered haloperidol (a dopamine antagonist) and to decrease with administration of bromocriptine (a dopamine agonist).16 This suggests that psychiatric patients might smoke to overcome the dopamine blockade caused by most typical antipsychotics, therefore alleviating their negative and extrapyramidal side effects.17

 

 

Alternatively, some studies suggest that a difference in dopamine receptor occupancy between typical and atypical antipsychotics leads to different effects on smoking behavior.18 When used long term, typical antipsychotics might increase dopamine receptors or dopamine sensitivity, and thus reinforce the positive effect of nicotine by increasing the number of receptors that can be stimulated, whereas atypical antipsychotics help stimulate the release of dopamine directly through partial agonist of serotonin 5-HT1A receptors.19,20 Atypical antipsychotics also appear to decrease cue-elicited cravings in people who are not mentally ill, whereas haloperidol does not.21

Based on these findings, switching patients with COPD from a typical to an atypical antipsychotic, if feasible, might make smoking cessation more manageable.22 Multiple studies have shown that clozapine is the preferred atypical antipsychotic because it is associated with the most significant decrease in smoking behaviors.23


First-line therapy: Nicotine replacement
Smoking cessation slows the progression of COPD and leads to marked improvements in cough, expectoration, breathlessness, and wheezing.24,25 Nicotine replacement therapy (NRT)—gum, inhaler, lozenges, nasal spray, and skin patch—is considered first-line pharmacotherapy. These nicotine substitutes can decrease withdrawal symptoms, although they do not appear to be as effective for light smokers (eg, <10 cigarettes/d), compared with heavy smokers (eg, ≥20 cigarettes/d).26

Long-term smoking abstinence can be improved with combination therapies. A nicotine patch, kept in place for as long as 24 hours, often is used with a nicotine gum or nasal spray. Another option combines the patch with a first-line, non-NRT intervention, such as sustained-release bupropion. Use bupropion with caution in psychiatric patients, however. Do not combine it with a monoamine oxidase inhibitor, and do not prescribe it to patients with an eating disorder or history of seizures.26 Bupropion could induce mania in patients with bipolar disorder.

Varenicline, a nicotinic receptor partial agonist indicated to aid in smoking cessation, has been shown to reduce pleasure gained from tobacco as well as cravings. It can increase the likelihood of abstinence from smoking for as long as 1 year, but it also can provoke behavioral changes, depressed mood, and suicidal ideation. These risks—described in an FDA black-box warning of serious neuropsychiatric events—warrant due caution when prescribing varenicline to patients with depression. The FDA also has warned that varenicline could lead to decreased alcohol tolerance and atypically aggressive behavior during intoxication, which is of particular concern because of the high rate of alcohol use among people with SMI.


Motivating and supporting change
When counseling patients with mental illness about smoking cessation, consider unique motivations that, if disregarded, could undermine your efforts. As described above, smoking can ameliorate negative and extrapyramidal symptoms associated with typical antipsychotics. This could explain the significantly higher rates of smoking associated with typical antipsychotics, compared with atypical antipsychotics.27 Patients also could use smoking as self-medication for depression and anxiety. Therefore, take care to offer alternate methods for coping, along with smoking cessation recommendations.22

Screen all adult patients for tobacco use, and offer prompt cessation counseling and pharmacologic interventions.28As a motivational intervention, the “5 As” framework—ask, advise, assess, assist, arrange—can help gauge patients’ smoking status and willingness to quit, as well as emphasize the importance of establishing a concrete, manageable plan.29

Keep in mind the barriers all patients face in their fight to quit smoking, such as nicotine withdrawal, weight gain, and loss of a coping mechanism for stress.29 Patients with schizophrenia can be motivated to quit smoking and participate in treatment for nicotine dependence.30

Besides encouraging smoking cessation, you can educate patients in behaviors that will improve COPD symptoms and management. These include:

 

  • reducing the risk of lung infections through vaccinations (influenza yearly, pneumonia once in adulthood) and avoiding crowds during peak cold and influenza season
  • participating in physical activity, which could slow lung function decline
  • adhering to prescribed medication
  • eating a balanced diet
  • seeking medical care early during an exacerbation.


Coaching patients in symptom control
Smoking cessation may have the greatest long-term benefit for patients with COPD, but symptom management is important as well (Figure 2). Pharmacotherapy for COPD usually is advanced in steps, but a more aggressive approach may be necessary for patients presenting with severe symptoms.


Mainstays of COPD therapy are inhaled bronchodilators, consisting of β2 agonists and anticholinergics, alone or in combination. Short-acting formulations are used for mild and intermittent symptoms; long-acting bronchodilators are added if symptoms persist.4 When dyspnea, wheezing, and activity intolerance are not well-controlled with bronchodilators, an inhaled corticosteroid can be tried, either alone or in combination with a long-acting bronchodilator.4

Adherence to medical recommendations is critical for successful COPD management, but inhaled therapy can be difficult for psychiatric patients—especially patients with cognitive or functional impairment. Asking them to demonstrate their inhaler technique can help assess treatment effectiveness.31

 

 

Referral to a pulmonologist is strongly advised in cases of:

 

  • advanced, end-stage COPD (FEV1 <50% predicted value despite adherence to recommended treatment, or rapid decline of FEV1)
  • COPD in patients age <50
  • frequent exacerbations
  • possible complications related to chronic heart failure
  • indications for oxygen treatment (eg, resting or ambulatory oxygen saturation ≤88% or PaO2 ≤55 mm Hg).32

 


Bottom Line
Chronic obstructive pulmonary disease (COPD) is a common condition with high morbidity and mortality. Comorbid COPD and tobacco smoking is especially prevalent in people with mental illness. Early diagnosis of COPD is important because appropriate management can alleviate symptoms, improve health status and exercise capacity, and prolong survival. You can play an integral role in COPD management by promoting smoking cessation and encouraging adherence to treatment.

 


Related Resources

 

  • Agency for Healthcare Research and Quality. Help for smokers and other tobacco users: quit smoking. www.ahrq.gov/professionals/clinicians-providers/guidelines-recommendations/tobacco/clinicians/tearsheets/helpsmokers.html.
  • Mayo Clinic. Diseases and conditions: COPD. www.mayoclinic.com/health/copd/DS00916.

 

  • Criner GJ, Bourbeau J, Diekemper RL, et al. Prevention of acute exacerbations of COPD: American College of Chest Physicians and Canadian Thoracic Society Guideline. Chest. 2015;147(4):894-942.

 

  • Global Initiative for Chronic Obstructive Lung Disease. Guidelines for professionals in the diagnosis and treatment of COPD. www.goldcopd.org/guidelines-global-strategy-for-diagnosis-management.html. 
     

Drug Brand Names
Bromocriptine • Parlodel
Bupropion • Wellbutrin SR, Zyban
Buspirone • BuSpar
Clozapine • Clozaril
Fluvoxamine • Luvox
Haloperidol • Haldol
Nortriptyline • Pamelor
Olanzapine • Zyprexa
Theophylline • Theolair
Varenicline • Chantix

 


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


1. Miller BJ, Paschall CB 3rd, Svendsen DP. Mortality and medical comorbidity among patients with serious mental illness. Psychiatr Serv. 2006;57(10):1482-1487.
2. Saha S, Chant D, McGrath J. A systematic review of mortality in schizophrenia: is the differential mortality gap worsening over time? Arch Gen Psychiatry. 2007;64(10):1123-1131.
3. Freeman E, Yoe JT. The poor health status of consumers of mental healthcare: behavioral disorders and chronic disease. Paper presented at: the National Association of State Mental Health Program Directors Medical Directors Workgroup; May 2006; Alexandria, VA.
4. Global Initiative for Chronic Obstructive Lung Disease. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease. http://www.goldcopd.org/uploads/users/files/GOLD_Report_2013_Feb20.pdf. Published February 20, 2013. Accessed March 2, 2016.
5. AntÒ JM, Vermeire P, Vestbo J, et al. Epidemiology of chronic obstructive pulmonary disease. Eur Respir J. 2001;17(5):982-994.
6. Newcomer JW. Antipsychotic medications: metabolic and cardiovascular risk. J Clin Psychiatry. 2007;68(suppl 4):8-13.
7. Calverley PM, Walker P. Chronic obstructive pulmonary disease. Lancet. 2003;362(9389):1053-1061.
8. Centers for Disease Control and Prevention. Chronic obstructive pulmonary disease among adults—United States, 2011. MMWR Morb Mortal Wkly Rep. 2012;61:938-943.
9. Feenstra TL, van Genugten ML, Hoogenveen RT, et al. The impact of aging and smoking on the future burden of chronic obstructive pulmonary disease: a model analysis in the Netherlands. Am J Respir Crit Care Med. 2001;164(4):590-596.
10. Celli BR, MacNee W; ATS/ERS Task Force. Standards for the diagnosis and treatment of patients with COPD: a summary of the ATS/ERS position paper [Erratum in: Eur Respir J. 2006;27(1):242]. Eur Respir J. 2004;23(6):932-946.
11. Matsuba K, Wright JL, Wiggs BR, et al. The changes in airways structure associated with reduced forced expiratory volume in one second. Eur Respir J. 1989;2(9):834-839.
12. O’Donnell DE, Revill SM, Webb KA. Dynamic hyperinflation and exercise intolerance in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2001;164(5):770-777.
13. Volkov VP. Respiratory diseases as a cause of death in schizophrenia [article in Russian]. Probl Tuberk Bolezn Legk. 2009;(6):24-27.
14. Kroon LA. Drug interactions and smoking: raising awareness for acute and critical care providers. Crit Care Nurs Clin North Am. 2006;18(1):53-62, xii.
15. Borson S, McDonald GJ, Gayle T, et al. Improvement in mood, physical symptoms, and function with nortriptyline for depression in patients with chronic obstructive pulmonary disease. Psychosomatics. 1992;33(2):190-201.
16. Caskey NH, Jarvik ME, Wirshing WC. The effects of dopaminergic D2 stimulation and blockade on smoking behavior. Exp Clin Psychopharmacol. 1999;7(1):72-78.
17. Dawe S, Gerada C, Russell MA, et al. Nicotine intake in smokers increases following a single dose of haloperidol. Psychopharmacol (Berl). 1995;117(1):110-115.
18. de Haan L, Booji J, Lavalaye J, et al. Occupancy of dopamine D2 receptors by antipsychotic drugs is related to nicotine addiction in young patients with schizophrenia. Psychopharmacology (Berl). 2006;183(4):500-505.
19. Hertel P, Nomikos GG, Iurlo M, et al. Risperidone: regional effects in vivo on release and metabolism of dopamine and serotonin in the rat brain. Psychopharmacology (Berl). 1996;124(1-2):74-86.
20. 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.
21. Hutchison KE, Rutter MC, Niaura R, et al. Olanzapine attenuates cue-elicited craving for tobacco. Psychopharmacology (Berl). 2004;175(4):407-413.
22. Kumari V, Postma P. Nicotine use in schizophrenia: the self medication hypotheses. Neurosci Biobehav Rev. 2005;29(6):1021-1034.
23. Procyshyn RM, Tse G, Sin O, et al. Concomitant clozapine reduces smoking in patients treated with risperidone. Eur Neuropsychopharmacol. 2002;12(1):77-80.
24. Anthonisen NR, Connett JE, Kiley JP, et al. Effects of smoking intervention and the use of an inhaled anticholinergic bronchodilator on the rate of decline of FEV1. The Lung Health Study. JAMA. 1994;272(19):1497-1505.
25. Pisinger C, Godtfredsen NS. Is there a health benefit of reduced tobacco consumption? A systematic review. Nicotine Tob Res. 2007;9(6):631-646.
26. Fiore MC, Jaén CR, Baker TB, et al. Treating tobacco use and dependence: 2008 update. Clinical Practice Guideline. Rockville, MD: Public Health Service, US Department of Health and Human Services; 2008.
27. Barnes M, Lawford BR, Burton SC, et al. Smoking and schizophrenia: is symptom profile related to smoking and which antipsychotic medication is of benefit in reducing cigarette use? Aust N Z J Psychiatry. 2006;40(6-7):575-580.
28. Screening for chronic obstructive pulmonary disease using spirometry: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2008;148(7):529-534.
29. Agency for Healthcare Research and Quality. Five major steps to intervention (The “5 A’s”). http://www.ahrq.gov/professionals/clinicians-providers/guidelines-recommendations/tobacco/5steps.html. Published 2012. Accessed March 2, 2016.
30. Addington J, el-Guebaly N, Campbell W, et al. Smoking cessation treatment for patients with schizophrenia. Am J Psychiatry. 1998;155(7):974-976.
31. Zarowitz BJ, O’Shea T. Chronic obstructive pulmonary disease: prevalence, characteristics, and pharmacologic treatment in nursing home residents with cognitive impairment. J Manag Care Pharm. 2012;18(8):598-606.
32. Schermer T, Smeenk F, van Weel C. Referral and consultation in asthma and COPD: an exploration of pulmonologists’ views. Neth J Med. 2003;61(3):71-81.

Article PDF
Author and Disclosure Information

 

Abdulkader Alam, MD
Assistant Professor of Psychiatry and Internal Medicine
Department of Psychiatry and Internal Medicine
Stony Brook University School of Medicine
Stony Brook, New York

R. Michael Huijon, MD
Resident, Combined Psychiatry and Family Medicine
University of Pittsburgh
Pittsburgh, Pennsylvania

Vijaya Alla, MD
Resident Family Medicine
Stony Brook University School of Medicine
Stony Brook, New York

Nicole Rivera, BS
Medical Student
Stony Brook University School of Medicine
Stony Brook, New York

Issue
Current Psychiatry - 15(5)
Publications
Topics
Page Number
68-74
Legacy Keywords
COPD, pulmonary, chronic obstructive pulmonary disease, cardiovascular disease, respiratory disease, somatic disorder, somatic disorders
Sections
Author and Disclosure Information

 

Abdulkader Alam, MD
Assistant Professor of Psychiatry and Internal Medicine
Department of Psychiatry and Internal Medicine
Stony Brook University School of Medicine
Stony Brook, New York

R. Michael Huijon, MD
Resident, Combined Psychiatry and Family Medicine
University of Pittsburgh
Pittsburgh, Pennsylvania

Vijaya Alla, MD
Resident Family Medicine
Stony Brook University School of Medicine
Stony Brook, New York

Nicole Rivera, BS
Medical Student
Stony Brook University School of Medicine
Stony Brook, New York

Author and Disclosure Information

 

Abdulkader Alam, MD
Assistant Professor of Psychiatry and Internal Medicine
Department of Psychiatry and Internal Medicine
Stony Brook University School of Medicine
Stony Brook, New York

R. Michael Huijon, MD
Resident, Combined Psychiatry and Family Medicine
University of Pittsburgh
Pittsburgh, Pennsylvania

Vijaya Alla, MD
Resident Family Medicine
Stony Brook University School of Medicine
Stony Brook, New York

Nicole Rivera, BS
Medical Student
Stony Brook University School of Medicine
Stony Brook, New York

Article PDF
Article PDF

Chronic obstructive pulmonary disease (COPD) usually is not diagnosed until clinically apparent and moderately advanced. Patients might not notice chronic dyspnea and smoker’s cough, or might consider their symptoms “normal” and not seek medical care. Delayed diagnosis is particularly prevalent in the psychiatric population, in which co-existing medical problems tend to remain unrecognized and untreated.1

Life expectancy of people with serious mental illness (SMI) is 13 to 30 years less than that of the general population—a gap that has widened over time.2 Pulmonary disease is a leading cause of elevated mortality risk in SMI, along with cardiovascular and infectious disease, diabetes, and barriers to care. Having a comorbid mental illness triples the mortality risk of chronic lower respiratory disease (Table 1).3


This article describes how you can intervene and improve quality of life for your patients with COPD by:

 

  • asking all patients, especially smokers, if they are experiencing classic symptoms of COPD
  • advocating for and supporting smoking cessation efforts
  • avoiding drug interactions and off-target dosing related to COPD and nicotine replacement therapy
  • considering, if feasible, a switch from typical to atypical antipsychotic therapy, which could reduce smoking behavior.


What is COPD?
COPD is preventable and treatable. It is characterized by “persistent airflow limitation that is usually progressive and associated with an enhanced chronic inflammatory response in the airways and the lungs to inhaled noxious particles or gases.”4

Smoking tobacco is the greatest risk factor for developing COPD.5 An estimated 50% to 80% of people with schizophrenia are smokers, as are 55% of people with bipolar disorder.6 COPD is a leading cause of morbidity and mortality worldwide,7,8 and its prevalence is projected to increase as the global population and smoking rates grow.9

A simplified schema of the pathophysiology of COPD implicates 4 lung areas: parenchyma, pulmonary vasculature, central airways, and peripheral airways.10 Variation in the areas affected and severity of change contributes to the disease’s heterogeneous presentation, which can include pulmonary hypertension, hypersecretion of mucus, ciliary dysfunction, airway hyperinflation, and impaired gas exchange.11,12 Many of these features lead to systemic effects as well, particularly on cardiac function.


When to test a patient for COPD
Early diagnosis and treatment can substantially improve quality-of-life outcomes for patients with COPD. The clinical approach (Figure 1) begins with recognizing classic symptoms. Consider COPD in any patient with:

 

  • dyspnea (particularly if becoming worse, persistent, or associated with exercise)
  • chronic cough
  • chronic sputum production
  • history of risk-factor exposure (particularly tobacco smoke)
  • family history of COPD.4


If the history and physical exam suggest COPD (Table 2), spirometry is the most reliable test to quantify and characterize lung dysfunction. It is not indicated as a screening tool for healthy adults or appropriate when a patient is acutely ill. Forced expiratory volume in the first second of expiration divided by the measured forced vital capacity (FEV1/FVC) < 0.7 defines clinical COPD and determines the need for pharmacologic intervention. Laboratory studies could be useful in certain clinical scenarios, such as serum testing for alpha1-antitrypsin deficiency in patients age <45 with emphysema. Plain film imaging might be useful to support a COPD diagnosis or rule out alternate diagnoses.


Psychopharmacology issues with comorbid COPD
Pharmacotherapy for psychiatric disorders can exacerbate comorbid COPD. For example, long-term use of phenothiazine-related typical antipsychotics for schizophrenia has been linked to an increased incidence of COPD.13 Antipsychotic side effects such as acute laryngeal dystonia and tardive dyskinesia, most commonly seen with first-generation antipsychotic use, can aggravate dyspnea caused by COPD. Opioids and most hypnotics, sedatives, and anxiolytics suppress the respiratory drive, and therefore should be used with caution in patients with COPD.

Carefully monitor serum levels of medications before and during attempts at smoking cessation. Nicotine’s induction of the cytochrome P450 1A2 system increases the metabolism of antipsychotics such as clozapine, fluvoxamine, olanzapine, and haloperidol. As a result, potentially toxic drug levels can occur when a smoker tries to quit.14

Screen patients with COPD for co­morbid psychiatric conditions. New psychiatric symptoms can emerge after COPD has been diagnosed, even in patients without pre-existing psychopathology.

Anxiety is a particularly common COPD comorbidity that can be difficult to manage. Selective serotonin reuptake inhibitors, buspirone, cognitive-behavioral therapy, and pulmonary rehabilitation can be helpful, although the effect of antidepressants on respiration is controversial. Nortriptyline has been shown to be effective in treating both anxiety and depressive symptoms in patients with COPD.15 Avoid using hypnotics to manage sleep problems related to COPD; instead, focus on minimizing sleep disturbance by limiting cough and dyspnea.


Antipsychotics and nicotine metabolism
Multiple studies have focused on the interplay among nicotine, dopamine, and antipsychotic agents. Nicotine receptors are present in the ventral tegmental dopaminergic cell bodies, which induce the release of dopamine and other neurotransmitters when stimulated. Smoking has been noted to increase in patients administered haloperidol (a dopamine antagonist) and to decrease with administration of bromocriptine (a dopamine agonist).16 This suggests that psychiatric patients might smoke to overcome the dopamine blockade caused by most typical antipsychotics, therefore alleviating their negative and extrapyramidal side effects.17

 

 

Alternatively, some studies suggest that a difference in dopamine receptor occupancy between typical and atypical antipsychotics leads to different effects on smoking behavior.18 When used long term, typical antipsychotics might increase dopamine receptors or dopamine sensitivity, and thus reinforce the positive effect of nicotine by increasing the number of receptors that can be stimulated, whereas atypical antipsychotics help stimulate the release of dopamine directly through partial agonist of serotonin 5-HT1A receptors.19,20 Atypical antipsychotics also appear to decrease cue-elicited cravings in people who are not mentally ill, whereas haloperidol does not.21

Based on these findings, switching patients with COPD from a typical to an atypical antipsychotic, if feasible, might make smoking cessation more manageable.22 Multiple studies have shown that clozapine is the preferred atypical antipsychotic because it is associated with the most significant decrease in smoking behaviors.23


First-line therapy: Nicotine replacement
Smoking cessation slows the progression of COPD and leads to marked improvements in cough, expectoration, breathlessness, and wheezing.24,25 Nicotine replacement therapy (NRT)—gum, inhaler, lozenges, nasal spray, and skin patch—is considered first-line pharmacotherapy. These nicotine substitutes can decrease withdrawal symptoms, although they do not appear to be as effective for light smokers (eg, <10 cigarettes/d), compared with heavy smokers (eg, ≥20 cigarettes/d).26

Long-term smoking abstinence can be improved with combination therapies. A nicotine patch, kept in place for as long as 24 hours, often is used with a nicotine gum or nasal spray. Another option combines the patch with a first-line, non-NRT intervention, such as sustained-release bupropion. Use bupropion with caution in psychiatric patients, however. Do not combine it with a monoamine oxidase inhibitor, and do not prescribe it to patients with an eating disorder or history of seizures.26 Bupropion could induce mania in patients with bipolar disorder.

Varenicline, a nicotinic receptor partial agonist indicated to aid in smoking cessation, has been shown to reduce pleasure gained from tobacco as well as cravings. It can increase the likelihood of abstinence from smoking for as long as 1 year, but it also can provoke behavioral changes, depressed mood, and suicidal ideation. These risks—described in an FDA black-box warning of serious neuropsychiatric events—warrant due caution when prescribing varenicline to patients with depression. The FDA also has warned that varenicline could lead to decreased alcohol tolerance and atypically aggressive behavior during intoxication, which is of particular concern because of the high rate of alcohol use among people with SMI.


Motivating and supporting change
When counseling patients with mental illness about smoking cessation, consider unique motivations that, if disregarded, could undermine your efforts. As described above, smoking can ameliorate negative and extrapyramidal symptoms associated with typical antipsychotics. This could explain the significantly higher rates of smoking associated with typical antipsychotics, compared with atypical antipsychotics.27 Patients also could use smoking as self-medication for depression and anxiety. Therefore, take care to offer alternate methods for coping, along with smoking cessation recommendations.22

Screen all adult patients for tobacco use, and offer prompt cessation counseling and pharmacologic interventions.28As a motivational intervention, the “5 As” framework—ask, advise, assess, assist, arrange—can help gauge patients’ smoking status and willingness to quit, as well as emphasize the importance of establishing a concrete, manageable plan.29

Keep in mind the barriers all patients face in their fight to quit smoking, such as nicotine withdrawal, weight gain, and loss of a coping mechanism for stress.29 Patients with schizophrenia can be motivated to quit smoking and participate in treatment for nicotine dependence.30

Besides encouraging smoking cessation, you can educate patients in behaviors that will improve COPD symptoms and management. These include:

 

  • reducing the risk of lung infections through vaccinations (influenza yearly, pneumonia once in adulthood) and avoiding crowds during peak cold and influenza season
  • participating in physical activity, which could slow lung function decline
  • adhering to prescribed medication
  • eating a balanced diet
  • seeking medical care early during an exacerbation.


Coaching patients in symptom control
Smoking cessation may have the greatest long-term benefit for patients with COPD, but symptom management is important as well (Figure 2). Pharmacotherapy for COPD usually is advanced in steps, but a more aggressive approach may be necessary for patients presenting with severe symptoms.


Mainstays of COPD therapy are inhaled bronchodilators, consisting of β2 agonists and anticholinergics, alone or in combination. Short-acting formulations are used for mild and intermittent symptoms; long-acting bronchodilators are added if symptoms persist.4 When dyspnea, wheezing, and activity intolerance are not well-controlled with bronchodilators, an inhaled corticosteroid can be tried, either alone or in combination with a long-acting bronchodilator.4

Adherence to medical recommendations is critical for successful COPD management, but inhaled therapy can be difficult for psychiatric patients—especially patients with cognitive or functional impairment. Asking them to demonstrate their inhaler technique can help assess treatment effectiveness.31

 

 

Referral to a pulmonologist is strongly advised in cases of:

 

  • advanced, end-stage COPD (FEV1 <50% predicted value despite adherence to recommended treatment, or rapid decline of FEV1)
  • COPD in patients age <50
  • frequent exacerbations
  • possible complications related to chronic heart failure
  • indications for oxygen treatment (eg, resting or ambulatory oxygen saturation ≤88% or PaO2 ≤55 mm Hg).32

 


Bottom Line
Chronic obstructive pulmonary disease (COPD) is a common condition with high morbidity and mortality. Comorbid COPD and tobacco smoking is especially prevalent in people with mental illness. Early diagnosis of COPD is important because appropriate management can alleviate symptoms, improve health status and exercise capacity, and prolong survival. You can play an integral role in COPD management by promoting smoking cessation and encouraging adherence to treatment.

 


Related Resources

 

  • Agency for Healthcare Research and Quality. Help for smokers and other tobacco users: quit smoking. www.ahrq.gov/professionals/clinicians-providers/guidelines-recommendations/tobacco/clinicians/tearsheets/helpsmokers.html.
  • Mayo Clinic. Diseases and conditions: COPD. www.mayoclinic.com/health/copd/DS00916.

 

  • Criner GJ, Bourbeau J, Diekemper RL, et al. Prevention of acute exacerbations of COPD: American College of Chest Physicians and Canadian Thoracic Society Guideline. Chest. 2015;147(4):894-942.

 

  • Global Initiative for Chronic Obstructive Lung Disease. Guidelines for professionals in the diagnosis and treatment of COPD. www.goldcopd.org/guidelines-global-strategy-for-diagnosis-management.html. 
     

Drug Brand Names
Bromocriptine • Parlodel
Bupropion • Wellbutrin SR, Zyban
Buspirone • BuSpar
Clozapine • Clozaril
Fluvoxamine • Luvox
Haloperidol • Haldol
Nortriptyline • Pamelor
Olanzapine • Zyprexa
Theophylline • Theolair
Varenicline • Chantix

 


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

Chronic obstructive pulmonary disease (COPD) usually is not diagnosed until clinically apparent and moderately advanced. Patients might not notice chronic dyspnea and smoker’s cough, or might consider their symptoms “normal” and not seek medical care. Delayed diagnosis is particularly prevalent in the psychiatric population, in which co-existing medical problems tend to remain unrecognized and untreated.1

Life expectancy of people with serious mental illness (SMI) is 13 to 30 years less than that of the general population—a gap that has widened over time.2 Pulmonary disease is a leading cause of elevated mortality risk in SMI, along with cardiovascular and infectious disease, diabetes, and barriers to care. Having a comorbid mental illness triples the mortality risk of chronic lower respiratory disease (Table 1).3


This article describes how you can intervene and improve quality of life for your patients with COPD by:

 

  • asking all patients, especially smokers, if they are experiencing classic symptoms of COPD
  • advocating for and supporting smoking cessation efforts
  • avoiding drug interactions and off-target dosing related to COPD and nicotine replacement therapy
  • considering, if feasible, a switch from typical to atypical antipsychotic therapy, which could reduce smoking behavior.


What is COPD?
COPD is preventable and treatable. It is characterized by “persistent airflow limitation that is usually progressive and associated with an enhanced chronic inflammatory response in the airways and the lungs to inhaled noxious particles or gases.”4

Smoking tobacco is the greatest risk factor for developing COPD.5 An estimated 50% to 80% of people with schizophrenia are smokers, as are 55% of people with bipolar disorder.6 COPD is a leading cause of morbidity and mortality worldwide,7,8 and its prevalence is projected to increase as the global population and smoking rates grow.9

A simplified schema of the pathophysiology of COPD implicates 4 lung areas: parenchyma, pulmonary vasculature, central airways, and peripheral airways.10 Variation in the areas affected and severity of change contributes to the disease’s heterogeneous presentation, which can include pulmonary hypertension, hypersecretion of mucus, ciliary dysfunction, airway hyperinflation, and impaired gas exchange.11,12 Many of these features lead to systemic effects as well, particularly on cardiac function.


When to test a patient for COPD
Early diagnosis and treatment can substantially improve quality-of-life outcomes for patients with COPD. The clinical approach (Figure 1) begins with recognizing classic symptoms. Consider COPD in any patient with:

 

  • dyspnea (particularly if becoming worse, persistent, or associated with exercise)
  • chronic cough
  • chronic sputum production
  • history of risk-factor exposure (particularly tobacco smoke)
  • family history of COPD.4


If the history and physical exam suggest COPD (Table 2), spirometry is the most reliable test to quantify and characterize lung dysfunction. It is not indicated as a screening tool for healthy adults or appropriate when a patient is acutely ill. Forced expiratory volume in the first second of expiration divided by the measured forced vital capacity (FEV1/FVC) < 0.7 defines clinical COPD and determines the need for pharmacologic intervention. Laboratory studies could be useful in certain clinical scenarios, such as serum testing for alpha1-antitrypsin deficiency in patients age <45 with emphysema. Plain film imaging might be useful to support a COPD diagnosis or rule out alternate diagnoses.


Psychopharmacology issues with comorbid COPD
Pharmacotherapy for psychiatric disorders can exacerbate comorbid COPD. For example, long-term use of phenothiazine-related typical antipsychotics for schizophrenia has been linked to an increased incidence of COPD.13 Antipsychotic side effects such as acute laryngeal dystonia and tardive dyskinesia, most commonly seen with first-generation antipsychotic use, can aggravate dyspnea caused by COPD. Opioids and most hypnotics, sedatives, and anxiolytics suppress the respiratory drive, and therefore should be used with caution in patients with COPD.

Carefully monitor serum levels of medications before and during attempts at smoking cessation. Nicotine’s induction of the cytochrome P450 1A2 system increases the metabolism of antipsychotics such as clozapine, fluvoxamine, olanzapine, and haloperidol. As a result, potentially toxic drug levels can occur when a smoker tries to quit.14

Screen patients with COPD for co­morbid psychiatric conditions. New psychiatric symptoms can emerge after COPD has been diagnosed, even in patients without pre-existing psychopathology.

Anxiety is a particularly common COPD comorbidity that can be difficult to manage. Selective serotonin reuptake inhibitors, buspirone, cognitive-behavioral therapy, and pulmonary rehabilitation can be helpful, although the effect of antidepressants on respiration is controversial. Nortriptyline has been shown to be effective in treating both anxiety and depressive symptoms in patients with COPD.15 Avoid using hypnotics to manage sleep problems related to COPD; instead, focus on minimizing sleep disturbance by limiting cough and dyspnea.


Antipsychotics and nicotine metabolism
Multiple studies have focused on the interplay among nicotine, dopamine, and antipsychotic agents. Nicotine receptors are present in the ventral tegmental dopaminergic cell bodies, which induce the release of dopamine and other neurotransmitters when stimulated. Smoking has been noted to increase in patients administered haloperidol (a dopamine antagonist) and to decrease with administration of bromocriptine (a dopamine agonist).16 This suggests that psychiatric patients might smoke to overcome the dopamine blockade caused by most typical antipsychotics, therefore alleviating their negative and extrapyramidal side effects.17

 

 

Alternatively, some studies suggest that a difference in dopamine receptor occupancy between typical and atypical antipsychotics leads to different effects on smoking behavior.18 When used long term, typical antipsychotics might increase dopamine receptors or dopamine sensitivity, and thus reinforce the positive effect of nicotine by increasing the number of receptors that can be stimulated, whereas atypical antipsychotics help stimulate the release of dopamine directly through partial agonist of serotonin 5-HT1A receptors.19,20 Atypical antipsychotics also appear to decrease cue-elicited cravings in people who are not mentally ill, whereas haloperidol does not.21

Based on these findings, switching patients with COPD from a typical to an atypical antipsychotic, if feasible, might make smoking cessation more manageable.22 Multiple studies have shown that clozapine is the preferred atypical antipsychotic because it is associated with the most significant decrease in smoking behaviors.23


First-line therapy: Nicotine replacement
Smoking cessation slows the progression of COPD and leads to marked improvements in cough, expectoration, breathlessness, and wheezing.24,25 Nicotine replacement therapy (NRT)—gum, inhaler, lozenges, nasal spray, and skin patch—is considered first-line pharmacotherapy. These nicotine substitutes can decrease withdrawal symptoms, although they do not appear to be as effective for light smokers (eg, <10 cigarettes/d), compared with heavy smokers (eg, ≥20 cigarettes/d).26

Long-term smoking abstinence can be improved with combination therapies. A nicotine patch, kept in place for as long as 24 hours, often is used with a nicotine gum or nasal spray. Another option combines the patch with a first-line, non-NRT intervention, such as sustained-release bupropion. Use bupropion with caution in psychiatric patients, however. Do not combine it with a monoamine oxidase inhibitor, and do not prescribe it to patients with an eating disorder or history of seizures.26 Bupropion could induce mania in patients with bipolar disorder.

Varenicline, a nicotinic receptor partial agonist indicated to aid in smoking cessation, has been shown to reduce pleasure gained from tobacco as well as cravings. It can increase the likelihood of abstinence from smoking for as long as 1 year, but it also can provoke behavioral changes, depressed mood, and suicidal ideation. These risks—described in an FDA black-box warning of serious neuropsychiatric events—warrant due caution when prescribing varenicline to patients with depression. The FDA also has warned that varenicline could lead to decreased alcohol tolerance and atypically aggressive behavior during intoxication, which is of particular concern because of the high rate of alcohol use among people with SMI.


Motivating and supporting change
When counseling patients with mental illness about smoking cessation, consider unique motivations that, if disregarded, could undermine your efforts. As described above, smoking can ameliorate negative and extrapyramidal symptoms associated with typical antipsychotics. This could explain the significantly higher rates of smoking associated with typical antipsychotics, compared with atypical antipsychotics.27 Patients also could use smoking as self-medication for depression and anxiety. Therefore, take care to offer alternate methods for coping, along with smoking cessation recommendations.22

Screen all adult patients for tobacco use, and offer prompt cessation counseling and pharmacologic interventions.28As a motivational intervention, the “5 As” framework—ask, advise, assess, assist, arrange—can help gauge patients’ smoking status and willingness to quit, as well as emphasize the importance of establishing a concrete, manageable plan.29

Keep in mind the barriers all patients face in their fight to quit smoking, such as nicotine withdrawal, weight gain, and loss of a coping mechanism for stress.29 Patients with schizophrenia can be motivated to quit smoking and participate in treatment for nicotine dependence.30

Besides encouraging smoking cessation, you can educate patients in behaviors that will improve COPD symptoms and management. These include:

 

  • reducing the risk of lung infections through vaccinations (influenza yearly, pneumonia once in adulthood) and avoiding crowds during peak cold and influenza season
  • participating in physical activity, which could slow lung function decline
  • adhering to prescribed medication
  • eating a balanced diet
  • seeking medical care early during an exacerbation.


Coaching patients in symptom control
Smoking cessation may have the greatest long-term benefit for patients with COPD, but symptom management is important as well (Figure 2). Pharmacotherapy for COPD usually is advanced in steps, but a more aggressive approach may be necessary for patients presenting with severe symptoms.


Mainstays of COPD therapy are inhaled bronchodilators, consisting of β2 agonists and anticholinergics, alone or in combination. Short-acting formulations are used for mild and intermittent symptoms; long-acting bronchodilators are added if symptoms persist.4 When dyspnea, wheezing, and activity intolerance are not well-controlled with bronchodilators, an inhaled corticosteroid can be tried, either alone or in combination with a long-acting bronchodilator.4

Adherence to medical recommendations is critical for successful COPD management, but inhaled therapy can be difficult for psychiatric patients—especially patients with cognitive or functional impairment. Asking them to demonstrate their inhaler technique can help assess treatment effectiveness.31

 

 

Referral to a pulmonologist is strongly advised in cases of:

 

  • advanced, end-stage COPD (FEV1 <50% predicted value despite adherence to recommended treatment, or rapid decline of FEV1)
  • COPD in patients age <50
  • frequent exacerbations
  • possible complications related to chronic heart failure
  • indications for oxygen treatment (eg, resting or ambulatory oxygen saturation ≤88% or PaO2 ≤55 mm Hg).32

 


Bottom Line
Chronic obstructive pulmonary disease (COPD) is a common condition with high morbidity and mortality. Comorbid COPD and tobacco smoking is especially prevalent in people with mental illness. Early diagnosis of COPD is important because appropriate management can alleviate symptoms, improve health status and exercise capacity, and prolong survival. You can play an integral role in COPD management by promoting smoking cessation and encouraging adherence to treatment.

 


Related Resources

 

  • Agency for Healthcare Research and Quality. Help for smokers and other tobacco users: quit smoking. www.ahrq.gov/professionals/clinicians-providers/guidelines-recommendations/tobacco/clinicians/tearsheets/helpsmokers.html.
  • Mayo Clinic. Diseases and conditions: COPD. www.mayoclinic.com/health/copd/DS00916.

 

  • Criner GJ, Bourbeau J, Diekemper RL, et al. Prevention of acute exacerbations of COPD: American College of Chest Physicians and Canadian Thoracic Society Guideline. Chest. 2015;147(4):894-942.

 

  • Global Initiative for Chronic Obstructive Lung Disease. Guidelines for professionals in the diagnosis and treatment of COPD. www.goldcopd.org/guidelines-global-strategy-for-diagnosis-management.html. 
     

Drug Brand Names
Bromocriptine • Parlodel
Bupropion • Wellbutrin SR, Zyban
Buspirone • BuSpar
Clozapine • Clozaril
Fluvoxamine • Luvox
Haloperidol • Haldol
Nortriptyline • Pamelor
Olanzapine • Zyprexa
Theophylline • Theolair
Varenicline • Chantix

 


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


1. Miller BJ, Paschall CB 3rd, Svendsen DP. Mortality and medical comorbidity among patients with serious mental illness. Psychiatr Serv. 2006;57(10):1482-1487.
2. Saha S, Chant D, McGrath J. A systematic review of mortality in schizophrenia: is the differential mortality gap worsening over time? Arch Gen Psychiatry. 2007;64(10):1123-1131.
3. Freeman E, Yoe JT. The poor health status of consumers of mental healthcare: behavioral disorders and chronic disease. Paper presented at: the National Association of State Mental Health Program Directors Medical Directors Workgroup; May 2006; Alexandria, VA.
4. Global Initiative for Chronic Obstructive Lung Disease. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease. http://www.goldcopd.org/uploads/users/files/GOLD_Report_2013_Feb20.pdf. Published February 20, 2013. Accessed March 2, 2016.
5. AntÒ JM, Vermeire P, Vestbo J, et al. Epidemiology of chronic obstructive pulmonary disease. Eur Respir J. 2001;17(5):982-994.
6. Newcomer JW. Antipsychotic medications: metabolic and cardiovascular risk. J Clin Psychiatry. 2007;68(suppl 4):8-13.
7. Calverley PM, Walker P. Chronic obstructive pulmonary disease. Lancet. 2003;362(9389):1053-1061.
8. Centers for Disease Control and Prevention. Chronic obstructive pulmonary disease among adults—United States, 2011. MMWR Morb Mortal Wkly Rep. 2012;61:938-943.
9. Feenstra TL, van Genugten ML, Hoogenveen RT, et al. The impact of aging and smoking on the future burden of chronic obstructive pulmonary disease: a model analysis in the Netherlands. Am J Respir Crit Care Med. 2001;164(4):590-596.
10. Celli BR, MacNee W; ATS/ERS Task Force. Standards for the diagnosis and treatment of patients with COPD: a summary of the ATS/ERS position paper [Erratum in: Eur Respir J. 2006;27(1):242]. Eur Respir J. 2004;23(6):932-946.
11. Matsuba K, Wright JL, Wiggs BR, et al. The changes in airways structure associated with reduced forced expiratory volume in one second. Eur Respir J. 1989;2(9):834-839.
12. O’Donnell DE, Revill SM, Webb KA. Dynamic hyperinflation and exercise intolerance in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2001;164(5):770-777.
13. Volkov VP. Respiratory diseases as a cause of death in schizophrenia [article in Russian]. Probl Tuberk Bolezn Legk. 2009;(6):24-27.
14. Kroon LA. Drug interactions and smoking: raising awareness for acute and critical care providers. Crit Care Nurs Clin North Am. 2006;18(1):53-62, xii.
15. Borson S, McDonald GJ, Gayle T, et al. Improvement in mood, physical symptoms, and function with nortriptyline for depression in patients with chronic obstructive pulmonary disease. Psychosomatics. 1992;33(2):190-201.
16. Caskey NH, Jarvik ME, Wirshing WC. The effects of dopaminergic D2 stimulation and blockade on smoking behavior. Exp Clin Psychopharmacol. 1999;7(1):72-78.
17. Dawe S, Gerada C, Russell MA, et al. Nicotine intake in smokers increases following a single dose of haloperidol. Psychopharmacol (Berl). 1995;117(1):110-115.
18. de Haan L, Booji J, Lavalaye J, et al. Occupancy of dopamine D2 receptors by antipsychotic drugs is related to nicotine addiction in young patients with schizophrenia. Psychopharmacology (Berl). 2006;183(4):500-505.
19. Hertel P, Nomikos GG, Iurlo M, et al. Risperidone: regional effects in vivo on release and metabolism of dopamine and serotonin in the rat brain. Psychopharmacology (Berl). 1996;124(1-2):74-86.
20. 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.
21. Hutchison KE, Rutter MC, Niaura R, et al. Olanzapine attenuates cue-elicited craving for tobacco. Psychopharmacology (Berl). 2004;175(4):407-413.
22. Kumari V, Postma P. Nicotine use in schizophrenia: the self medication hypotheses. Neurosci Biobehav Rev. 2005;29(6):1021-1034.
23. Procyshyn RM, Tse G, Sin O, et al. Concomitant clozapine reduces smoking in patients treated with risperidone. Eur Neuropsychopharmacol. 2002;12(1):77-80.
24. Anthonisen NR, Connett JE, Kiley JP, et al. Effects of smoking intervention and the use of an inhaled anticholinergic bronchodilator on the rate of decline of FEV1. The Lung Health Study. JAMA. 1994;272(19):1497-1505.
25. Pisinger C, Godtfredsen NS. Is there a health benefit of reduced tobacco consumption? A systematic review. Nicotine Tob Res. 2007;9(6):631-646.
26. Fiore MC, Jaén CR, Baker TB, et al. Treating tobacco use and dependence: 2008 update. Clinical Practice Guideline. Rockville, MD: Public Health Service, US Department of Health and Human Services; 2008.
27. Barnes M, Lawford BR, Burton SC, et al. Smoking and schizophrenia: is symptom profile related to smoking and which antipsychotic medication is of benefit in reducing cigarette use? Aust N Z J Psychiatry. 2006;40(6-7):575-580.
28. Screening for chronic obstructive pulmonary disease using spirometry: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2008;148(7):529-534.
29. Agency for Healthcare Research and Quality. Five major steps to intervention (The “5 A’s”). http://www.ahrq.gov/professionals/clinicians-providers/guidelines-recommendations/tobacco/5steps.html. Published 2012. Accessed March 2, 2016.
30. Addington J, el-Guebaly N, Campbell W, et al. Smoking cessation treatment for patients with schizophrenia. Am J Psychiatry. 1998;155(7):974-976.
31. Zarowitz BJ, O’Shea T. Chronic obstructive pulmonary disease: prevalence, characteristics, and pharmacologic treatment in nursing home residents with cognitive impairment. J Manag Care Pharm. 2012;18(8):598-606.
32. Schermer T, Smeenk F, van Weel C. Referral and consultation in asthma and COPD: an exploration of pulmonologists’ views. Neth J Med. 2003;61(3):71-81.

References


1. Miller BJ, Paschall CB 3rd, Svendsen DP. Mortality and medical comorbidity among patients with serious mental illness. Psychiatr Serv. 2006;57(10):1482-1487.
2. Saha S, Chant D, McGrath J. A systematic review of mortality in schizophrenia: is the differential mortality gap worsening over time? Arch Gen Psychiatry. 2007;64(10):1123-1131.
3. Freeman E, Yoe JT. The poor health status of consumers of mental healthcare: behavioral disorders and chronic disease. Paper presented at: the National Association of State Mental Health Program Directors Medical Directors Workgroup; May 2006; Alexandria, VA.
4. Global Initiative for Chronic Obstructive Lung Disease. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease. http://www.goldcopd.org/uploads/users/files/GOLD_Report_2013_Feb20.pdf. Published February 20, 2013. Accessed March 2, 2016.
5. AntÒ JM, Vermeire P, Vestbo J, et al. Epidemiology of chronic obstructive pulmonary disease. Eur Respir J. 2001;17(5):982-994.
6. Newcomer JW. Antipsychotic medications: metabolic and cardiovascular risk. J Clin Psychiatry. 2007;68(suppl 4):8-13.
7. Calverley PM, Walker P. Chronic obstructive pulmonary disease. Lancet. 2003;362(9389):1053-1061.
8. Centers for Disease Control and Prevention. Chronic obstructive pulmonary disease among adults—United States, 2011. MMWR Morb Mortal Wkly Rep. 2012;61:938-943.
9. Feenstra TL, van Genugten ML, Hoogenveen RT, et al. The impact of aging and smoking on the future burden of chronic obstructive pulmonary disease: a model analysis in the Netherlands. Am J Respir Crit Care Med. 2001;164(4):590-596.
10. Celli BR, MacNee W; ATS/ERS Task Force. Standards for the diagnosis and treatment of patients with COPD: a summary of the ATS/ERS position paper [Erratum in: Eur Respir J. 2006;27(1):242]. Eur Respir J. 2004;23(6):932-946.
11. Matsuba K, Wright JL, Wiggs BR, et al. The changes in airways structure associated with reduced forced expiratory volume in one second. Eur Respir J. 1989;2(9):834-839.
12. O’Donnell DE, Revill SM, Webb KA. Dynamic hyperinflation and exercise intolerance in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2001;164(5):770-777.
13. Volkov VP. Respiratory diseases as a cause of death in schizophrenia [article in Russian]. Probl Tuberk Bolezn Legk. 2009;(6):24-27.
14. Kroon LA. Drug interactions and smoking: raising awareness for acute and critical care providers. Crit Care Nurs Clin North Am. 2006;18(1):53-62, xii.
15. Borson S, McDonald GJ, Gayle T, et al. Improvement in mood, physical symptoms, and function with nortriptyline for depression in patients with chronic obstructive pulmonary disease. Psychosomatics. 1992;33(2):190-201.
16. Caskey NH, Jarvik ME, Wirshing WC. The effects of dopaminergic D2 stimulation and blockade on smoking behavior. Exp Clin Psychopharmacol. 1999;7(1):72-78.
17. Dawe S, Gerada C, Russell MA, et al. Nicotine intake in smokers increases following a single dose of haloperidol. Psychopharmacol (Berl). 1995;117(1):110-115.
18. de Haan L, Booji J, Lavalaye J, et al. Occupancy of dopamine D2 receptors by antipsychotic drugs is related to nicotine addiction in young patients with schizophrenia. Psychopharmacology (Berl). 2006;183(4):500-505.
19. Hertel P, Nomikos GG, Iurlo M, et al. Risperidone: regional effects in vivo on release and metabolism of dopamine and serotonin in the rat brain. Psychopharmacology (Berl). 1996;124(1-2):74-86.
20. 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.
21. Hutchison KE, Rutter MC, Niaura R, et al. Olanzapine attenuates cue-elicited craving for tobacco. Psychopharmacology (Berl). 2004;175(4):407-413.
22. Kumari V, Postma P. Nicotine use in schizophrenia: the self medication hypotheses. Neurosci Biobehav Rev. 2005;29(6):1021-1034.
23. Procyshyn RM, Tse G, Sin O, et al. Concomitant clozapine reduces smoking in patients treated with risperidone. Eur Neuropsychopharmacol. 2002;12(1):77-80.
24. Anthonisen NR, Connett JE, Kiley JP, et al. Effects of smoking intervention and the use of an inhaled anticholinergic bronchodilator on the rate of decline of FEV1. The Lung Health Study. JAMA. 1994;272(19):1497-1505.
25. Pisinger C, Godtfredsen NS. Is there a health benefit of reduced tobacco consumption? A systematic review. Nicotine Tob Res. 2007;9(6):631-646.
26. Fiore MC, Jaén CR, Baker TB, et al. Treating tobacco use and dependence: 2008 update. Clinical Practice Guideline. Rockville, MD: Public Health Service, US Department of Health and Human Services; 2008.
27. Barnes M, Lawford BR, Burton SC, et al. Smoking and schizophrenia: is symptom profile related to smoking and which antipsychotic medication is of benefit in reducing cigarette use? Aust N Z J Psychiatry. 2006;40(6-7):575-580.
28. Screening for chronic obstructive pulmonary disease using spirometry: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2008;148(7):529-534.
29. Agency for Healthcare Research and Quality. Five major steps to intervention (The “5 A’s”). http://www.ahrq.gov/professionals/clinicians-providers/guidelines-recommendations/tobacco/5steps.html. Published 2012. Accessed March 2, 2016.
30. Addington J, el-Guebaly N, Campbell W, et al. Smoking cessation treatment for patients with schizophrenia. Am J Psychiatry. 1998;155(7):974-976.
31. Zarowitz BJ, O’Shea T. Chronic obstructive pulmonary disease: prevalence, characteristics, and pharmacologic treatment in nursing home residents with cognitive impairment. J Manag Care Pharm. 2012;18(8):598-606.
32. Schermer T, Smeenk F, van Weel C. Referral and consultation in asthma and COPD: an exploration of pulmonologists’ views. Neth J Med. 2003;61(3):71-81.

Issue
Current Psychiatry - 15(5)
Issue
Current Psychiatry - 15(5)
Page Number
68-74
Page Number
68-74
Publications
Publications
Topics
Article Type
Display Headline
COPD comorbid with mental illness: What psychiatrists can do
Display Headline
COPD comorbid with mental illness: What psychiatrists can do
Legacy Keywords
COPD, pulmonary, chronic obstructive pulmonary disease, cardiovascular disease, respiratory disease, somatic disorder, somatic disorders
Legacy Keywords
COPD, pulmonary, chronic obstructive pulmonary disease, cardiovascular disease, respiratory disease, somatic disorder, somatic disorders
Sections
Disallow All Ads
Alternative CME
Article PDF Media

Can anti-inflammatory medications improve symptoms and reduce mortality in schizophrenia?

Article Type
Changed
Tue, 12/11/2018 - 15:01
Display Headline
Can anti-inflammatory medications improve symptoms and reduce mortality in schizophrenia?

Consider 3 observations:

  • Evidence is mounting that cytokine abnormalities are present in schizophrenia (Box1-8).
  • Reduced arterial compliance (change in volume divided by change in pressure [ΔV/ΔP] in an artery during the cardiac cycle) is an early marker of cardiovascular disease (CVD) and a robust predictor of mortality, and is associated with cytokine abnormalities.
  • People with schizophrenia experience increased mortality from CVD.

Taken together, the 3 statements hint at a hypothesis: a common inflammatory process involving cytokine imbalance is associated with symptoms of schizophrenia, reduced arterial compliance, and CVD.

Anti-inflammatory therapeutics that target specific cytokines might both decrease psychiatric symptoms and reduce cardiac mortality in people with schizophrenia. In this article, we (1) highlight the potential role of anti-inflammatory medications in reducing both psychiatric symptoms and cardiac mortality in people with schizophrenia and (2) review the pathophysiological basis of this inflammatory commonality and the evidence for its presence in schizophrenia.


The ‘membrane hypothesis’ of schizophrenia

In this hypothesis, a disturbance in the synthesis and structure of membrane phospholipids results in a subsequent disturbance in the function of neuronal membrane proteins, which might be associated with symptoms and mortality in schizophrenia.9-12 The synaptic vesicle protein synaptophysin, a marker for synaptic density, was found to be decreased in postmortem tissue from the gyrus cinguli in 11 patients with schizophrenia, compared with 13 controls.10 Intracellular phospholipases A2 (inPLA2) act as key enzymes in cell membrane repair and remodeling and in neuroplasticity, neurodevelopment, apoptosis, synaptic pruning, neurodegenerative processes, and neuroinflammation.

In a study, people with first-episode schizophrenia (n = 24) who were drug-naïve or off antipsychotic medication were compared with 25 healthy controls using voxel-based morphometry analysis of T1 high-resolution MRI. inPLA2 activity was increased in the patient group compared with controls; the analysis revealed abnormalities of the frontal and medial temporal cortices, hippocampus, and left-middle and superior temporal gyri in first-episode patients.11 In another study, inPLA2 activity was increased in 35 people with first-episode schizophrenia, compared with 22 controls, and was associated with symptom severity and outcome after 12 weeks of antipsychotic treatment.12


Early CVD mortality in schizophrenia

People with schizophrenia have an elevated rate of CVD compared with the general population; in part, this elevation is linked to magnified risk factors for CVD, including obesity, metabolic syndrome, cigarette smoking, and diabetes13-17; furthermore, most antipsychotics can cause or worsen metabolic syndrome.17

CVD is one of the most common causes of death among people with schizophrenia.17,18 Their life expectancy is reported to be 51 to 61 years—20 to 25 years less than what is seen in the general population.19-21


Arterial compliance in schizophrenia

Reduced arterial compliance has been found to be a robust predictor of athero­sclerosis, stroke, and myocardial infarction22-29:

  • In 376 subjects who had routine diagnostic coronary angiography associated with coronary stenosis, arterial compliance was reduced significantly—even after controlling for age, sex, smoking, diabetes, hypertension, hyperlipidemia, and obesity.24

In a cross-sectional study, 63 male U.S. veterans age 18 to 70 who had a psychiatric diagnosis (16 taking quetiapine, 19 taking risperidone, and 28 treated in the past but off antipsychotics for 2 months) had significantly reduced compliance in thigh- and calf-level arteries than male controls (n = 111), adjusting for body mass index and Framingham Risk Score (FRS). Of the 63 patients, 23 had a diagnosis of schizophrenia or schizoaffective disorder.30 (The FRS is an estimate of a person’s 10-year cardiovascular risk, calculated using age, sex, total cholesterol, high-density lipoprotein, smoker or not, systolic blood pressure, and whether taking an antihypertensive or not. Compliance was measured using computerized plethysmography). Although not statistically significant, secondary analyses from this data set (n = 77, including men for whom factors for metabolic syndrome were available) showed that calf-level compliance (1.82 vs 2.06 mL) and thigh-level compliance (3.6 vs 4.26 mL; P = .06) were reduced in subjects with schizophrenia, compared with those who had another psychiatric diagnosis.31

  • In another study, arterial compliance was significantly reduced in 10 subjects with schizophrenia, compared with 10 healthy controls.32
  • Last, reduced total arterial compliance has been shown to be a robust predictor of mortality in older people, compared with reduced local or regional arterial compliance.33


Cytokine abnormalities in arterial compliance

The mechanism by which reduced arterial compliance is associated with cardiovascular pathology is not entirely clear. Arterial compliance is a predictor of cardiovascular disorders independent of hypertension.34 Two studies show that vascular inflammation is associated with reduced arterial compliance.35,36 Reduced arterial compliance is associated with increased angiotensin II activity; increased nicotinamide adenine dinucleotide phosphate oxidase activity; reduced nitric oxide activity; and increased reactive oxygen species.37-39 Angiotensin-II signaling activates transforming growth factor-β, tumor necrosis factor (TNF)-α, interleukin (IL)-1, IL-17, IL-6, and C-reactive protein (CRP)—all of which are associated with reduced arterial compliance.39-46 In addition, high-sensitivity CRP is significantly associated with reduced arterial compliance.47-49

 

 

The overlap of cytokine abnormalities linked to schizophrenia and to arterial compliance is depicted in the Figure.


Anti-inflammatory medications and arterial compliance

Evidence suggests that anti-inflammatory medications increase arterial compliance:

  • In 10 patients who had coronary artery disease or diabetes, or both, simvastatin (40 mg/d) was administered for 4 months. Arterial compliance improved in all 10 after 2 months of treatment and increased by 34% after 4 months.27
  • Evidence also suggests that the use of omega-3 fatty acids was associated with increased arterial compliance in people with dyslipidemia.50
  • Last, in people with rheumatoid arthritis, infliximab, a monoclonal antibody against TNF-Symbol Stdα, reduced aortic inflammation; this effect correlated with an increase in aortic compliance.51


Anti-inflammatory medications in schizophrenia

Two studies have yielded notable findings:

  • A meta-analysis of 5 randomized controlled trials (RCTs) involving 264 subjects, comprising 4 studies of celecoxib and 1 of acetylsalicylic acid, had an effect size of 0.43 on total symptom severity. Investigators argued that acetylsalicylic acid might have the additional benefit of decreasing the risk of cardiac death in schizophrenia.52
  • A review of 26 RCTs examined the efficacy of anti-inflammatory medications on symptom severity in schizophrenia. Acetylsalicylic acid, N-acetylcysteine, and estrogens had an effect size of 0.3, 0.45, and 0.51, respectively.53


Significance of these findings

A revelation that cytokine abnormalities are associated with schizophrenia symptoms and co-occurring somatic illness might offer an important new avenue of therapeutic discovery. On average, people with schizophrenia die 20 to 25 years earlier than the general population; CVD is the major cause of their death. Measuring arterial compliance, a novel noninvasive technology in psychiatry, as well as metabolic parameters, could serve as an early biomarker for assessing risk of CVD.

Implications for psychiatric practice. If inflammation plays a role in CVD in schizophrenia—either independently of factors such as metabolic syndrome, obesity, and smoking, or on the causal pathway linking these factors to reduced arterial compliance and to CVD—treatment with anti-inflammatory medications might reduce the alarming disparity of mortality that accompanies schizophrenia. In short, anti-inflammatory medications may offer a double benefit in this setting. Furthermore, success in this approach could spur clarification of the role of abnormal cytokines in other psychiatric disorders.

At this time, for your patients, consider that anti-inflammatory medications routinely used in medical practice, such as nonsteroidal anti-inflammatory drugs, omega-3 fatty acids, and statins, might alleviate psychiatric symptoms and might reduce cardiovascular mortality in schizophrenia.


Future directions

Perhaps only a limited number of cytokines are common to schizophrenia and reduced arterial compliance. Targeting those specific cytokines might, however, provide the dual benefit in schizophrenia of:

  • alleviating symptoms
  • reducing the rate of CVD-related mortality.

Studies are warranted to determine the value of (1) anti-inflammatory medications, such as N-acetylcysteine and infliximab and (2) anti-inflammatory combination therapy for this dual purpose. In fact, recruitment of subjects is underway for a study, Anti-Inflammatory Combination Therapy for the Treatment of Schizophrenia, at the University of Maryland (ClinicalTrials.gov Identifier: NCT01514682).


Bottom Line

An emerging hypothesis posits a common inflammatory process involving cytokine imbalance that is associated with schizophrenia symptoms, reduced arterial compliance, and cardiovascular disease. Anti-inflammatory medications routinely used in medical practice, such as nonsteroidal anti-inflammatory drugs, omega-3 fatty acids, and statins, might alleviate psychiatric symptoms and reduce cardiac mortality in people with schizophrenia.


Related Resources

  • Müller N, Weidinger E, Leitner B, et al. The role of inflammation in schizophrenia. Front Neurosci. 2015;21(9):372. doi: 10.3389/fnins.2015.00372. eCollection 2015.
  • Xiong GL, Kenedi, CA. Aspirin to prevent cardiovascular events: Weighing risks and benefits. Current Psychiatry. 2010;9(2):55,56,62,63.


Drug Brand Names

Celecoxib • Celebrex
Infliximab • Remicade
Omega-3 fatty acids • Lovaza
Quetiapine • Seroquel
Simvastatin • Zocor
Risperidone • Risperdal


Disclosures

Dr. Koola’s work is underwritten by the American Psychiatric Association’s Kempf Fund Award for Research Development in Psychobiological Psychiatry and by National Institute of Mental Health-funded T32 Grant MH067533-07.

Drs. Raines and Hamilton report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. McMahon is a statistical consultant for Amgen, Inc.

References


1. Potvin S, Stip E, Sepehry AA, et al. Inflammatory cytokine alterations in schizophrenia: a systematic quantitative review. Biol Psychiatry. 2008;63(8):801-808.
2. Miller BJ, Buckley P, Seabolt W, et al. Meta-analysis of cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biol Psychiatry. 2011;70(7):663-671.
3. Frydecka D, Misiak B, Pawlak-Adamska E, et al. Interleukin-6: the missing element of the neurocognitive deterioration in schizophrenia? The focus on genetic underpinnings, cognitive impairment and clinical manifestation. Eur Arch Psychiatry Clin Neurosci. 2015;265(6):449-459.
4. Dickerson F, Stallings C, Origoni A, et al. Additive effects of elevated C-reactive protein and exposure to herpes simplex virus type 1 on cognitive impairment in individuals with schizophrenia. Schizophr Res. 2012;134(1):83-88.
5. Dickerson F, Stallings C, Origoni A, et al. C-reactive protein is associated with the severity of cognitive impairment but not of psychiatric symptoms in individuals with schizophrenia. Schizophr Res. 2007;93(1-3):261-265.
6. Asevedo E, Rizzo LB, Gadelha A, et al. Peripheral interleukin-2 level is associated with negative symptoms and cognitive performance in schizophrenia. Physiol Behav. 2014;129:194-198.
7. Miller BJ, Culpepper N, Rapaport MH. C-reactive protein levels in schizophrenia: a review and meta-analysis. Clin Schizophr Relat Psychoses. 2014;7(4):223-230.
8. Micoulaud-Franchi JA, Faugere M, Boyer L, et al. Elevated C-reactive protein is associated with sensory gating deficit in schizophrenia. Schizophr Res. 2015;165(1):94-96.
9. Horrobin DF. The membrane phospholipid hypothesis as a biochemical basis for the neurodevelopmental concept of schizophrenia. Schizophr Res. 1998;30(3):193-208.
10. Landén M, Davidsson P, Gottfries CG, et al. Reduction of the synaptophysin level but normal levels of glycerophospholipids in the gyrus cinguli in schizophrenia. Schizophr Res. 2002;55(1-2):83-98.
11. Smesny S, Milleit B, Nenadic I, et al. Phospholipase A2 activity is associated with structural brain changes in schizophrenia. Neuroimage. 2010;52(4):1314-1327.
12. Smesny S, Kunstmann C, Kunstmann S, et al. Phospholipase A2 activity in first episode schizophrenia: associations with symptom severity and outcome at week 12. World J Biol Psychiatry. 2011;12(8):598-607.
13. Fontaine KR, Heo M, Harrigan EP, et al. Estimating the consequences of anti-psychotic induced weight gain on health and mortality rate. Psychiatry Res. 2001;101(3):277-288.
14. Homel P, Casey D, Allison DB. Changes in body mass index for individuals with and without schizophrenia, 1987-1996. Schizophr Res. 2002;55(3):277-284.
15. Hedley AA, Ogden CL, Johnson CL, et al. Prevalence of overweight and obesity among US children, adolescents, and adults, 1999-2002. JAMA. 2004;291(23):2847-2850.
16. Dickerson FB, Brown CH, Kreyenbuhl JA, et al. Obesity among individuals with serious mental illness. Acta Psychiatr Scand. 2006;113(4):306-313.
17. Newcomer JW. Metabolic syndrome and mental illness. Am J Manag Care. 2007;13(suppl 7):S170-S177.
18. Healy D, Le Noury J, Harris M, et al. Mortality in schizophrenia and related psychoses: data from two cohorts, 1875-1924 and 1994-2010. BMJ Open. 2012;2(5). doi: 10.1136/bmjopen-2012-001810.
19. Newman SC, Bland RC. Mortality in a cohort of patients with schizophrenia: a record linkage study. Can J Psychiatry. 1991;36(4):239-245.
20. Harris EC, Barraclough B. Excess mortality of mental disorder. Br J Psychiatry. 1998;173:11-53.
21. Colton CW, Manderscheid RW. Congruencies in increased mortality rates, years of potential life lost, and causes of death among public mental health clients in eight states. Prev Chronic Dis. 2006;3(2):A42.
22. Farrar DJ, Bond MG, Riley WA, et al. Anatomic correlates of aortic pulse wave velocity and carotid artery elasticity during atherosclerosis progression and regression in monkeys. Circulation. 1991;83(5):1754-1763.
23. Wada T, Kodaira K, Fujishiro K, et al. Correlation of ultrasound-measured common carotid artery stiffness with pathological findings. Arterioscler Thromb. 1994;14(3):479-482.
24. Herrington DM, Kesler K, Reiber JH, et al. Arterial compliance adds to conventional risk factors for prediction of angiographic coronary artery disease. Am Heart J. 2013;146(4):662-667.
25. Willens HJ, Davis W, Herrington DM, et al. Relationship of peripheral arterial compliance and standard cardiovascular risk factors. Vasc Endovascular Surg. 2003;37(3):197-206.
26. Herrington DM, Brown WV, Mosca L, et al. Relationship between arterial stiffness and subclinical aortic atherosclerosis. Circulation. 2004;110(4):432-437.
27. Saliashvili G, Davis WW, Harris MT, et al. Simvastatin improved arterial compliance in high-risk patients. Vasc Endovascular Surg. 2004;38(6):519-523.
28. Le NA, Brown WV, Davis WW, et al. Comparison of the relation of triglyceride-rich lipoproteins and muscular artery compliance in healthy women versus healthy men. Am J Cardiol. 2005;95(9):1049-1054.
29. Willens HJ, Chirinos JA, Brown WV, et al. Usefulness of arterial compliance in the thigh in predicting exercise capacity in individuals without coronary heart disease. Am J Cardiol. 2005;96(2):306-310.
30. Koola MM, Brown WV, Qualls C, et al. Reduced arterial compliance in patients with psychiatric diagnoses. Schizophr Res. 2012;137(1-3):251-253.
31. Koola MM, Sorkin JD, Fargotstein M, et al. Predictors of calf arterial compliance in male veterans with psychiatric diagnoses. The Primary Care Companion for CNS Disorders. In press.
32. Phillips AA, Warburton DE, Flynn SW, et al. Assessment of arterial stiffness among schizophrenia-spectrum disorders using aortic pulse wave velocity and arterial compliance: a pilot study. Psychiatry Res. 2014;215(1):14-19.
33. Papaioannou TG, Protogerou AD, Stergiopulos N, et al. Total arterial compliance estimated by a novel method and all-cause mortality in the elderly: the PROTEGER study. Age (Dordr). 2014;36(3):9661.
34. Park S, Lakatta EG. Role of inflammation in the pathogenesis of arterial stiffness. Yonsei Med J. 2012;53(2):258-261.
35. Lakatta EG, Levy D. Arterial and cardiac aging: major shareholders in cardiovascular disease enterprises: Part II: the aging heart in health: links to heart disease. Circulation. 2003;107(2):346-354.
36. Zieman SJ, Melenovsky V, Kass DA. Mechanisms, pathophysiology, and therapy of arterial stiffness. Arterioscler Thromb Vasc Biol. 2005;25(5):932-943.
37. van der Loo B, Labugger R, Skepper JN, et al. Enhanced peroxynitrite formation is associated with vascular aging. J Exp Med. 2000;192(12):1731-1744.
38. Csiszar A, Ungvari Z, Edwards JG, et al. Aging-induced phenotypic changes and oxidative stress impair coronary arteriolar function. Circ Res. 2002;90(11):1159-1166.
39. Wang MC, Tsai WC, Chen JY, et al. Arterial stiffness correlated with cardiac remodelling in patients with chronic kidney disease. Nephrology (Carlton). 2007;12(6):591-597.
40. Belmin J, Bernard C, Corman B, et al. Increased production of tumor necrosis factor and interleukin-6 by arterial wall of aged rats. Am J Physiol. 1995;268(6 pt 2):H2288-2293.
41. Gerli R, Monti D, Bistoni O, et al. Chemokines, sTNF-Rs and sCD30 serum levels in healthy aged people and centenarians. Mech Ageing Dev. 2000;121(1-3):37-46.
42. Pasceri V, Willerson JT, Yeh ET. Direct proinflammatory effect of C-reactive protein on human endothelial cells. Circulation. 2000;102(18):2165-2168.
43. Torzewski M, Rist C, Mortensen RF, et al. C-reactive protein in the arterial intima: role of C-reactive protein receptor-dependent monocyte recruitment in atherogenesis. Arterioscler Thromb Vasc Biol. 2000;20(9):2094-2099.
44. Venugopal SK, Devaraj S, Yuhanna I, et al. Demonstration that C-reactive protein decreases eNOS expression and bioactivity in human aortic endothelial cells. Circulation. 2002;106(12):1439-1441.
45. Csiszar A, Ungvari Z, Koller A, et al. Aging-induced proinflammatory shift in cytokine expression profile in coronary arteries. FASEB J. 2003;17(9):1183-1185.
46. Spinetti G, Wang M, Monticone R, et al. Rat aortic MCP-1 and its receptor CCR2 increase with age and alter vascular smooth muscle cell function. Arterioscler Thromb Vasc Biol. 2004;24(8):1397-1402.
47. Mattace-Raso FU, van der Cammen TJ, van der Meer IM, et al. C-reactive protein and arterial stiffness in older adults: the Rotterdam Study. Atherosclerosis. 2004;176(1):111-116.
48. Mahmud A, Feely J. Arterial stiffness is related to systemic inflammation in essential hypertension. Hypertension. 2005;46(5):1118-1122.
49. Nagano M, Nakamura M, Sato K, et al. Association between serum C-reactive protein levels and pulse wave velocity: a population-based cross-sectional study in a general population. Atherosclerosis. 2005;180(1):189-195.
50. Nestel P, Shige H, Pomeroy S, et al. The n-3 fatty acids eicosapentaenoic acid and docosahexaenoic acid increase systemic arterial compliance in humans. Am J Clin Nutr. 2002;76(2):326-330.
51. Mäki-Petäjä KM, Elkhawad M, Cheriyan J, et al. Anti-tumor necrosis factor-α therapy reduces aortic inflammation and stiffness in patients with rheumatoid arthritis. Circulation. 2012;126(21):2473-2480.
52. Sommer IE, de Witte L, Begemann M, et al. Nonsteroidal anti-inflammatory drugs in schizophrenia: ready for practice or a good start? A meta-analysis. J Clin Psychiatry. 2012;73(4):414-419.
53. Sommer IE, van Westrhenen R, Begemann MJ, et al. Efficacy of anti-inflammatory agents to improve symptoms in patients with schizophrenia: an update. Schizophr Bull. 2014;40(1):181-191.

Article PDF
Author and Disclosure Information

Maju Mathew Koola, MD
Sheppard Pratt Health System
Baltimore, Maryland
Associate Professor
Department of Psychiatry and Behavioral Sciences
George Washington University School of Medicine and Health Sciences
Washington, DC

Jeffrey K. Raines, MD, PhD
Emeritus Professor
Department of Surgery
University of Miami
Miami, Florida
Department of Surgery
Harvard Medical School
Cambridge, Massachusetts

Robert G. Hamilton, PhD, D(ABMLI)
Professor
Departments of Medicine and Pathology
Johns Hopkins University School of Medicine
Baltimore, Maryland

Robert P. McMahon, PhD
Professor of Psychiatry
University of Maryland School of Medicine
Baltimore, Maryland

Issue
Current Psychiatry - 15(5)
Publications
Topics
Page Number
52-57
Legacy Keywords
anti-inflammatory medications, schizophrenia, psychosis, psychotic disorder, psychotic disorders, cardiovascular disease, mortality, cytokine
Sections
Author and Disclosure Information

Maju Mathew Koola, MD
Sheppard Pratt Health System
Baltimore, Maryland
Associate Professor
Department of Psychiatry and Behavioral Sciences
George Washington University School of Medicine and Health Sciences
Washington, DC

Jeffrey K. Raines, MD, PhD
Emeritus Professor
Department of Surgery
University of Miami
Miami, Florida
Department of Surgery
Harvard Medical School
Cambridge, Massachusetts

Robert G. Hamilton, PhD, D(ABMLI)
Professor
Departments of Medicine and Pathology
Johns Hopkins University School of Medicine
Baltimore, Maryland

Robert P. McMahon, PhD
Professor of Psychiatry
University of Maryland School of Medicine
Baltimore, Maryland

Author and Disclosure Information

Maju Mathew Koola, MD
Sheppard Pratt Health System
Baltimore, Maryland
Associate Professor
Department of Psychiatry and Behavioral Sciences
George Washington University School of Medicine and Health Sciences
Washington, DC

Jeffrey K. Raines, MD, PhD
Emeritus Professor
Department of Surgery
University of Miami
Miami, Florida
Department of Surgery
Harvard Medical School
Cambridge, Massachusetts

Robert G. Hamilton, PhD, D(ABMLI)
Professor
Departments of Medicine and Pathology
Johns Hopkins University School of Medicine
Baltimore, Maryland

Robert P. McMahon, PhD
Professor of Psychiatry
University of Maryland School of Medicine
Baltimore, Maryland

Article PDF
Article PDF

Consider 3 observations:

  • Evidence is mounting that cytokine abnormalities are present in schizophrenia (Box1-8).
  • Reduced arterial compliance (change in volume divided by change in pressure [ΔV/ΔP] in an artery during the cardiac cycle) is an early marker of cardiovascular disease (CVD) and a robust predictor of mortality, and is associated with cytokine abnormalities.
  • People with schizophrenia experience increased mortality from CVD.

Taken together, the 3 statements hint at a hypothesis: a common inflammatory process involving cytokine imbalance is associated with symptoms of schizophrenia, reduced arterial compliance, and CVD.

Anti-inflammatory therapeutics that target specific cytokines might both decrease psychiatric symptoms and reduce cardiac mortality in people with schizophrenia. In this article, we (1) highlight the potential role of anti-inflammatory medications in reducing both psychiatric symptoms and cardiac mortality in people with schizophrenia and (2) review the pathophysiological basis of this inflammatory commonality and the evidence for its presence in schizophrenia.


The ‘membrane hypothesis’ of schizophrenia

In this hypothesis, a disturbance in the synthesis and structure of membrane phospholipids results in a subsequent disturbance in the function of neuronal membrane proteins, which might be associated with symptoms and mortality in schizophrenia.9-12 The synaptic vesicle protein synaptophysin, a marker for synaptic density, was found to be decreased in postmortem tissue from the gyrus cinguli in 11 patients with schizophrenia, compared with 13 controls.10 Intracellular phospholipases A2 (inPLA2) act as key enzymes in cell membrane repair and remodeling and in neuroplasticity, neurodevelopment, apoptosis, synaptic pruning, neurodegenerative processes, and neuroinflammation.

In a study, people with first-episode schizophrenia (n = 24) who were drug-naïve or off antipsychotic medication were compared with 25 healthy controls using voxel-based morphometry analysis of T1 high-resolution MRI. inPLA2 activity was increased in the patient group compared with controls; the analysis revealed abnormalities of the frontal and medial temporal cortices, hippocampus, and left-middle and superior temporal gyri in first-episode patients.11 In another study, inPLA2 activity was increased in 35 people with first-episode schizophrenia, compared with 22 controls, and was associated with symptom severity and outcome after 12 weeks of antipsychotic treatment.12


Early CVD mortality in schizophrenia

People with schizophrenia have an elevated rate of CVD compared with the general population; in part, this elevation is linked to magnified risk factors for CVD, including obesity, metabolic syndrome, cigarette smoking, and diabetes13-17; furthermore, most antipsychotics can cause or worsen metabolic syndrome.17

CVD is one of the most common causes of death among people with schizophrenia.17,18 Their life expectancy is reported to be 51 to 61 years—20 to 25 years less than what is seen in the general population.19-21


Arterial compliance in schizophrenia

Reduced arterial compliance has been found to be a robust predictor of athero­sclerosis, stroke, and myocardial infarction22-29:

  • In 376 subjects who had routine diagnostic coronary angiography associated with coronary stenosis, arterial compliance was reduced significantly—even after controlling for age, sex, smoking, diabetes, hypertension, hyperlipidemia, and obesity.24

In a cross-sectional study, 63 male U.S. veterans age 18 to 70 who had a psychiatric diagnosis (16 taking quetiapine, 19 taking risperidone, and 28 treated in the past but off antipsychotics for 2 months) had significantly reduced compliance in thigh- and calf-level arteries than male controls (n = 111), adjusting for body mass index and Framingham Risk Score (FRS). Of the 63 patients, 23 had a diagnosis of schizophrenia or schizoaffective disorder.30 (The FRS is an estimate of a person’s 10-year cardiovascular risk, calculated using age, sex, total cholesterol, high-density lipoprotein, smoker or not, systolic blood pressure, and whether taking an antihypertensive or not. Compliance was measured using computerized plethysmography). Although not statistically significant, secondary analyses from this data set (n = 77, including men for whom factors for metabolic syndrome were available) showed that calf-level compliance (1.82 vs 2.06 mL) and thigh-level compliance (3.6 vs 4.26 mL; P = .06) were reduced in subjects with schizophrenia, compared with those who had another psychiatric diagnosis.31

  • In another study, arterial compliance was significantly reduced in 10 subjects with schizophrenia, compared with 10 healthy controls.32
  • Last, reduced total arterial compliance has been shown to be a robust predictor of mortality in older people, compared with reduced local or regional arterial compliance.33


Cytokine abnormalities in arterial compliance

The mechanism by which reduced arterial compliance is associated with cardiovascular pathology is not entirely clear. Arterial compliance is a predictor of cardiovascular disorders independent of hypertension.34 Two studies show that vascular inflammation is associated with reduced arterial compliance.35,36 Reduced arterial compliance is associated with increased angiotensin II activity; increased nicotinamide adenine dinucleotide phosphate oxidase activity; reduced nitric oxide activity; and increased reactive oxygen species.37-39 Angiotensin-II signaling activates transforming growth factor-β, tumor necrosis factor (TNF)-α, interleukin (IL)-1, IL-17, IL-6, and C-reactive protein (CRP)—all of which are associated with reduced arterial compliance.39-46 In addition, high-sensitivity CRP is significantly associated with reduced arterial compliance.47-49

 

 

The overlap of cytokine abnormalities linked to schizophrenia and to arterial compliance is depicted in the Figure.


Anti-inflammatory medications and arterial compliance

Evidence suggests that anti-inflammatory medications increase arterial compliance:

  • In 10 patients who had coronary artery disease or diabetes, or both, simvastatin (40 mg/d) was administered for 4 months. Arterial compliance improved in all 10 after 2 months of treatment and increased by 34% after 4 months.27
  • Evidence also suggests that the use of omega-3 fatty acids was associated with increased arterial compliance in people with dyslipidemia.50
  • Last, in people with rheumatoid arthritis, infliximab, a monoclonal antibody against TNF-Symbol Stdα, reduced aortic inflammation; this effect correlated with an increase in aortic compliance.51


Anti-inflammatory medications in schizophrenia

Two studies have yielded notable findings:

  • A meta-analysis of 5 randomized controlled trials (RCTs) involving 264 subjects, comprising 4 studies of celecoxib and 1 of acetylsalicylic acid, had an effect size of 0.43 on total symptom severity. Investigators argued that acetylsalicylic acid might have the additional benefit of decreasing the risk of cardiac death in schizophrenia.52
  • A review of 26 RCTs examined the efficacy of anti-inflammatory medications on symptom severity in schizophrenia. Acetylsalicylic acid, N-acetylcysteine, and estrogens had an effect size of 0.3, 0.45, and 0.51, respectively.53


Significance of these findings

A revelation that cytokine abnormalities are associated with schizophrenia symptoms and co-occurring somatic illness might offer an important new avenue of therapeutic discovery. On average, people with schizophrenia die 20 to 25 years earlier than the general population; CVD is the major cause of their death. Measuring arterial compliance, a novel noninvasive technology in psychiatry, as well as metabolic parameters, could serve as an early biomarker for assessing risk of CVD.

Implications for psychiatric practice. If inflammation plays a role in CVD in schizophrenia—either independently of factors such as metabolic syndrome, obesity, and smoking, or on the causal pathway linking these factors to reduced arterial compliance and to CVD—treatment with anti-inflammatory medications might reduce the alarming disparity of mortality that accompanies schizophrenia. In short, anti-inflammatory medications may offer a double benefit in this setting. Furthermore, success in this approach could spur clarification of the role of abnormal cytokines in other psychiatric disorders.

At this time, for your patients, consider that anti-inflammatory medications routinely used in medical practice, such as nonsteroidal anti-inflammatory drugs, omega-3 fatty acids, and statins, might alleviate psychiatric symptoms and might reduce cardiovascular mortality in schizophrenia.


Future directions

Perhaps only a limited number of cytokines are common to schizophrenia and reduced arterial compliance. Targeting those specific cytokines might, however, provide the dual benefit in schizophrenia of:

  • alleviating symptoms
  • reducing the rate of CVD-related mortality.

Studies are warranted to determine the value of (1) anti-inflammatory medications, such as N-acetylcysteine and infliximab and (2) anti-inflammatory combination therapy for this dual purpose. In fact, recruitment of subjects is underway for a study, Anti-Inflammatory Combination Therapy for the Treatment of Schizophrenia, at the University of Maryland (ClinicalTrials.gov Identifier: NCT01514682).


Bottom Line

An emerging hypothesis posits a common inflammatory process involving cytokine imbalance that is associated with schizophrenia symptoms, reduced arterial compliance, and cardiovascular disease. Anti-inflammatory medications routinely used in medical practice, such as nonsteroidal anti-inflammatory drugs, omega-3 fatty acids, and statins, might alleviate psychiatric symptoms and reduce cardiac mortality in people with schizophrenia.


Related Resources

  • Müller N, Weidinger E, Leitner B, et al. The role of inflammation in schizophrenia. Front Neurosci. 2015;21(9):372. doi: 10.3389/fnins.2015.00372. eCollection 2015.
  • Xiong GL, Kenedi, CA. Aspirin to prevent cardiovascular events: Weighing risks and benefits. Current Psychiatry. 2010;9(2):55,56,62,63.


Drug Brand Names

Celecoxib • Celebrex
Infliximab • Remicade
Omega-3 fatty acids • Lovaza
Quetiapine • Seroquel
Simvastatin • Zocor
Risperidone • Risperdal


Disclosures

Dr. Koola’s work is underwritten by the American Psychiatric Association’s Kempf Fund Award for Research Development in Psychobiological Psychiatry and by National Institute of Mental Health-funded T32 Grant MH067533-07.

Drs. Raines and Hamilton report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. McMahon is a statistical consultant for Amgen, Inc.

Consider 3 observations:

  • Evidence is mounting that cytokine abnormalities are present in schizophrenia (Box1-8).
  • Reduced arterial compliance (change in volume divided by change in pressure [ΔV/ΔP] in an artery during the cardiac cycle) is an early marker of cardiovascular disease (CVD) and a robust predictor of mortality, and is associated with cytokine abnormalities.
  • People with schizophrenia experience increased mortality from CVD.

Taken together, the 3 statements hint at a hypothesis: a common inflammatory process involving cytokine imbalance is associated with symptoms of schizophrenia, reduced arterial compliance, and CVD.

Anti-inflammatory therapeutics that target specific cytokines might both decrease psychiatric symptoms and reduce cardiac mortality in people with schizophrenia. In this article, we (1) highlight the potential role of anti-inflammatory medications in reducing both psychiatric symptoms and cardiac mortality in people with schizophrenia and (2) review the pathophysiological basis of this inflammatory commonality and the evidence for its presence in schizophrenia.


The ‘membrane hypothesis’ of schizophrenia

In this hypothesis, a disturbance in the synthesis and structure of membrane phospholipids results in a subsequent disturbance in the function of neuronal membrane proteins, which might be associated with symptoms and mortality in schizophrenia.9-12 The synaptic vesicle protein synaptophysin, a marker for synaptic density, was found to be decreased in postmortem tissue from the gyrus cinguli in 11 patients with schizophrenia, compared with 13 controls.10 Intracellular phospholipases A2 (inPLA2) act as key enzymes in cell membrane repair and remodeling and in neuroplasticity, neurodevelopment, apoptosis, synaptic pruning, neurodegenerative processes, and neuroinflammation.

In a study, people with first-episode schizophrenia (n = 24) who were drug-naïve or off antipsychotic medication were compared with 25 healthy controls using voxel-based morphometry analysis of T1 high-resolution MRI. inPLA2 activity was increased in the patient group compared with controls; the analysis revealed abnormalities of the frontal and medial temporal cortices, hippocampus, and left-middle and superior temporal gyri in first-episode patients.11 In another study, inPLA2 activity was increased in 35 people with first-episode schizophrenia, compared with 22 controls, and was associated with symptom severity and outcome after 12 weeks of antipsychotic treatment.12


Early CVD mortality in schizophrenia

People with schizophrenia have an elevated rate of CVD compared with the general population; in part, this elevation is linked to magnified risk factors for CVD, including obesity, metabolic syndrome, cigarette smoking, and diabetes13-17; furthermore, most antipsychotics can cause or worsen metabolic syndrome.17

CVD is one of the most common causes of death among people with schizophrenia.17,18 Their life expectancy is reported to be 51 to 61 years—20 to 25 years less than what is seen in the general population.19-21


Arterial compliance in schizophrenia

Reduced arterial compliance has been found to be a robust predictor of athero­sclerosis, stroke, and myocardial infarction22-29:

  • In 376 subjects who had routine diagnostic coronary angiography associated with coronary stenosis, arterial compliance was reduced significantly—even after controlling for age, sex, smoking, diabetes, hypertension, hyperlipidemia, and obesity.24

In a cross-sectional study, 63 male U.S. veterans age 18 to 70 who had a psychiatric diagnosis (16 taking quetiapine, 19 taking risperidone, and 28 treated in the past but off antipsychotics for 2 months) had significantly reduced compliance in thigh- and calf-level arteries than male controls (n = 111), adjusting for body mass index and Framingham Risk Score (FRS). Of the 63 patients, 23 had a diagnosis of schizophrenia or schizoaffective disorder.30 (The FRS is an estimate of a person’s 10-year cardiovascular risk, calculated using age, sex, total cholesterol, high-density lipoprotein, smoker or not, systolic blood pressure, and whether taking an antihypertensive or not. Compliance was measured using computerized plethysmography). Although not statistically significant, secondary analyses from this data set (n = 77, including men for whom factors for metabolic syndrome were available) showed that calf-level compliance (1.82 vs 2.06 mL) and thigh-level compliance (3.6 vs 4.26 mL; P = .06) were reduced in subjects with schizophrenia, compared with those who had another psychiatric diagnosis.31

  • In another study, arterial compliance was significantly reduced in 10 subjects with schizophrenia, compared with 10 healthy controls.32
  • Last, reduced total arterial compliance has been shown to be a robust predictor of mortality in older people, compared with reduced local or regional arterial compliance.33


Cytokine abnormalities in arterial compliance

The mechanism by which reduced arterial compliance is associated with cardiovascular pathology is not entirely clear. Arterial compliance is a predictor of cardiovascular disorders independent of hypertension.34 Two studies show that vascular inflammation is associated with reduced arterial compliance.35,36 Reduced arterial compliance is associated with increased angiotensin II activity; increased nicotinamide adenine dinucleotide phosphate oxidase activity; reduced nitric oxide activity; and increased reactive oxygen species.37-39 Angiotensin-II signaling activates transforming growth factor-β, tumor necrosis factor (TNF)-α, interleukin (IL)-1, IL-17, IL-6, and C-reactive protein (CRP)—all of which are associated with reduced arterial compliance.39-46 In addition, high-sensitivity CRP is significantly associated with reduced arterial compliance.47-49

 

 

The overlap of cytokine abnormalities linked to schizophrenia and to arterial compliance is depicted in the Figure.


Anti-inflammatory medications and arterial compliance

Evidence suggests that anti-inflammatory medications increase arterial compliance:

  • In 10 patients who had coronary artery disease or diabetes, or both, simvastatin (40 mg/d) was administered for 4 months. Arterial compliance improved in all 10 after 2 months of treatment and increased by 34% after 4 months.27
  • Evidence also suggests that the use of omega-3 fatty acids was associated with increased arterial compliance in people with dyslipidemia.50
  • Last, in people with rheumatoid arthritis, infliximab, a monoclonal antibody against TNF-Symbol Stdα, reduced aortic inflammation; this effect correlated with an increase in aortic compliance.51


Anti-inflammatory medications in schizophrenia

Two studies have yielded notable findings:

  • A meta-analysis of 5 randomized controlled trials (RCTs) involving 264 subjects, comprising 4 studies of celecoxib and 1 of acetylsalicylic acid, had an effect size of 0.43 on total symptom severity. Investigators argued that acetylsalicylic acid might have the additional benefit of decreasing the risk of cardiac death in schizophrenia.52
  • A review of 26 RCTs examined the efficacy of anti-inflammatory medications on symptom severity in schizophrenia. Acetylsalicylic acid, N-acetylcysteine, and estrogens had an effect size of 0.3, 0.45, and 0.51, respectively.53


Significance of these findings

A revelation that cytokine abnormalities are associated with schizophrenia symptoms and co-occurring somatic illness might offer an important new avenue of therapeutic discovery. On average, people with schizophrenia die 20 to 25 years earlier than the general population; CVD is the major cause of their death. Measuring arterial compliance, a novel noninvasive technology in psychiatry, as well as metabolic parameters, could serve as an early biomarker for assessing risk of CVD.

Implications for psychiatric practice. If inflammation plays a role in CVD in schizophrenia—either independently of factors such as metabolic syndrome, obesity, and smoking, or on the causal pathway linking these factors to reduced arterial compliance and to CVD—treatment with anti-inflammatory medications might reduce the alarming disparity of mortality that accompanies schizophrenia. In short, anti-inflammatory medications may offer a double benefit in this setting. Furthermore, success in this approach could spur clarification of the role of abnormal cytokines in other psychiatric disorders.

At this time, for your patients, consider that anti-inflammatory medications routinely used in medical practice, such as nonsteroidal anti-inflammatory drugs, omega-3 fatty acids, and statins, might alleviate psychiatric symptoms and might reduce cardiovascular mortality in schizophrenia.


Future directions

Perhaps only a limited number of cytokines are common to schizophrenia and reduced arterial compliance. Targeting those specific cytokines might, however, provide the dual benefit in schizophrenia of:

  • alleviating symptoms
  • reducing the rate of CVD-related mortality.

Studies are warranted to determine the value of (1) anti-inflammatory medications, such as N-acetylcysteine and infliximab and (2) anti-inflammatory combination therapy for this dual purpose. In fact, recruitment of subjects is underway for a study, Anti-Inflammatory Combination Therapy for the Treatment of Schizophrenia, at the University of Maryland (ClinicalTrials.gov Identifier: NCT01514682).


Bottom Line

An emerging hypothesis posits a common inflammatory process involving cytokine imbalance that is associated with schizophrenia symptoms, reduced arterial compliance, and cardiovascular disease. Anti-inflammatory medications routinely used in medical practice, such as nonsteroidal anti-inflammatory drugs, omega-3 fatty acids, and statins, might alleviate psychiatric symptoms and reduce cardiac mortality in people with schizophrenia.


Related Resources

  • Müller N, Weidinger E, Leitner B, et al. The role of inflammation in schizophrenia. Front Neurosci. 2015;21(9):372. doi: 10.3389/fnins.2015.00372. eCollection 2015.
  • Xiong GL, Kenedi, CA. Aspirin to prevent cardiovascular events: Weighing risks and benefits. Current Psychiatry. 2010;9(2):55,56,62,63.


Drug Brand Names

Celecoxib • Celebrex
Infliximab • Remicade
Omega-3 fatty acids • Lovaza
Quetiapine • Seroquel
Simvastatin • Zocor
Risperidone • Risperdal


Disclosures

Dr. Koola’s work is underwritten by the American Psychiatric Association’s Kempf Fund Award for Research Development in Psychobiological Psychiatry and by National Institute of Mental Health-funded T32 Grant MH067533-07.

Drs. Raines and Hamilton report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

Dr. McMahon is a statistical consultant for Amgen, Inc.

References


1. Potvin S, Stip E, Sepehry AA, et al. Inflammatory cytokine alterations in schizophrenia: a systematic quantitative review. Biol Psychiatry. 2008;63(8):801-808.
2. Miller BJ, Buckley P, Seabolt W, et al. Meta-analysis of cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biol Psychiatry. 2011;70(7):663-671.
3. Frydecka D, Misiak B, Pawlak-Adamska E, et al. Interleukin-6: the missing element of the neurocognitive deterioration in schizophrenia? The focus on genetic underpinnings, cognitive impairment and clinical manifestation. Eur Arch Psychiatry Clin Neurosci. 2015;265(6):449-459.
4. Dickerson F, Stallings C, Origoni A, et al. Additive effects of elevated C-reactive protein and exposure to herpes simplex virus type 1 on cognitive impairment in individuals with schizophrenia. Schizophr Res. 2012;134(1):83-88.
5. Dickerson F, Stallings C, Origoni A, et al. C-reactive protein is associated with the severity of cognitive impairment but not of psychiatric symptoms in individuals with schizophrenia. Schizophr Res. 2007;93(1-3):261-265.
6. Asevedo E, Rizzo LB, Gadelha A, et al. Peripheral interleukin-2 level is associated with negative symptoms and cognitive performance in schizophrenia. Physiol Behav. 2014;129:194-198.
7. Miller BJ, Culpepper N, Rapaport MH. C-reactive protein levels in schizophrenia: a review and meta-analysis. Clin Schizophr Relat Psychoses. 2014;7(4):223-230.
8. Micoulaud-Franchi JA, Faugere M, Boyer L, et al. Elevated C-reactive protein is associated with sensory gating deficit in schizophrenia. Schizophr Res. 2015;165(1):94-96.
9. Horrobin DF. The membrane phospholipid hypothesis as a biochemical basis for the neurodevelopmental concept of schizophrenia. Schizophr Res. 1998;30(3):193-208.
10. Landén M, Davidsson P, Gottfries CG, et al. Reduction of the synaptophysin level but normal levels of glycerophospholipids in the gyrus cinguli in schizophrenia. Schizophr Res. 2002;55(1-2):83-98.
11. Smesny S, Milleit B, Nenadic I, et al. Phospholipase A2 activity is associated with structural brain changes in schizophrenia. Neuroimage. 2010;52(4):1314-1327.
12. Smesny S, Kunstmann C, Kunstmann S, et al. Phospholipase A2 activity in first episode schizophrenia: associations with symptom severity and outcome at week 12. World J Biol Psychiatry. 2011;12(8):598-607.
13. Fontaine KR, Heo M, Harrigan EP, et al. Estimating the consequences of anti-psychotic induced weight gain on health and mortality rate. Psychiatry Res. 2001;101(3):277-288.
14. Homel P, Casey D, Allison DB. Changes in body mass index for individuals with and without schizophrenia, 1987-1996. Schizophr Res. 2002;55(3):277-284.
15. Hedley AA, Ogden CL, Johnson CL, et al. Prevalence of overweight and obesity among US children, adolescents, and adults, 1999-2002. JAMA. 2004;291(23):2847-2850.
16. Dickerson FB, Brown CH, Kreyenbuhl JA, et al. Obesity among individuals with serious mental illness. Acta Psychiatr Scand. 2006;113(4):306-313.
17. Newcomer JW. Metabolic syndrome and mental illness. Am J Manag Care. 2007;13(suppl 7):S170-S177.
18. Healy D, Le Noury J, Harris M, et al. Mortality in schizophrenia and related psychoses: data from two cohorts, 1875-1924 and 1994-2010. BMJ Open. 2012;2(5). doi: 10.1136/bmjopen-2012-001810.
19. Newman SC, Bland RC. Mortality in a cohort of patients with schizophrenia: a record linkage study. Can J Psychiatry. 1991;36(4):239-245.
20. Harris EC, Barraclough B. Excess mortality of mental disorder. Br J Psychiatry. 1998;173:11-53.
21. Colton CW, Manderscheid RW. Congruencies in increased mortality rates, years of potential life lost, and causes of death among public mental health clients in eight states. Prev Chronic Dis. 2006;3(2):A42.
22. Farrar DJ, Bond MG, Riley WA, et al. Anatomic correlates of aortic pulse wave velocity and carotid artery elasticity during atherosclerosis progression and regression in monkeys. Circulation. 1991;83(5):1754-1763.
23. Wada T, Kodaira K, Fujishiro K, et al. Correlation of ultrasound-measured common carotid artery stiffness with pathological findings. Arterioscler Thromb. 1994;14(3):479-482.
24. Herrington DM, Kesler K, Reiber JH, et al. Arterial compliance adds to conventional risk factors for prediction of angiographic coronary artery disease. Am Heart J. 2013;146(4):662-667.
25. Willens HJ, Davis W, Herrington DM, et al. Relationship of peripheral arterial compliance and standard cardiovascular risk factors. Vasc Endovascular Surg. 2003;37(3):197-206.
26. Herrington DM, Brown WV, Mosca L, et al. Relationship between arterial stiffness and subclinical aortic atherosclerosis. Circulation. 2004;110(4):432-437.
27. Saliashvili G, Davis WW, Harris MT, et al. Simvastatin improved arterial compliance in high-risk patients. Vasc Endovascular Surg. 2004;38(6):519-523.
28. Le NA, Brown WV, Davis WW, et al. Comparison of the relation of triglyceride-rich lipoproteins and muscular artery compliance in healthy women versus healthy men. Am J Cardiol. 2005;95(9):1049-1054.
29. Willens HJ, Chirinos JA, Brown WV, et al. Usefulness of arterial compliance in the thigh in predicting exercise capacity in individuals without coronary heart disease. Am J Cardiol. 2005;96(2):306-310.
30. Koola MM, Brown WV, Qualls C, et al. Reduced arterial compliance in patients with psychiatric diagnoses. Schizophr Res. 2012;137(1-3):251-253.
31. Koola MM, Sorkin JD, Fargotstein M, et al. Predictors of calf arterial compliance in male veterans with psychiatric diagnoses. The Primary Care Companion for CNS Disorders. In press.
32. Phillips AA, Warburton DE, Flynn SW, et al. Assessment of arterial stiffness among schizophrenia-spectrum disorders using aortic pulse wave velocity and arterial compliance: a pilot study. Psychiatry Res. 2014;215(1):14-19.
33. Papaioannou TG, Protogerou AD, Stergiopulos N, et al. Total arterial compliance estimated by a novel method and all-cause mortality in the elderly: the PROTEGER study. Age (Dordr). 2014;36(3):9661.
34. Park S, Lakatta EG. Role of inflammation in the pathogenesis of arterial stiffness. Yonsei Med J. 2012;53(2):258-261.
35. Lakatta EG, Levy D. Arterial and cardiac aging: major shareholders in cardiovascular disease enterprises: Part II: the aging heart in health: links to heart disease. Circulation. 2003;107(2):346-354.
36. Zieman SJ, Melenovsky V, Kass DA. Mechanisms, pathophysiology, and therapy of arterial stiffness. Arterioscler Thromb Vasc Biol. 2005;25(5):932-943.
37. van der Loo B, Labugger R, Skepper JN, et al. Enhanced peroxynitrite formation is associated with vascular aging. J Exp Med. 2000;192(12):1731-1744.
38. Csiszar A, Ungvari Z, Edwards JG, et al. Aging-induced phenotypic changes and oxidative stress impair coronary arteriolar function. Circ Res. 2002;90(11):1159-1166.
39. Wang MC, Tsai WC, Chen JY, et al. Arterial stiffness correlated with cardiac remodelling in patients with chronic kidney disease. Nephrology (Carlton). 2007;12(6):591-597.
40. Belmin J, Bernard C, Corman B, et al. Increased production of tumor necrosis factor and interleukin-6 by arterial wall of aged rats. Am J Physiol. 1995;268(6 pt 2):H2288-2293.
41. Gerli R, Monti D, Bistoni O, et al. Chemokines, sTNF-Rs and sCD30 serum levels in healthy aged people and centenarians. Mech Ageing Dev. 2000;121(1-3):37-46.
42. Pasceri V, Willerson JT, Yeh ET. Direct proinflammatory effect of C-reactive protein on human endothelial cells. Circulation. 2000;102(18):2165-2168.
43. Torzewski M, Rist C, Mortensen RF, et al. C-reactive protein in the arterial intima: role of C-reactive protein receptor-dependent monocyte recruitment in atherogenesis. Arterioscler Thromb Vasc Biol. 2000;20(9):2094-2099.
44. Venugopal SK, Devaraj S, Yuhanna I, et al. Demonstration that C-reactive protein decreases eNOS expression and bioactivity in human aortic endothelial cells. Circulation. 2002;106(12):1439-1441.
45. Csiszar A, Ungvari Z, Koller A, et al. Aging-induced proinflammatory shift in cytokine expression profile in coronary arteries. FASEB J. 2003;17(9):1183-1185.
46. Spinetti G, Wang M, Monticone R, et al. Rat aortic MCP-1 and its receptor CCR2 increase with age and alter vascular smooth muscle cell function. Arterioscler Thromb Vasc Biol. 2004;24(8):1397-1402.
47. Mattace-Raso FU, van der Cammen TJ, van der Meer IM, et al. C-reactive protein and arterial stiffness in older adults: the Rotterdam Study. Atherosclerosis. 2004;176(1):111-116.
48. Mahmud A, Feely J. Arterial stiffness is related to systemic inflammation in essential hypertension. Hypertension. 2005;46(5):1118-1122.
49. Nagano M, Nakamura M, Sato K, et al. Association between serum C-reactive protein levels and pulse wave velocity: a population-based cross-sectional study in a general population. Atherosclerosis. 2005;180(1):189-195.
50. Nestel P, Shige H, Pomeroy S, et al. The n-3 fatty acids eicosapentaenoic acid and docosahexaenoic acid increase systemic arterial compliance in humans. Am J Clin Nutr. 2002;76(2):326-330.
51. Mäki-Petäjä KM, Elkhawad M, Cheriyan J, et al. Anti-tumor necrosis factor-α therapy reduces aortic inflammation and stiffness in patients with rheumatoid arthritis. Circulation. 2012;126(21):2473-2480.
52. Sommer IE, de Witte L, Begemann M, et al. Nonsteroidal anti-inflammatory drugs in schizophrenia: ready for practice or a good start? A meta-analysis. J Clin Psychiatry. 2012;73(4):414-419.
53. Sommer IE, van Westrhenen R, Begemann MJ, et al. Efficacy of anti-inflammatory agents to improve symptoms in patients with schizophrenia: an update. Schizophr Bull. 2014;40(1):181-191.

References


1. Potvin S, Stip E, Sepehry AA, et al. Inflammatory cytokine alterations in schizophrenia: a systematic quantitative review. Biol Psychiatry. 2008;63(8):801-808.
2. Miller BJ, Buckley P, Seabolt W, et al. Meta-analysis of cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biol Psychiatry. 2011;70(7):663-671.
3. Frydecka D, Misiak B, Pawlak-Adamska E, et al. Interleukin-6: the missing element of the neurocognitive deterioration in schizophrenia? The focus on genetic underpinnings, cognitive impairment and clinical manifestation. Eur Arch Psychiatry Clin Neurosci. 2015;265(6):449-459.
4. Dickerson F, Stallings C, Origoni A, et al. Additive effects of elevated C-reactive protein and exposure to herpes simplex virus type 1 on cognitive impairment in individuals with schizophrenia. Schizophr Res. 2012;134(1):83-88.
5. Dickerson F, Stallings C, Origoni A, et al. C-reactive protein is associated with the severity of cognitive impairment but not of psychiatric symptoms in individuals with schizophrenia. Schizophr Res. 2007;93(1-3):261-265.
6. Asevedo E, Rizzo LB, Gadelha A, et al. Peripheral interleukin-2 level is associated with negative symptoms and cognitive performance in schizophrenia. Physiol Behav. 2014;129:194-198.
7. Miller BJ, Culpepper N, Rapaport MH. C-reactive protein levels in schizophrenia: a review and meta-analysis. Clin Schizophr Relat Psychoses. 2014;7(4):223-230.
8. Micoulaud-Franchi JA, Faugere M, Boyer L, et al. Elevated C-reactive protein is associated with sensory gating deficit in schizophrenia. Schizophr Res. 2015;165(1):94-96.
9. Horrobin DF. The membrane phospholipid hypothesis as a biochemical basis for the neurodevelopmental concept of schizophrenia. Schizophr Res. 1998;30(3):193-208.
10. Landén M, Davidsson P, Gottfries CG, et al. Reduction of the synaptophysin level but normal levels of glycerophospholipids in the gyrus cinguli in schizophrenia. Schizophr Res. 2002;55(1-2):83-98.
11. Smesny S, Milleit B, Nenadic I, et al. Phospholipase A2 activity is associated with structural brain changes in schizophrenia. Neuroimage. 2010;52(4):1314-1327.
12. Smesny S, Kunstmann C, Kunstmann S, et al. Phospholipase A2 activity in first episode schizophrenia: associations with symptom severity and outcome at week 12. World J Biol Psychiatry. 2011;12(8):598-607.
13. Fontaine KR, Heo M, Harrigan EP, et al. Estimating the consequences of anti-psychotic induced weight gain on health and mortality rate. Psychiatry Res. 2001;101(3):277-288.
14. Homel P, Casey D, Allison DB. Changes in body mass index for individuals with and without schizophrenia, 1987-1996. Schizophr Res. 2002;55(3):277-284.
15. Hedley AA, Ogden CL, Johnson CL, et al. Prevalence of overweight and obesity among US children, adolescents, and adults, 1999-2002. JAMA. 2004;291(23):2847-2850.
16. Dickerson FB, Brown CH, Kreyenbuhl JA, et al. Obesity among individuals with serious mental illness. Acta Psychiatr Scand. 2006;113(4):306-313.
17. Newcomer JW. Metabolic syndrome and mental illness. Am J Manag Care. 2007;13(suppl 7):S170-S177.
18. Healy D, Le Noury J, Harris M, et al. Mortality in schizophrenia and related psychoses: data from two cohorts, 1875-1924 and 1994-2010. BMJ Open. 2012;2(5). doi: 10.1136/bmjopen-2012-001810.
19. Newman SC, Bland RC. Mortality in a cohort of patients with schizophrenia: a record linkage study. Can J Psychiatry. 1991;36(4):239-245.
20. Harris EC, Barraclough B. Excess mortality of mental disorder. Br J Psychiatry. 1998;173:11-53.
21. Colton CW, Manderscheid RW. Congruencies in increased mortality rates, years of potential life lost, and causes of death among public mental health clients in eight states. Prev Chronic Dis. 2006;3(2):A42.
22. Farrar DJ, Bond MG, Riley WA, et al. Anatomic correlates of aortic pulse wave velocity and carotid artery elasticity during atherosclerosis progression and regression in monkeys. Circulation. 1991;83(5):1754-1763.
23. Wada T, Kodaira K, Fujishiro K, et al. Correlation of ultrasound-measured common carotid artery stiffness with pathological findings. Arterioscler Thromb. 1994;14(3):479-482.
24. Herrington DM, Kesler K, Reiber JH, et al. Arterial compliance adds to conventional risk factors for prediction of angiographic coronary artery disease. Am Heart J. 2013;146(4):662-667.
25. Willens HJ, Davis W, Herrington DM, et al. Relationship of peripheral arterial compliance and standard cardiovascular risk factors. Vasc Endovascular Surg. 2003;37(3):197-206.
26. Herrington DM, Brown WV, Mosca L, et al. Relationship between arterial stiffness and subclinical aortic atherosclerosis. Circulation. 2004;110(4):432-437.
27. Saliashvili G, Davis WW, Harris MT, et al. Simvastatin improved arterial compliance in high-risk patients. Vasc Endovascular Surg. 2004;38(6):519-523.
28. Le NA, Brown WV, Davis WW, et al. Comparison of the relation of triglyceride-rich lipoproteins and muscular artery compliance in healthy women versus healthy men. Am J Cardiol. 2005;95(9):1049-1054.
29. Willens HJ, Chirinos JA, Brown WV, et al. Usefulness of arterial compliance in the thigh in predicting exercise capacity in individuals without coronary heart disease. Am J Cardiol. 2005;96(2):306-310.
30. Koola MM, Brown WV, Qualls C, et al. Reduced arterial compliance in patients with psychiatric diagnoses. Schizophr Res. 2012;137(1-3):251-253.
31. Koola MM, Sorkin JD, Fargotstein M, et al. Predictors of calf arterial compliance in male veterans with psychiatric diagnoses. The Primary Care Companion for CNS Disorders. In press.
32. Phillips AA, Warburton DE, Flynn SW, et al. Assessment of arterial stiffness among schizophrenia-spectrum disorders using aortic pulse wave velocity and arterial compliance: a pilot study. Psychiatry Res. 2014;215(1):14-19.
33. Papaioannou TG, Protogerou AD, Stergiopulos N, et al. Total arterial compliance estimated by a novel method and all-cause mortality in the elderly: the PROTEGER study. Age (Dordr). 2014;36(3):9661.
34. Park S, Lakatta EG. Role of inflammation in the pathogenesis of arterial stiffness. Yonsei Med J. 2012;53(2):258-261.
35. Lakatta EG, Levy D. Arterial and cardiac aging: major shareholders in cardiovascular disease enterprises: Part II: the aging heart in health: links to heart disease. Circulation. 2003;107(2):346-354.
36. Zieman SJ, Melenovsky V, Kass DA. Mechanisms, pathophysiology, and therapy of arterial stiffness. Arterioscler Thromb Vasc Biol. 2005;25(5):932-943.
37. van der Loo B, Labugger R, Skepper JN, et al. Enhanced peroxynitrite formation is associated with vascular aging. J Exp Med. 2000;192(12):1731-1744.
38. Csiszar A, Ungvari Z, Edwards JG, et al. Aging-induced phenotypic changes and oxidative stress impair coronary arteriolar function. Circ Res. 2002;90(11):1159-1166.
39. Wang MC, Tsai WC, Chen JY, et al. Arterial stiffness correlated with cardiac remodelling in patients with chronic kidney disease. Nephrology (Carlton). 2007;12(6):591-597.
40. Belmin J, Bernard C, Corman B, et al. Increased production of tumor necrosis factor and interleukin-6 by arterial wall of aged rats. Am J Physiol. 1995;268(6 pt 2):H2288-2293.
41. Gerli R, Monti D, Bistoni O, et al. Chemokines, sTNF-Rs and sCD30 serum levels in healthy aged people and centenarians. Mech Ageing Dev. 2000;121(1-3):37-46.
42. Pasceri V, Willerson JT, Yeh ET. Direct proinflammatory effect of C-reactive protein on human endothelial cells. Circulation. 2000;102(18):2165-2168.
43. Torzewski M, Rist C, Mortensen RF, et al. C-reactive protein in the arterial intima: role of C-reactive protein receptor-dependent monocyte recruitment in atherogenesis. Arterioscler Thromb Vasc Biol. 2000;20(9):2094-2099.
44. Venugopal SK, Devaraj S, Yuhanna I, et al. Demonstration that C-reactive protein decreases eNOS expression and bioactivity in human aortic endothelial cells. Circulation. 2002;106(12):1439-1441.
45. Csiszar A, Ungvari Z, Koller A, et al. Aging-induced proinflammatory shift in cytokine expression profile in coronary arteries. FASEB J. 2003;17(9):1183-1185.
46. Spinetti G, Wang M, Monticone R, et al. Rat aortic MCP-1 and its receptor CCR2 increase with age and alter vascular smooth muscle cell function. Arterioscler Thromb Vasc Biol. 2004;24(8):1397-1402.
47. Mattace-Raso FU, van der Cammen TJ, van der Meer IM, et al. C-reactive protein and arterial stiffness in older adults: the Rotterdam Study. Atherosclerosis. 2004;176(1):111-116.
48. Mahmud A, Feely J. Arterial stiffness is related to systemic inflammation in essential hypertension. Hypertension. 2005;46(5):1118-1122.
49. Nagano M, Nakamura M, Sato K, et al. Association between serum C-reactive protein levels and pulse wave velocity: a population-based cross-sectional study in a general population. Atherosclerosis. 2005;180(1):189-195.
50. Nestel P, Shige H, Pomeroy S, et al. The n-3 fatty acids eicosapentaenoic acid and docosahexaenoic acid increase systemic arterial compliance in humans. Am J Clin Nutr. 2002;76(2):326-330.
51. Mäki-Petäjä KM, Elkhawad M, Cheriyan J, et al. Anti-tumor necrosis factor-α therapy reduces aortic inflammation and stiffness in patients with rheumatoid arthritis. Circulation. 2012;126(21):2473-2480.
52. Sommer IE, de Witte L, Begemann M, et al. Nonsteroidal anti-inflammatory drugs in schizophrenia: ready for practice or a good start? A meta-analysis. J Clin Psychiatry. 2012;73(4):414-419.
53. Sommer IE, van Westrhenen R, Begemann MJ, et al. Efficacy of anti-inflammatory agents to improve symptoms in patients with schizophrenia: an update. Schizophr Bull. 2014;40(1):181-191.

Issue
Current Psychiatry - 15(5)
Issue
Current Psychiatry - 15(5)
Page Number
52-57
Page Number
52-57
Publications
Publications
Topics
Article Type
Display Headline
Can anti-inflammatory medications improve symptoms and reduce mortality in schizophrenia?
Display Headline
Can anti-inflammatory medications improve symptoms and reduce mortality in schizophrenia?
Legacy Keywords
anti-inflammatory medications, schizophrenia, psychosis, psychotic disorder, psychotic disorders, cardiovascular disease, mortality, cytokine
Legacy Keywords
anti-inflammatory medications, schizophrenia, psychosis, psychotic disorder, psychotic disorders, cardiovascular disease, mortality, cytokine
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Is it a 'senior moment' or early dementia? Addressing memory concerns in older patients

Article Type
Changed
Tue, 12/11/2018 - 15:01
Display Headline
Is it a 'senior moment' or early dementia? Addressing memory concerns in older patients

Many older patients are concerned about their memory. The “worried well” may come into your office with a list of things they can’t recall, yet they remember each “deficit” quite well. Anticipatory anxiety about one’s own decline is common, and is most often concerned with changes in memory.1,2

Patients with dementia or early cognitive decline often are oblivious to their cognitive changes, however. Of particular concern is progressive dementia, especially Alzheimer’s disease (AD). Although jokes about “senior moments” are common, concern about AD incurs deep-seated worry. It is essential for clinicians to differentiate normal cognitive changes of aging—particularly those in memory—from early signs of neurodegenerative disease (Table 13).


In this article, we review typical memory changes in persons age >65, and differentiate these from mild cognitive impairment (MCI), an increasingly recognized prodrome of AD. Clinicians armed with knowledge of MCI are able to reassure the worried well, or recommend neuropsychological testing as indicated.


Is memory change inevitable with aging?

Memory loss is a common problem in aging, with variable severity. Research is establishing norms in cognitive functioning through the ninth decade of life.4 Controversy about sampling, measures, and methods abound,5 and drives prolific research on the subject, which is beyond the scope of this article. It has been demonstrated that there are a few “optimally aging” persons who avoid memory decline altogether.5,6 Most researchers and clinicians agree, however, that memory change is pervasive with advancing age.

Memory change follows a gradient with recent memories lost to a greater degree than remote memories (Ribot’s Law).7 Forgetfulness is characteristic of normal aging, and frequently manifests with misplaced objects and short-term lapses. However, this is not pathological—as long as the item or memory is recalled within 24 to 48 hours.

Compared with younger adults, healthy older adults are less efficient at encoding new information. Subsequently, they have more difficulty retrieving data, particularly after a delay. The time needed to learn and use new information increases, which is referred to as processing inefficiency. This influences changes in test performance across all cognitive domains, with decreases in measures of mental processing speed, working memory, and problem-solving.

Many patients who complain about “forgetfulness” are experiencing this normal change. It is not uncommon for a patient to offer a list of things she has forgotten recently, along with the dates and circumstances in which she forgot them. Because she sometimes forgets things, but remembers them later, there likely is nothing to worry about. If reminders—such as her list—help, this too is a good sign, because it shows her resourcefulness in using accommodations. If the patient is managing her normal activities, reassurance is warranted.


Mild cognitive impairment

Since at least 1958,8 clinical observations and research have recognized a prodrome that differentiates cognitive changes predictive of dementia from those that represent typical aging. Several studies and methods have converged toward consensus that MCI is a valid construct for that purpose, with ecological validity and sound predictive value. Clinical value is evident when a patient does not meet criteria for MCI; in this case, the clinician can reassure the worried well with conviction.

Revealing the diagnosis of MCI to patients requires sensitivity and assurance that you will reevaluate the condition annually. Although there is no evidence-based remedy for MCI or means to slow its progression to dementia, data are rapidly accruing regarding the value of lifestyle changes and other nonpharmacologic interventions.9

Recognizing MCI most simply requires 2 criteria:

The patient’s expressed concern about decline in cognitive functioning from a previous level of performance. Alternately, a caretaker’s report is valuable because the patient might lack insight. You are not looking for an inability to perform activities of daily living, which is indicative of frank dementia; rather, you want to determine whether the person’s independence in functional abilities is preserved, although less efficient. Patients might repeatedly report occurrences of new problems, although modest, in some cases. Although problems with memory often are the most frequently reported symptoms, changes can be observed in any cognitive domain. Uncharacteristic inability to understand instructions, frustration with new tasks, and inflexibility are common.

Quantified clinical assessment that the patient’s cognitive decline exceeds norms of his age cohort. Clinicians are already familiar with many of these tests (5-minute recall, clock face drawing, etc.). For MCI, we recommend the Montreal Cognitive Assessment (MoCA), which is specifically designed for MCI.10 It takes only 10 minutes to administer. Multiple versions of the MoCA, and instructions for its administration are available for provider use at www.mocatest.org.

When these criteria are met—a decline in previous functioning and an objective clinical confirmation—referral for neuropsychological testing is recommended. Subtypes of MCI—amnestic and non-amnestic—have been employed to specify the subtype (amnesic) that is most consistent with prodromal AD. However, this dichotomous scheme does not adequately explain or capture the heterogeneity of MCI.11,12

 

 


Medical considerations
Just as all domains of cognition are correlated to some degree, the overall health status of a person influences evaluation of memory. Variables, such as fatigue, test anxiety, mood, motivation, visual and auditory acuity, education, language fluency, attention, and pain, affect test performance. In addition, clinician rapport and the manner in which tests are administered must be considered.

Depression can mimic MCI. A depressed patient often has poor expectations of himself and slowed thinking, and might exaggerate symptoms. He might give up on tests or refuse to complete them. His presentation initially could suggest cognitive decline, but depression is revealed when the clinician pays attention to vegetative signs (insomnia, poor appetite) or suicidal ideation. There is growing evidence that subjective complaints of memory loss are more frequently associated with depression than with objective measures of cognitive impairment.13,14

Other treatable conditions can present with cognitive change (the so-called reversible dementias). A deficiency of vitamin B12, thiamine, or folate often is seen because quality of nutrition generally decreases with age. Hyponatremia and dehydration can present with confusion and memory impairment. Other treatable conditions include:

  • cerebral vasculitis, which could improve with immune suppressants
  • endocrine diseases, which might respond to hormonal or surgical treatment
  • normal pressure hydrocephalus, which can be relieved by surgical placement of a shunt.

Take a complete history. What exactly is the nature of the patient or caregiver’s complaint? You need to attempt to engage the patient in conversation, observing his behavior during the evaluation. Is there notable delay in response, difficulty in attention and focus, or in understanding questions?

The content of speech is an indicator of the patient’s information processing. Ask the patient to recite as many animals from the jungle as possible. Most people can come up with at least 15. The person with MCI will likely name fewer animals, but may respond well to cueing, and perform better in recognition (eg, pictures or drawings) vs retrieval. When asked to describe a typical day, the patient may offer a vague, nonchalant response eg, “I keep busy watching the news.” This kind of response may be evidence of confabulation; with further questioning, he is unable to identify current issues of interest.

Substance abuse. It is essential that clinicians recognize that elders are not exempt from alcohol and other drug abuse that affects cognition. Skilled history taking, including attention to non-verbal responses, is indicated. A defensive tone, rolling of eyes, or silent yet affirmative nodding are means by which caregivers offer essential “clues” to the provider.

A quick screening tool for the office is valuable; many clinicians are most familiar with the Mini-Mental State Examination or the Saint Louis University Mental Status Examination, which are known to be sensitive in detecting memory problems and other cognitive defects. As we noted, the MoCA is now recommended for differentiating more subtle changes of MCI.10,15 It is important to remember that common conditions such as an urinary tract infection or trauma after anesthesia for routine procedures such as colonoscopy can cause cognitive impairment. Again, eliciting history from a family member is valuable because the patient may have forgotten vital data.

A good physical exam is important when evaluating for dementia. Look for any neurologic anomaly. Check for disinhibition of primitive reflexes, eg, abnormal grasp or snout response or Babinski sign. Compare the symmetry and strength of deep tendon reflexes. Look for neurologic soft signs. Any pathological reflex response can be an important clue about neurodegeneration or space-occupying lesions. We recall seeing a 62-year-old man whose spouse brought him for evaluation for new-onset reckless driving and marked inattention to personal hygiene that developed over the previous 3 months. On examination, he appeared disheveled and had a dull affect, although disinhibited and careless. His mentation and gait were slowed. He denied distress of any kind. Frontal release signs were noted on exam. An MRI revealed a space-occupying lesion of the frontal lobe measuring 3 cm wide with a thickness of 2 cm, which pathology confirmed as a benign tumor.

Always check for arrhythmia and hypertension. These are significant risk factors for ischemic brain disease, multiple-infarct stroke, or other forms of vascular dementia. A shuffling gait suggests Parkinson’s disease, or even Lewy body dementia, or medication-related conditions, for example, from antipsychotics.

Take a medication history. Many common treatments for anxiety and insomnia can cause symptoms that mimic dementia. Digitalis toxicity results in poor recall and confusion. Combinations of common medicines (antacids, antihistamines, and others) compete for metabolic pathways and lead to altered mental status. Referencing the Beers List16 is valuable; anticholinergics, benzodiazepines, and narcotic analgesics are of special concern. The latter could still be useful for comfort care at the end of life.

 

 

It is common for seniors to take a variety of untested and unproven supplements in the hope of preventing or lessening memory problems. In addition to incurring significant costs, the indiscriminate use of supplements poses risks of toxicity, including unintended interactions with prescribed medications. Many older adults do not disclose their use of these supplements to providers because they do not consider them “medicine.”

Labs. The next level of evaluation calls for a basic laboratory workup. Check complete blood count, liver enzymes, thyroid function tests, vitamin D, B12 and folate levels; perform urinalysis and a complete metabolic panel. Look at a general hormone panel; abnormal values could reveal a pituitary adenoma. (In the past 33 years, the first author has found 42 pituitary tumors in the workup of mental status change.)

We use imaging, such as a CT or MRI of the brain, in almost all cases of suspected dementia. Cerebral atrophy, space-occupying lesions, and shifting of the ventricles often correspond with cognitive decline.


Treatment

Effective treatment of dementia remains elusive. Other than for the “reversible dementias,” pharmacotherapy has shown less progress than had been expected. Donepezil, galantamine, rivastigmine, and memantine could slow disease progression in some cases. There have been many studies for dementia preventives and treatments. Extensive reviews and meta-analyses, including those of randomized controlled trials17-19 abound for a variety of herbs, supplements, and antioxidants; none have shown compelling results. Table 2 lists Institute of Medicine recommendations supported by evidence that could reduce effects of cognitive aging.20



Recommendations from collaboration between the National Institute on Aging and the Alzheimer’s Association21 state that research should focus on biomarkers, such as neural substrates or genotypes. Indicators of oxidative stress (cytokines) and inflammation (isoprostanes) show promise as measures of brain changes that correspond with increased risk of AD or other dementias.


Summing up

Older adults are a heterogeneous group. Intellectual capacity does not diminish with advancing age. Many elders now exceed expectations for productivity, athletic ability, scientific achievement, and the creative arts. Others live longer with diminished quality of life, their health compromised by progressive neurodegenerative disease.

Age-associated memory change often is exaggerated and feared by older adults and, regrettably, is associated with inevitable functional impairment and is seen as heralding the loss of autonomy. The worried well are anxious, although the stigma associated with cognitive decline may preclude confiding their concerns.

Providers need the tools and acumen to treat patients along an increasingly long continuum of time, including conveyance of evidence-based encouragement toward optimal health and vitality.


Bottom Line

Variations in cognition occur over the lifespan. Be aware that mild cognitive impairment (MCI) is not a benign change but a harbinger of dementia for most affected people. We are able to differentiate the worried well from patients with MCI. The importance of early treatment for reversible forms of dementia is vital to reducing patient suffering and the overall burden on caregivers. Early identification of MCI will assist further research toward prevention or delay of progression to Alzheimer’s dementia.


Related Resources

  • American Psychological Association. Memory and Aging. www.apa.org/pi/aging/memory-and-aging.pdf.
  • Desai AK, Schwarz L. Subjective cognitive impairment: when to be concerned about ‘senior moments.’ Current Psychiatry. 2011;10(4):31,32,39,40,42,44,A.


Drug Brand Names

Donepezil • Aricept
Galantamine • Reminyl
Memantine • Namenda
Rivastigmine • Exelon


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

References


1. Serby MJ, Yhap C, Landron EY. A study of herbal remedies for memory complaints. J Neuropsychiatry Clin Neurosci. 2010;22(3):345-347.
2. Jaremka LM, Derry HM, Bornstein R, et al. Omega-3 supplementation and loneliness-related memory problems: secondary analyses of a randomized controlled trial. Psychosom Med. 2014;76(8):650-658.
3. Depp CA, Harmell A, Vania IV. Successful cognitive aging. In: Pardon MC, Bondi MW, eds. Behavioral neurobiology of aging. New York, NY: Springer-Verlag; 2012:35-50.
4. Invik RJ, Malec JF, Smith GE, et al. Mayo’s older Americans normative studies: WAIS-R, WMS-R, and AVLT norms for ages 56 to 97. Clin Neuropsychol. 1992;6(suppl 1):1-104.
5. Powell DH, Whitla DK. Profiles in cognitive aging. Boston, MA: Harvard University Press; 1994.
6. Negash S, Smith GE, Pankratz SE, et al. Successful aging: definitions and prediction of longevity and conversion to mild cognitive impairment. Am J Geriatr Psychiatry. 2011;19(6):581-588.
7. Ribot T. Diseases of memory: an essay in the positive psychology. London, United Kingdom: Kegan Paul Trench; 1882.
8. Kral VA. Neuropsychiatric observations in old peoples home: studies of memory dysfunction in senescence. J Gerontol. 1958;13(2):169-176.
9. Gitlin LN, Kales HC, Lyketsos CG. Nonpharmacologic management of behavioral symptoms in dementia. JAMA. 2012;308(19):2020-2029.
10. Nasreddine ZS, Phillips NA, Bédirian V, et al. The Montreal Cognitive Assessment, MoCA: a brief screening tool for mild cognitive assessment. J Am Geriatr Soc. 2005;53(4):695-699.
11. Clark LR, Delano-Wood L, Lisbon DJ, et al. Are empirically-derived subtypes of mild cognitive impairment consistent with conventional subtypes? J Intl Neuropsychol Soc. 2013;19(6):1-11.
12. Ganguli M, Snitz BE, Saxton JA, et al. Outcomes of mild cognitive impairment by definition: a population study. Arch Neurol. 2011;68(6):761-767.
13. Bartley M, Bokde AL, Ewers M, et al. Subjective memory complaints in community dwelling older people: the influence of brain and psychopathology. Intl J Geriatr Psychiatry. 2012;27(8):836-843.
14. Chung JC, Man DW. Self-appraised, informant-reported, and objective memory and cognitive function in mild cognitive impairment. Dement Geriatr Cogn Disord. 2009;27(2):187-193.
15. Tsoi KK, Chan JY, Hirai HW, et al. Cognitive tests to detect dementia: a systematic review and meta-analysis. JAMA Intern Med. 2015;175(9):1450-1458.
16. American Geriatrics Society 2012 Beers Criteria Update Expert Panel. American Geriatrics Society updated Beers Criteria for potentially inappropriate medication use in older adults. J Am Geriatr Soc. 2012;60(4):616-631.
17. May BH, Yang AW, Zhang AL, et al. Chinese herbal medicine for mild cognitive impairment and age associated memory impairment: a review of randomised controlled trials. Biogerontology. 2009;10(2):109-123.
18. Loef M, Walach H. The omega-6/omega-3 ratio and dementia or cognitive decline: a systematic review on human studies and biological evidence. J Nutr Gerontol Geriatr. 2013;32(1):1-23.
19. Solfrizzi VP, Panza F. Plant-based nutraceutical interventions against cognitive impairment and dementia: meta-analytic evidence of efficacy of a standardized Gingko biloba extract. J Alzheimers Dis. 2015;43(2):605-611.
20. Institute of Medicine. Cognitive aging: progress in understanding and opportunities for action. Washington, DC: National Academies Press; 2015.
21. Albert MS, DeKosky ST, Dickson D, et al. The diagnosis of mild cognitive impairment due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7(3):270-279.

Article PDF
Author and Disclosure Information

James A. Wilcox, MD, PhD
Professor of Clinical Psychiatry
University of Arizona
Staff Psychiatrist
Southern Arizona Veterans Administration Health Care System
Tucson, Arizona

P. Reid Duffy, PhD, RN
Mental Health Research Coordinator
Southern Arizona Veterans Administration Health Care System
Tucson, Arizona

Issue
Current Psychiatry - 15(5)
Publications
Topics
Page Number
28-30,32-34,40
Legacy Keywords
senior moment, senior moments, cognitive decline, cognitive deficit, dementia, memory problem, memory problems, memory concerns, geriatric, Alzheimer's disease, Alzheimer disease, cognitive disorder, cognitive disorders
Sections
Author and Disclosure Information

James A. Wilcox, MD, PhD
Professor of Clinical Psychiatry
University of Arizona
Staff Psychiatrist
Southern Arizona Veterans Administration Health Care System
Tucson, Arizona

P. Reid Duffy, PhD, RN
Mental Health Research Coordinator
Southern Arizona Veterans Administration Health Care System
Tucson, Arizona

Author and Disclosure Information

James A. Wilcox, MD, PhD
Professor of Clinical Psychiatry
University of Arizona
Staff Psychiatrist
Southern Arizona Veterans Administration Health Care System
Tucson, Arizona

P. Reid Duffy, PhD, RN
Mental Health Research Coordinator
Southern Arizona Veterans Administration Health Care System
Tucson, Arizona

Article PDF
Article PDF

Many older patients are concerned about their memory. The “worried well” may come into your office with a list of things they can’t recall, yet they remember each “deficit” quite well. Anticipatory anxiety about one’s own decline is common, and is most often concerned with changes in memory.1,2

Patients with dementia or early cognitive decline often are oblivious to their cognitive changes, however. Of particular concern is progressive dementia, especially Alzheimer’s disease (AD). Although jokes about “senior moments” are common, concern about AD incurs deep-seated worry. It is essential for clinicians to differentiate normal cognitive changes of aging—particularly those in memory—from early signs of neurodegenerative disease (Table 13).


In this article, we review typical memory changes in persons age >65, and differentiate these from mild cognitive impairment (MCI), an increasingly recognized prodrome of AD. Clinicians armed with knowledge of MCI are able to reassure the worried well, or recommend neuropsychological testing as indicated.


Is memory change inevitable with aging?

Memory loss is a common problem in aging, with variable severity. Research is establishing norms in cognitive functioning through the ninth decade of life.4 Controversy about sampling, measures, and methods abound,5 and drives prolific research on the subject, which is beyond the scope of this article. It has been demonstrated that there are a few “optimally aging” persons who avoid memory decline altogether.5,6 Most researchers and clinicians agree, however, that memory change is pervasive with advancing age.

Memory change follows a gradient with recent memories lost to a greater degree than remote memories (Ribot’s Law).7 Forgetfulness is characteristic of normal aging, and frequently manifests with misplaced objects and short-term lapses. However, this is not pathological—as long as the item or memory is recalled within 24 to 48 hours.

Compared with younger adults, healthy older adults are less efficient at encoding new information. Subsequently, they have more difficulty retrieving data, particularly after a delay. The time needed to learn and use new information increases, which is referred to as processing inefficiency. This influences changes in test performance across all cognitive domains, with decreases in measures of mental processing speed, working memory, and problem-solving.

Many patients who complain about “forgetfulness” are experiencing this normal change. It is not uncommon for a patient to offer a list of things she has forgotten recently, along with the dates and circumstances in which she forgot them. Because she sometimes forgets things, but remembers them later, there likely is nothing to worry about. If reminders—such as her list—help, this too is a good sign, because it shows her resourcefulness in using accommodations. If the patient is managing her normal activities, reassurance is warranted.


Mild cognitive impairment

Since at least 1958,8 clinical observations and research have recognized a prodrome that differentiates cognitive changes predictive of dementia from those that represent typical aging. Several studies and methods have converged toward consensus that MCI is a valid construct for that purpose, with ecological validity and sound predictive value. Clinical value is evident when a patient does not meet criteria for MCI; in this case, the clinician can reassure the worried well with conviction.

Revealing the diagnosis of MCI to patients requires sensitivity and assurance that you will reevaluate the condition annually. Although there is no evidence-based remedy for MCI or means to slow its progression to dementia, data are rapidly accruing regarding the value of lifestyle changes and other nonpharmacologic interventions.9

Recognizing MCI most simply requires 2 criteria:

The patient’s expressed concern about decline in cognitive functioning from a previous level of performance. Alternately, a caretaker’s report is valuable because the patient might lack insight. You are not looking for an inability to perform activities of daily living, which is indicative of frank dementia; rather, you want to determine whether the person’s independence in functional abilities is preserved, although less efficient. Patients might repeatedly report occurrences of new problems, although modest, in some cases. Although problems with memory often are the most frequently reported symptoms, changes can be observed in any cognitive domain. Uncharacteristic inability to understand instructions, frustration with new tasks, and inflexibility are common.

Quantified clinical assessment that the patient’s cognitive decline exceeds norms of his age cohort. Clinicians are already familiar with many of these tests (5-minute recall, clock face drawing, etc.). For MCI, we recommend the Montreal Cognitive Assessment (MoCA), which is specifically designed for MCI.10 It takes only 10 minutes to administer. Multiple versions of the MoCA, and instructions for its administration are available for provider use at www.mocatest.org.

When these criteria are met—a decline in previous functioning and an objective clinical confirmation—referral for neuropsychological testing is recommended. Subtypes of MCI—amnestic and non-amnestic—have been employed to specify the subtype (amnesic) that is most consistent with prodromal AD. However, this dichotomous scheme does not adequately explain or capture the heterogeneity of MCI.11,12

 

 


Medical considerations
Just as all domains of cognition are correlated to some degree, the overall health status of a person influences evaluation of memory. Variables, such as fatigue, test anxiety, mood, motivation, visual and auditory acuity, education, language fluency, attention, and pain, affect test performance. In addition, clinician rapport and the manner in which tests are administered must be considered.

Depression can mimic MCI. A depressed patient often has poor expectations of himself and slowed thinking, and might exaggerate symptoms. He might give up on tests or refuse to complete them. His presentation initially could suggest cognitive decline, but depression is revealed when the clinician pays attention to vegetative signs (insomnia, poor appetite) or suicidal ideation. There is growing evidence that subjective complaints of memory loss are more frequently associated with depression than with objective measures of cognitive impairment.13,14

Other treatable conditions can present with cognitive change (the so-called reversible dementias). A deficiency of vitamin B12, thiamine, or folate often is seen because quality of nutrition generally decreases with age. Hyponatremia and dehydration can present with confusion and memory impairment. Other treatable conditions include:

  • cerebral vasculitis, which could improve with immune suppressants
  • endocrine diseases, which might respond to hormonal or surgical treatment
  • normal pressure hydrocephalus, which can be relieved by surgical placement of a shunt.

Take a complete history. What exactly is the nature of the patient or caregiver’s complaint? You need to attempt to engage the patient in conversation, observing his behavior during the evaluation. Is there notable delay in response, difficulty in attention and focus, or in understanding questions?

The content of speech is an indicator of the patient’s information processing. Ask the patient to recite as many animals from the jungle as possible. Most people can come up with at least 15. The person with MCI will likely name fewer animals, but may respond well to cueing, and perform better in recognition (eg, pictures or drawings) vs retrieval. When asked to describe a typical day, the patient may offer a vague, nonchalant response eg, “I keep busy watching the news.” This kind of response may be evidence of confabulation; with further questioning, he is unable to identify current issues of interest.

Substance abuse. It is essential that clinicians recognize that elders are not exempt from alcohol and other drug abuse that affects cognition. Skilled history taking, including attention to non-verbal responses, is indicated. A defensive tone, rolling of eyes, or silent yet affirmative nodding are means by which caregivers offer essential “clues” to the provider.

A quick screening tool for the office is valuable; many clinicians are most familiar with the Mini-Mental State Examination or the Saint Louis University Mental Status Examination, which are known to be sensitive in detecting memory problems and other cognitive defects. As we noted, the MoCA is now recommended for differentiating more subtle changes of MCI.10,15 It is important to remember that common conditions such as an urinary tract infection or trauma after anesthesia for routine procedures such as colonoscopy can cause cognitive impairment. Again, eliciting history from a family member is valuable because the patient may have forgotten vital data.

A good physical exam is important when evaluating for dementia. Look for any neurologic anomaly. Check for disinhibition of primitive reflexes, eg, abnormal grasp or snout response or Babinski sign. Compare the symmetry and strength of deep tendon reflexes. Look for neurologic soft signs. Any pathological reflex response can be an important clue about neurodegeneration or space-occupying lesions. We recall seeing a 62-year-old man whose spouse brought him for evaluation for new-onset reckless driving and marked inattention to personal hygiene that developed over the previous 3 months. On examination, he appeared disheveled and had a dull affect, although disinhibited and careless. His mentation and gait were slowed. He denied distress of any kind. Frontal release signs were noted on exam. An MRI revealed a space-occupying lesion of the frontal lobe measuring 3 cm wide with a thickness of 2 cm, which pathology confirmed as a benign tumor.

Always check for arrhythmia and hypertension. These are significant risk factors for ischemic brain disease, multiple-infarct stroke, or other forms of vascular dementia. A shuffling gait suggests Parkinson’s disease, or even Lewy body dementia, or medication-related conditions, for example, from antipsychotics.

Take a medication history. Many common treatments for anxiety and insomnia can cause symptoms that mimic dementia. Digitalis toxicity results in poor recall and confusion. Combinations of common medicines (antacids, antihistamines, and others) compete for metabolic pathways and lead to altered mental status. Referencing the Beers List16 is valuable; anticholinergics, benzodiazepines, and narcotic analgesics are of special concern. The latter could still be useful for comfort care at the end of life.

 

 

It is common for seniors to take a variety of untested and unproven supplements in the hope of preventing or lessening memory problems. In addition to incurring significant costs, the indiscriminate use of supplements poses risks of toxicity, including unintended interactions with prescribed medications. Many older adults do not disclose their use of these supplements to providers because they do not consider them “medicine.”

Labs. The next level of evaluation calls for a basic laboratory workup. Check complete blood count, liver enzymes, thyroid function tests, vitamin D, B12 and folate levels; perform urinalysis and a complete metabolic panel. Look at a general hormone panel; abnormal values could reveal a pituitary adenoma. (In the past 33 years, the first author has found 42 pituitary tumors in the workup of mental status change.)

We use imaging, such as a CT or MRI of the brain, in almost all cases of suspected dementia. Cerebral atrophy, space-occupying lesions, and shifting of the ventricles often correspond with cognitive decline.


Treatment

Effective treatment of dementia remains elusive. Other than for the “reversible dementias,” pharmacotherapy has shown less progress than had been expected. Donepezil, galantamine, rivastigmine, and memantine could slow disease progression in some cases. There have been many studies for dementia preventives and treatments. Extensive reviews and meta-analyses, including those of randomized controlled trials17-19 abound for a variety of herbs, supplements, and antioxidants; none have shown compelling results. Table 2 lists Institute of Medicine recommendations supported by evidence that could reduce effects of cognitive aging.20



Recommendations from collaboration between the National Institute on Aging and the Alzheimer’s Association21 state that research should focus on biomarkers, such as neural substrates or genotypes. Indicators of oxidative stress (cytokines) and inflammation (isoprostanes) show promise as measures of brain changes that correspond with increased risk of AD or other dementias.


Summing up

Older adults are a heterogeneous group. Intellectual capacity does not diminish with advancing age. Many elders now exceed expectations for productivity, athletic ability, scientific achievement, and the creative arts. Others live longer with diminished quality of life, their health compromised by progressive neurodegenerative disease.

Age-associated memory change often is exaggerated and feared by older adults and, regrettably, is associated with inevitable functional impairment and is seen as heralding the loss of autonomy. The worried well are anxious, although the stigma associated with cognitive decline may preclude confiding their concerns.

Providers need the tools and acumen to treat patients along an increasingly long continuum of time, including conveyance of evidence-based encouragement toward optimal health and vitality.


Bottom Line

Variations in cognition occur over the lifespan. Be aware that mild cognitive impairment (MCI) is not a benign change but a harbinger of dementia for most affected people. We are able to differentiate the worried well from patients with MCI. The importance of early treatment for reversible forms of dementia is vital to reducing patient suffering and the overall burden on caregivers. Early identification of MCI will assist further research toward prevention or delay of progression to Alzheimer’s dementia.


Related Resources

  • American Psychological Association. Memory and Aging. www.apa.org/pi/aging/memory-and-aging.pdf.
  • Desai AK, Schwarz L. Subjective cognitive impairment: when to be concerned about ‘senior moments.’ Current Psychiatry. 2011;10(4):31,32,39,40,42,44,A.


Drug Brand Names

Donepezil • Aricept
Galantamine • Reminyl
Memantine • Namenda
Rivastigmine • Exelon


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

Many older patients are concerned about their memory. The “worried well” may come into your office with a list of things they can’t recall, yet they remember each “deficit” quite well. Anticipatory anxiety about one’s own decline is common, and is most often concerned with changes in memory.1,2

Patients with dementia or early cognitive decline often are oblivious to their cognitive changes, however. Of particular concern is progressive dementia, especially Alzheimer’s disease (AD). Although jokes about “senior moments” are common, concern about AD incurs deep-seated worry. It is essential for clinicians to differentiate normal cognitive changes of aging—particularly those in memory—from early signs of neurodegenerative disease (Table 13).


In this article, we review typical memory changes in persons age >65, and differentiate these from mild cognitive impairment (MCI), an increasingly recognized prodrome of AD. Clinicians armed with knowledge of MCI are able to reassure the worried well, or recommend neuropsychological testing as indicated.


Is memory change inevitable with aging?

Memory loss is a common problem in aging, with variable severity. Research is establishing norms in cognitive functioning through the ninth decade of life.4 Controversy about sampling, measures, and methods abound,5 and drives prolific research on the subject, which is beyond the scope of this article. It has been demonstrated that there are a few “optimally aging” persons who avoid memory decline altogether.5,6 Most researchers and clinicians agree, however, that memory change is pervasive with advancing age.

Memory change follows a gradient with recent memories lost to a greater degree than remote memories (Ribot’s Law).7 Forgetfulness is characteristic of normal aging, and frequently manifests with misplaced objects and short-term lapses. However, this is not pathological—as long as the item or memory is recalled within 24 to 48 hours.

Compared with younger adults, healthy older adults are less efficient at encoding new information. Subsequently, they have more difficulty retrieving data, particularly after a delay. The time needed to learn and use new information increases, which is referred to as processing inefficiency. This influences changes in test performance across all cognitive domains, with decreases in measures of mental processing speed, working memory, and problem-solving.

Many patients who complain about “forgetfulness” are experiencing this normal change. It is not uncommon for a patient to offer a list of things she has forgotten recently, along with the dates and circumstances in which she forgot them. Because she sometimes forgets things, but remembers them later, there likely is nothing to worry about. If reminders—such as her list—help, this too is a good sign, because it shows her resourcefulness in using accommodations. If the patient is managing her normal activities, reassurance is warranted.


Mild cognitive impairment

Since at least 1958,8 clinical observations and research have recognized a prodrome that differentiates cognitive changes predictive of dementia from those that represent typical aging. Several studies and methods have converged toward consensus that MCI is a valid construct for that purpose, with ecological validity and sound predictive value. Clinical value is evident when a patient does not meet criteria for MCI; in this case, the clinician can reassure the worried well with conviction.

Revealing the diagnosis of MCI to patients requires sensitivity and assurance that you will reevaluate the condition annually. Although there is no evidence-based remedy for MCI or means to slow its progression to dementia, data are rapidly accruing regarding the value of lifestyle changes and other nonpharmacologic interventions.9

Recognizing MCI most simply requires 2 criteria:

The patient’s expressed concern about decline in cognitive functioning from a previous level of performance. Alternately, a caretaker’s report is valuable because the patient might lack insight. You are not looking for an inability to perform activities of daily living, which is indicative of frank dementia; rather, you want to determine whether the person’s independence in functional abilities is preserved, although less efficient. Patients might repeatedly report occurrences of new problems, although modest, in some cases. Although problems with memory often are the most frequently reported symptoms, changes can be observed in any cognitive domain. Uncharacteristic inability to understand instructions, frustration with new tasks, and inflexibility are common.

Quantified clinical assessment that the patient’s cognitive decline exceeds norms of his age cohort. Clinicians are already familiar with many of these tests (5-minute recall, clock face drawing, etc.). For MCI, we recommend the Montreal Cognitive Assessment (MoCA), which is specifically designed for MCI.10 It takes only 10 minutes to administer. Multiple versions of the MoCA, and instructions for its administration are available for provider use at www.mocatest.org.

When these criteria are met—a decline in previous functioning and an objective clinical confirmation—referral for neuropsychological testing is recommended. Subtypes of MCI—amnestic and non-amnestic—have been employed to specify the subtype (amnesic) that is most consistent with prodromal AD. However, this dichotomous scheme does not adequately explain or capture the heterogeneity of MCI.11,12

 

 


Medical considerations
Just as all domains of cognition are correlated to some degree, the overall health status of a person influences evaluation of memory. Variables, such as fatigue, test anxiety, mood, motivation, visual and auditory acuity, education, language fluency, attention, and pain, affect test performance. In addition, clinician rapport and the manner in which tests are administered must be considered.

Depression can mimic MCI. A depressed patient often has poor expectations of himself and slowed thinking, and might exaggerate symptoms. He might give up on tests or refuse to complete them. His presentation initially could suggest cognitive decline, but depression is revealed when the clinician pays attention to vegetative signs (insomnia, poor appetite) or suicidal ideation. There is growing evidence that subjective complaints of memory loss are more frequently associated with depression than with objective measures of cognitive impairment.13,14

Other treatable conditions can present with cognitive change (the so-called reversible dementias). A deficiency of vitamin B12, thiamine, or folate often is seen because quality of nutrition generally decreases with age. Hyponatremia and dehydration can present with confusion and memory impairment. Other treatable conditions include:

  • cerebral vasculitis, which could improve with immune suppressants
  • endocrine diseases, which might respond to hormonal or surgical treatment
  • normal pressure hydrocephalus, which can be relieved by surgical placement of a shunt.

Take a complete history. What exactly is the nature of the patient or caregiver’s complaint? You need to attempt to engage the patient in conversation, observing his behavior during the evaluation. Is there notable delay in response, difficulty in attention and focus, or in understanding questions?

The content of speech is an indicator of the patient’s information processing. Ask the patient to recite as many animals from the jungle as possible. Most people can come up with at least 15. The person with MCI will likely name fewer animals, but may respond well to cueing, and perform better in recognition (eg, pictures or drawings) vs retrieval. When asked to describe a typical day, the patient may offer a vague, nonchalant response eg, “I keep busy watching the news.” This kind of response may be evidence of confabulation; with further questioning, he is unable to identify current issues of interest.

Substance abuse. It is essential that clinicians recognize that elders are not exempt from alcohol and other drug abuse that affects cognition. Skilled history taking, including attention to non-verbal responses, is indicated. A defensive tone, rolling of eyes, or silent yet affirmative nodding are means by which caregivers offer essential “clues” to the provider.

A quick screening tool for the office is valuable; many clinicians are most familiar with the Mini-Mental State Examination or the Saint Louis University Mental Status Examination, which are known to be sensitive in detecting memory problems and other cognitive defects. As we noted, the MoCA is now recommended for differentiating more subtle changes of MCI.10,15 It is important to remember that common conditions such as an urinary tract infection or trauma after anesthesia for routine procedures such as colonoscopy can cause cognitive impairment. Again, eliciting history from a family member is valuable because the patient may have forgotten vital data.

A good physical exam is important when evaluating for dementia. Look for any neurologic anomaly. Check for disinhibition of primitive reflexes, eg, abnormal grasp or snout response or Babinski sign. Compare the symmetry and strength of deep tendon reflexes. Look for neurologic soft signs. Any pathological reflex response can be an important clue about neurodegeneration or space-occupying lesions. We recall seeing a 62-year-old man whose spouse brought him for evaluation for new-onset reckless driving and marked inattention to personal hygiene that developed over the previous 3 months. On examination, he appeared disheveled and had a dull affect, although disinhibited and careless. His mentation and gait were slowed. He denied distress of any kind. Frontal release signs were noted on exam. An MRI revealed a space-occupying lesion of the frontal lobe measuring 3 cm wide with a thickness of 2 cm, which pathology confirmed as a benign tumor.

Always check for arrhythmia and hypertension. These are significant risk factors for ischemic brain disease, multiple-infarct stroke, or other forms of vascular dementia. A shuffling gait suggests Parkinson’s disease, or even Lewy body dementia, or medication-related conditions, for example, from antipsychotics.

Take a medication history. Many common treatments for anxiety and insomnia can cause symptoms that mimic dementia. Digitalis toxicity results in poor recall and confusion. Combinations of common medicines (antacids, antihistamines, and others) compete for metabolic pathways and lead to altered mental status. Referencing the Beers List16 is valuable; anticholinergics, benzodiazepines, and narcotic analgesics are of special concern. The latter could still be useful for comfort care at the end of life.

 

 

It is common for seniors to take a variety of untested and unproven supplements in the hope of preventing or lessening memory problems. In addition to incurring significant costs, the indiscriminate use of supplements poses risks of toxicity, including unintended interactions with prescribed medications. Many older adults do not disclose their use of these supplements to providers because they do not consider them “medicine.”

Labs. The next level of evaluation calls for a basic laboratory workup. Check complete blood count, liver enzymes, thyroid function tests, vitamin D, B12 and folate levels; perform urinalysis and a complete metabolic panel. Look at a general hormone panel; abnormal values could reveal a pituitary adenoma. (In the past 33 years, the first author has found 42 pituitary tumors in the workup of mental status change.)

We use imaging, such as a CT or MRI of the brain, in almost all cases of suspected dementia. Cerebral atrophy, space-occupying lesions, and shifting of the ventricles often correspond with cognitive decline.


Treatment

Effective treatment of dementia remains elusive. Other than for the “reversible dementias,” pharmacotherapy has shown less progress than had been expected. Donepezil, galantamine, rivastigmine, and memantine could slow disease progression in some cases. There have been many studies for dementia preventives and treatments. Extensive reviews and meta-analyses, including those of randomized controlled trials17-19 abound for a variety of herbs, supplements, and antioxidants; none have shown compelling results. Table 2 lists Institute of Medicine recommendations supported by evidence that could reduce effects of cognitive aging.20



Recommendations from collaboration between the National Institute on Aging and the Alzheimer’s Association21 state that research should focus on biomarkers, such as neural substrates or genotypes. Indicators of oxidative stress (cytokines) and inflammation (isoprostanes) show promise as measures of brain changes that correspond with increased risk of AD or other dementias.


Summing up

Older adults are a heterogeneous group. Intellectual capacity does not diminish with advancing age. Many elders now exceed expectations for productivity, athletic ability, scientific achievement, and the creative arts. Others live longer with diminished quality of life, their health compromised by progressive neurodegenerative disease.

Age-associated memory change often is exaggerated and feared by older adults and, regrettably, is associated with inevitable functional impairment and is seen as heralding the loss of autonomy. The worried well are anxious, although the stigma associated with cognitive decline may preclude confiding their concerns.

Providers need the tools and acumen to treat patients along an increasingly long continuum of time, including conveyance of evidence-based encouragement toward optimal health and vitality.


Bottom Line

Variations in cognition occur over the lifespan. Be aware that mild cognitive impairment (MCI) is not a benign change but a harbinger of dementia for most affected people. We are able to differentiate the worried well from patients with MCI. The importance of early treatment for reversible forms of dementia is vital to reducing patient suffering and the overall burden on caregivers. Early identification of MCI will assist further research toward prevention or delay of progression to Alzheimer’s dementia.


Related Resources

  • American Psychological Association. Memory and Aging. www.apa.org/pi/aging/memory-and-aging.pdf.
  • Desai AK, Schwarz L. Subjective cognitive impairment: when to be concerned about ‘senior moments.’ Current Psychiatry. 2011;10(4):31,32,39,40,42,44,A.


Drug Brand Names

Donepezil • Aricept
Galantamine • Reminyl
Memantine • Namenda
Rivastigmine • Exelon


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

References


1. Serby MJ, Yhap C, Landron EY. A study of herbal remedies for memory complaints. J Neuropsychiatry Clin Neurosci. 2010;22(3):345-347.
2. Jaremka LM, Derry HM, Bornstein R, et al. Omega-3 supplementation and loneliness-related memory problems: secondary analyses of a randomized controlled trial. Psychosom Med. 2014;76(8):650-658.
3. Depp CA, Harmell A, Vania IV. Successful cognitive aging. In: Pardon MC, Bondi MW, eds. Behavioral neurobiology of aging. New York, NY: Springer-Verlag; 2012:35-50.
4. Invik RJ, Malec JF, Smith GE, et al. Mayo’s older Americans normative studies: WAIS-R, WMS-R, and AVLT norms for ages 56 to 97. Clin Neuropsychol. 1992;6(suppl 1):1-104.
5. Powell DH, Whitla DK. Profiles in cognitive aging. Boston, MA: Harvard University Press; 1994.
6. Negash S, Smith GE, Pankratz SE, et al. Successful aging: definitions and prediction of longevity and conversion to mild cognitive impairment. Am J Geriatr Psychiatry. 2011;19(6):581-588.
7. Ribot T. Diseases of memory: an essay in the positive psychology. London, United Kingdom: Kegan Paul Trench; 1882.
8. Kral VA. Neuropsychiatric observations in old peoples home: studies of memory dysfunction in senescence. J Gerontol. 1958;13(2):169-176.
9. Gitlin LN, Kales HC, Lyketsos CG. Nonpharmacologic management of behavioral symptoms in dementia. JAMA. 2012;308(19):2020-2029.
10. Nasreddine ZS, Phillips NA, Bédirian V, et al. The Montreal Cognitive Assessment, MoCA: a brief screening tool for mild cognitive assessment. J Am Geriatr Soc. 2005;53(4):695-699.
11. Clark LR, Delano-Wood L, Lisbon DJ, et al. Are empirically-derived subtypes of mild cognitive impairment consistent with conventional subtypes? J Intl Neuropsychol Soc. 2013;19(6):1-11.
12. Ganguli M, Snitz BE, Saxton JA, et al. Outcomes of mild cognitive impairment by definition: a population study. Arch Neurol. 2011;68(6):761-767.
13. Bartley M, Bokde AL, Ewers M, et al. Subjective memory complaints in community dwelling older people: the influence of brain and psychopathology. Intl J Geriatr Psychiatry. 2012;27(8):836-843.
14. Chung JC, Man DW. Self-appraised, informant-reported, and objective memory and cognitive function in mild cognitive impairment. Dement Geriatr Cogn Disord. 2009;27(2):187-193.
15. Tsoi KK, Chan JY, Hirai HW, et al. Cognitive tests to detect dementia: a systematic review and meta-analysis. JAMA Intern Med. 2015;175(9):1450-1458.
16. American Geriatrics Society 2012 Beers Criteria Update Expert Panel. American Geriatrics Society updated Beers Criteria for potentially inappropriate medication use in older adults. J Am Geriatr Soc. 2012;60(4):616-631.
17. May BH, Yang AW, Zhang AL, et al. Chinese herbal medicine for mild cognitive impairment and age associated memory impairment: a review of randomised controlled trials. Biogerontology. 2009;10(2):109-123.
18. Loef M, Walach H. The omega-6/omega-3 ratio and dementia or cognitive decline: a systematic review on human studies and biological evidence. J Nutr Gerontol Geriatr. 2013;32(1):1-23.
19. Solfrizzi VP, Panza F. Plant-based nutraceutical interventions against cognitive impairment and dementia: meta-analytic evidence of efficacy of a standardized Gingko biloba extract. J Alzheimers Dis. 2015;43(2):605-611.
20. Institute of Medicine. Cognitive aging: progress in understanding and opportunities for action. Washington, DC: National Academies Press; 2015.
21. Albert MS, DeKosky ST, Dickson D, et al. The diagnosis of mild cognitive impairment due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7(3):270-279.

References


1. Serby MJ, Yhap C, Landron EY. A study of herbal remedies for memory complaints. J Neuropsychiatry Clin Neurosci. 2010;22(3):345-347.
2. Jaremka LM, Derry HM, Bornstein R, et al. Omega-3 supplementation and loneliness-related memory problems: secondary analyses of a randomized controlled trial. Psychosom Med. 2014;76(8):650-658.
3. Depp CA, Harmell A, Vania IV. Successful cognitive aging. In: Pardon MC, Bondi MW, eds. Behavioral neurobiology of aging. New York, NY: Springer-Verlag; 2012:35-50.
4. Invik RJ, Malec JF, Smith GE, et al. Mayo’s older Americans normative studies: WAIS-R, WMS-R, and AVLT norms for ages 56 to 97. Clin Neuropsychol. 1992;6(suppl 1):1-104.
5. Powell DH, Whitla DK. Profiles in cognitive aging. Boston, MA: Harvard University Press; 1994.
6. Negash S, Smith GE, Pankratz SE, et al. Successful aging: definitions and prediction of longevity and conversion to mild cognitive impairment. Am J Geriatr Psychiatry. 2011;19(6):581-588.
7. Ribot T. Diseases of memory: an essay in the positive psychology. London, United Kingdom: Kegan Paul Trench; 1882.
8. Kral VA. Neuropsychiatric observations in old peoples home: studies of memory dysfunction in senescence. J Gerontol. 1958;13(2):169-176.
9. Gitlin LN, Kales HC, Lyketsos CG. Nonpharmacologic management of behavioral symptoms in dementia. JAMA. 2012;308(19):2020-2029.
10. Nasreddine ZS, Phillips NA, Bédirian V, et al. The Montreal Cognitive Assessment, MoCA: a brief screening tool for mild cognitive assessment. J Am Geriatr Soc. 2005;53(4):695-699.
11. Clark LR, Delano-Wood L, Lisbon DJ, et al. Are empirically-derived subtypes of mild cognitive impairment consistent with conventional subtypes? J Intl Neuropsychol Soc. 2013;19(6):1-11.
12. Ganguli M, Snitz BE, Saxton JA, et al. Outcomes of mild cognitive impairment by definition: a population study. Arch Neurol. 2011;68(6):761-767.
13. Bartley M, Bokde AL, Ewers M, et al. Subjective memory complaints in community dwelling older people: the influence of brain and psychopathology. Intl J Geriatr Psychiatry. 2012;27(8):836-843.
14. Chung JC, Man DW. Self-appraised, informant-reported, and objective memory and cognitive function in mild cognitive impairment. Dement Geriatr Cogn Disord. 2009;27(2):187-193.
15. Tsoi KK, Chan JY, Hirai HW, et al. Cognitive tests to detect dementia: a systematic review and meta-analysis. JAMA Intern Med. 2015;175(9):1450-1458.
16. American Geriatrics Society 2012 Beers Criteria Update Expert Panel. American Geriatrics Society updated Beers Criteria for potentially inappropriate medication use in older adults. J Am Geriatr Soc. 2012;60(4):616-631.
17. May BH, Yang AW, Zhang AL, et al. Chinese herbal medicine for mild cognitive impairment and age associated memory impairment: a review of randomised controlled trials. Biogerontology. 2009;10(2):109-123.
18. Loef M, Walach H. The omega-6/omega-3 ratio and dementia or cognitive decline: a systematic review on human studies and biological evidence. J Nutr Gerontol Geriatr. 2013;32(1):1-23.
19. Solfrizzi VP, Panza F. Plant-based nutraceutical interventions against cognitive impairment and dementia: meta-analytic evidence of efficacy of a standardized Gingko biloba extract. J Alzheimers Dis. 2015;43(2):605-611.
20. Institute of Medicine. Cognitive aging: progress in understanding and opportunities for action. Washington, DC: National Academies Press; 2015.
21. Albert MS, DeKosky ST, Dickson D, et al. The diagnosis of mild cognitive impairment due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7(3):270-279.

Issue
Current Psychiatry - 15(5)
Issue
Current Psychiatry - 15(5)
Page Number
28-30,32-34,40
Page Number
28-30,32-34,40
Publications
Publications
Topics
Article Type
Display Headline
Is it a 'senior moment' or early dementia? Addressing memory concerns in older patients
Display Headline
Is it a 'senior moment' or early dementia? Addressing memory concerns in older patients
Legacy Keywords
senior moment, senior moments, cognitive decline, cognitive deficit, dementia, memory problem, memory problems, memory concerns, geriatric, Alzheimer's disease, Alzheimer disease, cognitive disorder, cognitive disorders
Legacy Keywords
senior moment, senior moments, cognitive decline, cognitive deficit, dementia, memory problem, memory problems, memory concerns, geriatric, Alzheimer's disease, Alzheimer disease, cognitive disorder, cognitive disorders
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Don’t assume that psychiatric patients lack capacity to make decisions about care

Article Type
Changed
Tue, 12/11/2018 - 14:33
Display Headline
Don’t assume that psychiatric patients lack capacity to make decisions about care

Some practitioners of medicine—including psychiatrists—might equate “psychosis” with incapacity, but that isn’t necessarily true. Even patients who, by most measures, are deemed psychotic might be capable of making wise and thoughtful decisions about their life. The case I describe in this article demonstrates that fact.

While rotating on a busy consultation service, I was asked to evaluate the capacity of a woman who had a diagnosis of schizophrenia and was being seen for worsening auditory hallucinations and progressive weight loss. She had a complicated medical course that eventually led to multiple requests to the consult team for a capacity evaluation.

The question of capacity in this patient, and in the psychiatric population generally, motivated me to review the literature, because the assumption by many on the medical teams involved in this patient’s care was that psychiatric patients do not have the capacity to participate in their own care. My goal here is to clarify the misconceptions in regard to this situation.


CASE REPORT
Schizophrenia, weight loss, back pain
Ms. V, age 67, a resident of a group home for the past 6 years, was brought to the emergency department (ED) because of weight loss and auditory hallucinations that had developed during the past few months. She had a history of paranoid schizophrenia that included several psychiatric hospitalizations but no known medical history.

The patient appeared cachectic and dehydrated. When approached, she was pleasant and reported hearing voices of the “devil.”

“They are not scary,” she confided. “They talk to me about art and literature.”

Over the past 6 months, Ms. V had lost 60 lb; she was now bedridden because of back pain. Collateral information obtained from staff members at the group home indicated that she had refused to get out of bed, and only intermittently took her medications or ate meals during the past few months. In general, however, she had been relatively stable over the course of her psychiatric illness, was adherent to psychiatric treatment, and had had no psychiatric hospitalizations in the past 3 decades.

Ominous development. Although the ED staff was convinced that Ms. V needed psychiatric admission, we (the consult team) first requested a detailed medical workup, including imaging studies. A CT scan showed multiple metastatic foci throughout her spine. She was admitted to the medical service.

Respiratory distress developed; her condition deteriorated. Numerous capacity consults were requested because she refused a medical workup or to sign do-not-resuscitate and do-not-intubate orders. Each time an evaluation was performed, Ms. V was deemed by various clinicians on the consult service to have decision-making capacity.

The patient grew unhappy with the staff’s insistence that she undergo more tests regardless of her stated wishes. The palliative care service determined that further workup would not benefit her medically: Ms. V’s condition would be grave and her prognosis poor regardless of what treatment she received.

The medical team continued to believe that, because that this patient had a mental illness and was actively hallucinating, she did not have the capacity to refuse any proposed treatments and tests.


What is capacity?
Capacity is an assessment of a person’s ability to make rational decisions. This includes the ability to understand, appreciate, and manipulate information in reaching such decisions. Determining whether a patient has the capacity to accept or refuse treatment is a medical decision that any physician can make; however, consultation−liaison psychiatrists are the experts who often are involved in this activity, particularly in patients who have a psychiatric comorbidity.

Capacity is evaluated by assessing 4 standards; that is, whether a patient can:

 

  • communicate choice about proposed treatment
  • understand her (his) medical situation
  • appreciate the situation and its consequences
  • manipulate information to reach a rational decision.1-3
  • manipulate information to reach a rational decision.
     

CASE REPORT continued
Although Ms. V’s health was deteriorating and her auditory hallucinations were becoming worse, she appeared insightful about her medical problems, understood her prognosis, and wanted comfort care. She understood that having multiple metastases meant a poor prognosis, and that a biopsy might yield a medical diagnosis. She stated, “If it were caught earlier and I was better able to tolerate treatment, it would make sense to know for sure, but now it doesn’t make sense. I just want to have no pain in my end.”
 

Misconceptions
In a study by Ganzini et al,4,5 395 consultation−liaison psychiatrists, geriatricians, and geriatric psychologists responded to a survey in which they rated types of misunderstandings by clinicians who refer patients for assessment of decision-making capacity. Seventy percent reported that it is common that, when a patient has a mental illness such as schizophrenia, practitioners think that the patient lacks capacity to make medical decisions. However, results of a meta-analysis by Jeste et al,6 in which the magnitude of impairment of decisional capacity in patients with schizophrenia was assessed in comparison to that of normal subjects, suggest that the presence of schizophrenia does not necessarily mean the patient has impaired capacity.

 

 

Voluntary participation in research. Many patients with schizophrenia volunteer to participate in clinical trials even when they are acutely psychotic and admitted to a psychiatric hospital. Given the importance placed on participants’ voluntary informed consent as a prerequisite for ethical conduct of research, the cognitive and emotional impairments associated with schizophrenia raise questions about patients’ capacity to consent.

As is true in other areas of functional capacity, the ability of patients with schizophrenia to make competent decisions relates more to their overall cognitive functioning than to the presence or absence of specific symptoms of the disorder.7 Documentation of longitudinal consent-related abilities among research participants with schizophrenia in the long-term Clinical Antipsychotic Trials of Intervention Effectiveness study indicated that most participants had stable or improved consent-related abilities. Although almost 25% of participants experienced substantial worsening, only 4% fell below the study’s capacity threshold for enrollment.8


What I learned from Ms. V
A diagnosis of schizophrenia does not automatically render a person unable to make decisions about medical care. Even patients who have severe mental illness might have significant intact areas of reality testing. Ethically, it is important to at least consider that the chronically mentally ill can understand treatment options and express consistent choices.

Healthcare providers might tend to exclude psychiatric patients from end-of-life decisions because they (1) are worried about the emotional fragility of such patients and (2) assume that patients lack capacity to participate in making such important decisions. The case presented here is an example of a patient with a severe psychiatric diagnosis being able to participate in her care despite her mental state.

 


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


1. Appelbaum PS, Grisso T. Assessing patients’ capacities to consent to treatment. N Engl J Med. 1988;319(25):1635-1638.
2. Leo RJ. Competency and the capacity to make treatment decisions: a primer for primary care physicians. Prim Care Companion J Clin Psychiatry. 1999;1(5):131-141.
3. White MM, Lofwall MR. Challenges of the capacity evaluation for the consultation-liaison psychiatrist. J Psychiatr Pract. 2015;21(2):160-170.
4. Ganzini L, Volicer L, Nelson W, et al. Pitfalls in assessment of decision-making capacity. Psychosomatics. 2003;44(3):237-243.
5. Ganzini L, Volicer L, Nelson WA, et al. Ten myths about decision-making capacity. J Am Med Dir Assoc. 2005;6(3):S100-S104.
6. Jeste DV, Depp CA, Palmer BW. Magnitude of impairment in decisional capacity in people with schizophrenia compared to normal subjects: an overview. Schizophr Bull. 2005;32(1):121-128.
7. Appelbaum PS. Decisional capacity of patients with schizophrenia to consent to research: taking stock. Schizophr Bull. 2005;32(1):22-25.
8. Stroup TS, Appelbaum PS, Gu H, et al. Longitudinal consent-related abilities among research participants with schizophrenia: results from the CATIE study. Schizophr Res. 2011;130(1-3):47-52.

Article PDF
Author and Disclosure Information

 

Sahil Munjal, MD
PGY-3 Resident in Psychiatry
New York Medical College at Westchester Medical Center
Valhalla, New York

Issue
Current Psychiatry - 15(4)
Publications
Topics
Page Number
e4-e6
Legacy Keywords
capacity, severe mental illness, SMI, making decisions, evaluating capacity, schizophrenia. psychosis, psychotic disorder, psychotic disorders
Sections
Author and Disclosure Information

 

Sahil Munjal, MD
PGY-3 Resident in Psychiatry
New York Medical College at Westchester Medical Center
Valhalla, New York

Author and Disclosure Information

 

Sahil Munjal, MD
PGY-3 Resident in Psychiatry
New York Medical College at Westchester Medical Center
Valhalla, New York

Article PDF
Article PDF

Some practitioners of medicine—including psychiatrists—might equate “psychosis” with incapacity, but that isn’t necessarily true. Even patients who, by most measures, are deemed psychotic might be capable of making wise and thoughtful decisions about their life. The case I describe in this article demonstrates that fact.

While rotating on a busy consultation service, I was asked to evaluate the capacity of a woman who had a diagnosis of schizophrenia and was being seen for worsening auditory hallucinations and progressive weight loss. She had a complicated medical course that eventually led to multiple requests to the consult team for a capacity evaluation.

The question of capacity in this patient, and in the psychiatric population generally, motivated me to review the literature, because the assumption by many on the medical teams involved in this patient’s care was that psychiatric patients do not have the capacity to participate in their own care. My goal here is to clarify the misconceptions in regard to this situation.


CASE REPORT
Schizophrenia, weight loss, back pain
Ms. V, age 67, a resident of a group home for the past 6 years, was brought to the emergency department (ED) because of weight loss and auditory hallucinations that had developed during the past few months. She had a history of paranoid schizophrenia that included several psychiatric hospitalizations but no known medical history.

The patient appeared cachectic and dehydrated. When approached, she was pleasant and reported hearing voices of the “devil.”

“They are not scary,” she confided. “They talk to me about art and literature.”

Over the past 6 months, Ms. V had lost 60 lb; she was now bedridden because of back pain. Collateral information obtained from staff members at the group home indicated that she had refused to get out of bed, and only intermittently took her medications or ate meals during the past few months. In general, however, she had been relatively stable over the course of her psychiatric illness, was adherent to psychiatric treatment, and had had no psychiatric hospitalizations in the past 3 decades.

Ominous development. Although the ED staff was convinced that Ms. V needed psychiatric admission, we (the consult team) first requested a detailed medical workup, including imaging studies. A CT scan showed multiple metastatic foci throughout her spine. She was admitted to the medical service.

Respiratory distress developed; her condition deteriorated. Numerous capacity consults were requested because she refused a medical workup or to sign do-not-resuscitate and do-not-intubate orders. Each time an evaluation was performed, Ms. V was deemed by various clinicians on the consult service to have decision-making capacity.

The patient grew unhappy with the staff’s insistence that she undergo more tests regardless of her stated wishes. The palliative care service determined that further workup would not benefit her medically: Ms. V’s condition would be grave and her prognosis poor regardless of what treatment she received.

The medical team continued to believe that, because that this patient had a mental illness and was actively hallucinating, she did not have the capacity to refuse any proposed treatments and tests.


What is capacity?
Capacity is an assessment of a person’s ability to make rational decisions. This includes the ability to understand, appreciate, and manipulate information in reaching such decisions. Determining whether a patient has the capacity to accept or refuse treatment is a medical decision that any physician can make; however, consultation−liaison psychiatrists are the experts who often are involved in this activity, particularly in patients who have a psychiatric comorbidity.

Capacity is evaluated by assessing 4 standards; that is, whether a patient can:

 

  • communicate choice about proposed treatment
  • understand her (his) medical situation
  • appreciate the situation and its consequences
  • manipulate information to reach a rational decision.1-3
  • manipulate information to reach a rational decision.
     

CASE REPORT continued
Although Ms. V’s health was deteriorating and her auditory hallucinations were becoming worse, she appeared insightful about her medical problems, understood her prognosis, and wanted comfort care. She understood that having multiple metastases meant a poor prognosis, and that a biopsy might yield a medical diagnosis. She stated, “If it were caught earlier and I was better able to tolerate treatment, it would make sense to know for sure, but now it doesn’t make sense. I just want to have no pain in my end.”
 

Misconceptions
In a study by Ganzini et al,4,5 395 consultation−liaison psychiatrists, geriatricians, and geriatric psychologists responded to a survey in which they rated types of misunderstandings by clinicians who refer patients for assessment of decision-making capacity. Seventy percent reported that it is common that, when a patient has a mental illness such as schizophrenia, practitioners think that the patient lacks capacity to make medical decisions. However, results of a meta-analysis by Jeste et al,6 in which the magnitude of impairment of decisional capacity in patients with schizophrenia was assessed in comparison to that of normal subjects, suggest that the presence of schizophrenia does not necessarily mean the patient has impaired capacity.

 

 

Voluntary participation in research. Many patients with schizophrenia volunteer to participate in clinical trials even when they are acutely psychotic and admitted to a psychiatric hospital. Given the importance placed on participants’ voluntary informed consent as a prerequisite for ethical conduct of research, the cognitive and emotional impairments associated with schizophrenia raise questions about patients’ capacity to consent.

As is true in other areas of functional capacity, the ability of patients with schizophrenia to make competent decisions relates more to their overall cognitive functioning than to the presence or absence of specific symptoms of the disorder.7 Documentation of longitudinal consent-related abilities among research participants with schizophrenia in the long-term Clinical Antipsychotic Trials of Intervention Effectiveness study indicated that most participants had stable or improved consent-related abilities. Although almost 25% of participants experienced substantial worsening, only 4% fell below the study’s capacity threshold for enrollment.8


What I learned from Ms. V
A diagnosis of schizophrenia does not automatically render a person unable to make decisions about medical care. Even patients who have severe mental illness might have significant intact areas of reality testing. Ethically, it is important to at least consider that the chronically mentally ill can understand treatment options and express consistent choices.

Healthcare providers might tend to exclude psychiatric patients from end-of-life decisions because they (1) are worried about the emotional fragility of such patients and (2) assume that patients lack capacity to participate in making such important decisions. The case presented here is an example of a patient with a severe psychiatric diagnosis being able to participate in her care despite her mental state.

 


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

Some practitioners of medicine—including psychiatrists—might equate “psychosis” with incapacity, but that isn’t necessarily true. Even patients who, by most measures, are deemed psychotic might be capable of making wise and thoughtful decisions about their life. The case I describe in this article demonstrates that fact.

While rotating on a busy consultation service, I was asked to evaluate the capacity of a woman who had a diagnosis of schizophrenia and was being seen for worsening auditory hallucinations and progressive weight loss. She had a complicated medical course that eventually led to multiple requests to the consult team for a capacity evaluation.

The question of capacity in this patient, and in the psychiatric population generally, motivated me to review the literature, because the assumption by many on the medical teams involved in this patient’s care was that psychiatric patients do not have the capacity to participate in their own care. My goal here is to clarify the misconceptions in regard to this situation.


CASE REPORT
Schizophrenia, weight loss, back pain
Ms. V, age 67, a resident of a group home for the past 6 years, was brought to the emergency department (ED) because of weight loss and auditory hallucinations that had developed during the past few months. She had a history of paranoid schizophrenia that included several psychiatric hospitalizations but no known medical history.

The patient appeared cachectic and dehydrated. When approached, she was pleasant and reported hearing voices of the “devil.”

“They are not scary,” she confided. “They talk to me about art and literature.”

Over the past 6 months, Ms. V had lost 60 lb; she was now bedridden because of back pain. Collateral information obtained from staff members at the group home indicated that she had refused to get out of bed, and only intermittently took her medications or ate meals during the past few months. In general, however, she had been relatively stable over the course of her psychiatric illness, was adherent to psychiatric treatment, and had had no psychiatric hospitalizations in the past 3 decades.

Ominous development. Although the ED staff was convinced that Ms. V needed psychiatric admission, we (the consult team) first requested a detailed medical workup, including imaging studies. A CT scan showed multiple metastatic foci throughout her spine. She was admitted to the medical service.

Respiratory distress developed; her condition deteriorated. Numerous capacity consults were requested because she refused a medical workup or to sign do-not-resuscitate and do-not-intubate orders. Each time an evaluation was performed, Ms. V was deemed by various clinicians on the consult service to have decision-making capacity.

The patient grew unhappy with the staff’s insistence that she undergo more tests regardless of her stated wishes. The palliative care service determined that further workup would not benefit her medically: Ms. V’s condition would be grave and her prognosis poor regardless of what treatment she received.

The medical team continued to believe that, because that this patient had a mental illness and was actively hallucinating, she did not have the capacity to refuse any proposed treatments and tests.


What is capacity?
Capacity is an assessment of a person’s ability to make rational decisions. This includes the ability to understand, appreciate, and manipulate information in reaching such decisions. Determining whether a patient has the capacity to accept or refuse treatment is a medical decision that any physician can make; however, consultation−liaison psychiatrists are the experts who often are involved in this activity, particularly in patients who have a psychiatric comorbidity.

Capacity is evaluated by assessing 4 standards; that is, whether a patient can:

 

  • communicate choice about proposed treatment
  • understand her (his) medical situation
  • appreciate the situation and its consequences
  • manipulate information to reach a rational decision.1-3
  • manipulate information to reach a rational decision.
     

CASE REPORT continued
Although Ms. V’s health was deteriorating and her auditory hallucinations were becoming worse, she appeared insightful about her medical problems, understood her prognosis, and wanted comfort care. She understood that having multiple metastases meant a poor prognosis, and that a biopsy might yield a medical diagnosis. She stated, “If it were caught earlier and I was better able to tolerate treatment, it would make sense to know for sure, but now it doesn’t make sense. I just want to have no pain in my end.”
 

Misconceptions
In a study by Ganzini et al,4,5 395 consultation−liaison psychiatrists, geriatricians, and geriatric psychologists responded to a survey in which they rated types of misunderstandings by clinicians who refer patients for assessment of decision-making capacity. Seventy percent reported that it is common that, when a patient has a mental illness such as schizophrenia, practitioners think that the patient lacks capacity to make medical decisions. However, results of a meta-analysis by Jeste et al,6 in which the magnitude of impairment of decisional capacity in patients with schizophrenia was assessed in comparison to that of normal subjects, suggest that the presence of schizophrenia does not necessarily mean the patient has impaired capacity.

 

 

Voluntary participation in research. Many patients with schizophrenia volunteer to participate in clinical trials even when they are acutely psychotic and admitted to a psychiatric hospital. Given the importance placed on participants’ voluntary informed consent as a prerequisite for ethical conduct of research, the cognitive and emotional impairments associated with schizophrenia raise questions about patients’ capacity to consent.

As is true in other areas of functional capacity, the ability of patients with schizophrenia to make competent decisions relates more to their overall cognitive functioning than to the presence or absence of specific symptoms of the disorder.7 Documentation of longitudinal consent-related abilities among research participants with schizophrenia in the long-term Clinical Antipsychotic Trials of Intervention Effectiveness study indicated that most participants had stable or improved consent-related abilities. Although almost 25% of participants experienced substantial worsening, only 4% fell below the study’s capacity threshold for enrollment.8


What I learned from Ms. V
A diagnosis of schizophrenia does not automatically render a person unable to make decisions about medical care. Even patients who have severe mental illness might have significant intact areas of reality testing. Ethically, it is important to at least consider that the chronically mentally ill can understand treatment options and express consistent choices.

Healthcare providers might tend to exclude psychiatric patients from end-of-life decisions because they (1) are worried about the emotional fragility of such patients and (2) assume that patients lack capacity to participate in making such important decisions. The case presented here is an example of a patient with a severe psychiatric diagnosis being able to participate in her care despite her mental state.

 


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


1. Appelbaum PS, Grisso T. Assessing patients’ capacities to consent to treatment. N Engl J Med. 1988;319(25):1635-1638.
2. Leo RJ. Competency and the capacity to make treatment decisions: a primer for primary care physicians. Prim Care Companion J Clin Psychiatry. 1999;1(5):131-141.
3. White MM, Lofwall MR. Challenges of the capacity evaluation for the consultation-liaison psychiatrist. J Psychiatr Pract. 2015;21(2):160-170.
4. Ganzini L, Volicer L, Nelson W, et al. Pitfalls in assessment of decision-making capacity. Psychosomatics. 2003;44(3):237-243.
5. Ganzini L, Volicer L, Nelson WA, et al. Ten myths about decision-making capacity. J Am Med Dir Assoc. 2005;6(3):S100-S104.
6. Jeste DV, Depp CA, Palmer BW. Magnitude of impairment in decisional capacity in people with schizophrenia compared to normal subjects: an overview. Schizophr Bull. 2005;32(1):121-128.
7. Appelbaum PS. Decisional capacity of patients with schizophrenia to consent to research: taking stock. Schizophr Bull. 2005;32(1):22-25.
8. Stroup TS, Appelbaum PS, Gu H, et al. Longitudinal consent-related abilities among research participants with schizophrenia: results from the CATIE study. Schizophr Res. 2011;130(1-3):47-52.

References


1. Appelbaum PS, Grisso T. Assessing patients’ capacities to consent to treatment. N Engl J Med. 1988;319(25):1635-1638.
2. Leo RJ. Competency and the capacity to make treatment decisions: a primer for primary care physicians. Prim Care Companion J Clin Psychiatry. 1999;1(5):131-141.
3. White MM, Lofwall MR. Challenges of the capacity evaluation for the consultation-liaison psychiatrist. J Psychiatr Pract. 2015;21(2):160-170.
4. Ganzini L, Volicer L, Nelson W, et al. Pitfalls in assessment of decision-making capacity. Psychosomatics. 2003;44(3):237-243.
5. Ganzini L, Volicer L, Nelson WA, et al. Ten myths about decision-making capacity. J Am Med Dir Assoc. 2005;6(3):S100-S104.
6. Jeste DV, Depp CA, Palmer BW. Magnitude of impairment in decisional capacity in people with schizophrenia compared to normal subjects: an overview. Schizophr Bull. 2005;32(1):121-128.
7. Appelbaum PS. Decisional capacity of patients with schizophrenia to consent to research: taking stock. Schizophr Bull. 2005;32(1):22-25.
8. Stroup TS, Appelbaum PS, Gu H, et al. Longitudinal consent-related abilities among research participants with schizophrenia: results from the CATIE study. Schizophr Res. 2011;130(1-3):47-52.

Issue
Current Psychiatry - 15(4)
Issue
Current Psychiatry - 15(4)
Page Number
e4-e6
Page Number
e4-e6
Publications
Publications
Topics
Article Type
Display Headline
Don’t assume that psychiatric patients lack capacity to make decisions about care
Display Headline
Don’t assume that psychiatric patients lack capacity to make decisions about care
Legacy Keywords
capacity, severe mental illness, SMI, making decisions, evaluating capacity, schizophrenia. psychosis, psychotic disorder, psychotic disorders
Legacy Keywords
capacity, severe mental illness, SMI, making decisions, evaluating capacity, schizophrenia. psychosis, psychotic disorder, psychotic disorders
Sections
Disallow All Ads
Alternative CME
Article PDF Media

The ‘worried well’ and the ‘walking wounded’: How will we know them?; ‘Struggling with inner demons’

Article Type
Changed
Thu, 03/28/2019 - 15:10
Display Headline
The ‘worried well’ and the ‘walking wounded’: How will we know them?; ‘Struggling with inner demons’

The ‘worried well’ and the ‘walking wounded’: How will we know them?
One of Dr. Henry A. Nasrallah’s resolutions (16 New Year’s resolutions for psychiatrists in 2016, From the Editor, January 2016, p. 23,24) stated that a significant percentage of one’s practice should be dedicated to the sickest patients, followed by the statement, “There are enough non-physician mental health professionals to handle the walking wounded and worried well.”

Who are the “walking wounded” and the “worried well”? These are commonly used terms, but who falls into these categories? I think it is important to get a sense of who is in these groups, because my takeaway from this editorial is that it is acceptable to let the walking wounded and worried well be treated by lesser-trained clinicians. Do these terms refer to a diagnostic group? Level of functioning? Severity of symptoms? Or severity plus chronicity? Level of suffering? Ability to “fake” looking less severe? 

I wonder, am I a walking wounded or worried well? Are some of my friends, or my family members? When I see a patient, I ask myself if he (she) might be in that category.

Susan Fredriksen, MD
Private Practice
Hayesville, North Carolina


Dr. Nasrallah responds

I use those terms to refer to persons who have psychiatric symptoms but are not disabled socially or vocationally. They deserve a full psychiatric evaluation when they initially seek help, but generally do well with various types of psychotherapy, including cognitive-behavioral therapy, interpersonal therapy, psychodynamic therapy, or dialectic behavior therapy. There are many well-trained psychologists and licensed therapists who can administer those therapies as well as, or better than, some psychiatrists. 

I recommended that psychiatrists dedicate a significant percentage (more than 50%) of their practice to more severely ill patients (those with psychosis, bipolar disorder, major depressive disorder, panic disorders, obsessive-compulsive disorder, posttraumatic stress disorder, etc.) because we are the only mental health professionals who can competently integrate biopsychosocial treatments for these patients and administer pharmacotherapeutic agents in addition to non-drug approaches. The supply of psychiatrists is short, and the number of seriously ill patients who need the medical expertise we can provide is large.

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


‘Struggling with inner demons’

I would hope that Dr. Nasrallah would understand that the use of the metaphor, “struggling with inner demons,” does not suggest “stupid” (Stop blaming ‘demons’ for bizarre delusions or behavior!, From the Editor, February 2016, p. 19,20,22). A celebrity, or any other person, might be struggling with intense, conflicting emotions that create chaos and distress. I would shudder if I read in The New York Times, “Well known actor’s divorce and drug use clearly leading to hypertrophied amygdala.” The term inner demons does not necessarily imply medieval superstition, but rather a well-established use of creative language.

Ron Samarian, MD
Chief, Department of Psychiatry
William Beaumont Hospital
Royal Oak, Michigan
Chair, Oakland University
William Beaumont Medical School
Rochester, Michigan


Dr. Nasrallah responds
Dr. Samarian missed the reason for my umbrage with the “inner demons” metaphor. As a psychiatrist, educator, and researcher, I am exquisitely sensitive to the poor understanding of mental illness and the rampant stigma associated with psychiatric disorders despite the incredible neurobiologic advances. Thus, I regard the metaphor that employs words like “demons” when describing intense struggles with emotional upheavals and stress as having an unfortunate connotation to the obsolete beliefs that abnormal behavior, thoughts, or mood are due to the devil and his nefarious demons.

I would welcome a metaphor that describes a depressed person as having a shrunken hippocampus, which would regrow with antidepressant or electroconvulsive therapy, because that’s the biologic truth and has no misleading connotations; the same with Dr. Samarian’s example of a hypertrophied amygdala in a person with chronic stress.

References

Article PDF
Author and Disclosure Information

Issue
Current Psychiatry - 15(4)
Publications
Topics
Page Number
e3
Legacy Keywords
worried well and walking wounded, struggling with inner demons, letters to the editor, letter to the editor
Sections
Author and Disclosure Information

Author and Disclosure Information

Article PDF
Article PDF

The ‘worried well’ and the ‘walking wounded’: How will we know them?
One of Dr. Henry A. Nasrallah’s resolutions (16 New Year’s resolutions for psychiatrists in 2016, From the Editor, January 2016, p. 23,24) stated that a significant percentage of one’s practice should be dedicated to the sickest patients, followed by the statement, “There are enough non-physician mental health professionals to handle the walking wounded and worried well.”

Who are the “walking wounded” and the “worried well”? These are commonly used terms, but who falls into these categories? I think it is important to get a sense of who is in these groups, because my takeaway from this editorial is that it is acceptable to let the walking wounded and worried well be treated by lesser-trained clinicians. Do these terms refer to a diagnostic group? Level of functioning? Severity of symptoms? Or severity plus chronicity? Level of suffering? Ability to “fake” looking less severe? 

I wonder, am I a walking wounded or worried well? Are some of my friends, or my family members? When I see a patient, I ask myself if he (she) might be in that category.

Susan Fredriksen, MD
Private Practice
Hayesville, North Carolina


Dr. Nasrallah responds

I use those terms to refer to persons who have psychiatric symptoms but are not disabled socially or vocationally. They deserve a full psychiatric evaluation when they initially seek help, but generally do well with various types of psychotherapy, including cognitive-behavioral therapy, interpersonal therapy, psychodynamic therapy, or dialectic behavior therapy. There are many well-trained psychologists and licensed therapists who can administer those therapies as well as, or better than, some psychiatrists. 

I recommended that psychiatrists dedicate a significant percentage (more than 50%) of their practice to more severely ill patients (those with psychosis, bipolar disorder, major depressive disorder, panic disorders, obsessive-compulsive disorder, posttraumatic stress disorder, etc.) because we are the only mental health professionals who can competently integrate biopsychosocial treatments for these patients and administer pharmacotherapeutic agents in addition to non-drug approaches. The supply of psychiatrists is short, and the number of seriously ill patients who need the medical expertise we can provide is large.

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


‘Struggling with inner demons’

I would hope that Dr. Nasrallah would understand that the use of the metaphor, “struggling with inner demons,” does not suggest “stupid” (Stop blaming ‘demons’ for bizarre delusions or behavior!, From the Editor, February 2016, p. 19,20,22). A celebrity, or any other person, might be struggling with intense, conflicting emotions that create chaos and distress. I would shudder if I read in The New York Times, “Well known actor’s divorce and drug use clearly leading to hypertrophied amygdala.” The term inner demons does not necessarily imply medieval superstition, but rather a well-established use of creative language.

Ron Samarian, MD
Chief, Department of Psychiatry
William Beaumont Hospital
Royal Oak, Michigan
Chair, Oakland University
William Beaumont Medical School
Rochester, Michigan


Dr. Nasrallah responds
Dr. Samarian missed the reason for my umbrage with the “inner demons” metaphor. As a psychiatrist, educator, and researcher, I am exquisitely sensitive to the poor understanding of mental illness and the rampant stigma associated with psychiatric disorders despite the incredible neurobiologic advances. Thus, I regard the metaphor that employs words like “demons” when describing intense struggles with emotional upheavals and stress as having an unfortunate connotation to the obsolete beliefs that abnormal behavior, thoughts, or mood are due to the devil and his nefarious demons.

I would welcome a metaphor that describes a depressed person as having a shrunken hippocampus, which would regrow with antidepressant or electroconvulsive therapy, because that’s the biologic truth and has no misleading connotations; the same with Dr. Samarian’s example of a hypertrophied amygdala in a person with chronic stress.

The ‘worried well’ and the ‘walking wounded’: How will we know them?
One of Dr. Henry A. Nasrallah’s resolutions (16 New Year’s resolutions for psychiatrists in 2016, From the Editor, January 2016, p. 23,24) stated that a significant percentage of one’s practice should be dedicated to the sickest patients, followed by the statement, “There are enough non-physician mental health professionals to handle the walking wounded and worried well.”

Who are the “walking wounded” and the “worried well”? These are commonly used terms, but who falls into these categories? I think it is important to get a sense of who is in these groups, because my takeaway from this editorial is that it is acceptable to let the walking wounded and worried well be treated by lesser-trained clinicians. Do these terms refer to a diagnostic group? Level of functioning? Severity of symptoms? Or severity plus chronicity? Level of suffering? Ability to “fake” looking less severe? 

I wonder, am I a walking wounded or worried well? Are some of my friends, or my family members? When I see a patient, I ask myself if he (she) might be in that category.

Susan Fredriksen, MD
Private Practice
Hayesville, North Carolina


Dr. Nasrallah responds

I use those terms to refer to persons who have psychiatric symptoms but are not disabled socially or vocationally. They deserve a full psychiatric evaluation when they initially seek help, but generally do well with various types of psychotherapy, including cognitive-behavioral therapy, interpersonal therapy, psychodynamic therapy, or dialectic behavior therapy. There are many well-trained psychologists and licensed therapists who can administer those therapies as well as, or better than, some psychiatrists. 

I recommended that psychiatrists dedicate a significant percentage (more than 50%) of their practice to more severely ill patients (those with psychosis, bipolar disorder, major depressive disorder, panic disorders, obsessive-compulsive disorder, posttraumatic stress disorder, etc.) because we are the only mental health professionals who can competently integrate biopsychosocial treatments for these patients and administer pharmacotherapeutic agents in addition to non-drug approaches. The supply of psychiatrists is short, and the number of seriously ill patients who need the medical expertise we can provide is large.

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


‘Struggling with inner demons’

I would hope that Dr. Nasrallah would understand that the use of the metaphor, “struggling with inner demons,” does not suggest “stupid” (Stop blaming ‘demons’ for bizarre delusions or behavior!, From the Editor, February 2016, p. 19,20,22). A celebrity, or any other person, might be struggling with intense, conflicting emotions that create chaos and distress. I would shudder if I read in The New York Times, “Well known actor’s divorce and drug use clearly leading to hypertrophied amygdala.” The term inner demons does not necessarily imply medieval superstition, but rather a well-established use of creative language.

Ron Samarian, MD
Chief, Department of Psychiatry
William Beaumont Hospital
Royal Oak, Michigan
Chair, Oakland University
William Beaumont Medical School
Rochester, Michigan


Dr. Nasrallah responds
Dr. Samarian missed the reason for my umbrage with the “inner demons” metaphor. As a psychiatrist, educator, and researcher, I am exquisitely sensitive to the poor understanding of mental illness and the rampant stigma associated with psychiatric disorders despite the incredible neurobiologic advances. Thus, I regard the metaphor that employs words like “demons” when describing intense struggles with emotional upheavals and stress as having an unfortunate connotation to the obsolete beliefs that abnormal behavior, thoughts, or mood are due to the devil and his nefarious demons.

I would welcome a metaphor that describes a depressed person as having a shrunken hippocampus, which would regrow with antidepressant or electroconvulsive therapy, because that’s the biologic truth and has no misleading connotations; the same with Dr. Samarian’s example of a hypertrophied amygdala in a person with chronic stress.

References

References

Issue
Current Psychiatry - 15(4)
Issue
Current Psychiatry - 15(4)
Page Number
e3
Page Number
e3
Publications
Publications
Topics
Article Type
Display Headline
The ‘worried well’ and the ‘walking wounded’: How will we know them?; ‘Struggling with inner demons’
Display Headline
The ‘worried well’ and the ‘walking wounded’: How will we know them?; ‘Struggling with inner demons’
Legacy Keywords
worried well and walking wounded, struggling with inner demons, letters to the editor, letter to the editor
Legacy Keywords
worried well and walking wounded, struggling with inner demons, letters to the editor, letter to the editor
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Treat insomnia in depressed, even suicidal, people

Article Type
Changed
Mon, 06/17/2019 - 14:16
Display Headline
Treat insomnia in depressed, even suicidal, people

Author and Disclosure Information

Dr. Vaughn is Professor and Case Distinguished Chair,  Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta, Georgia.

Issue
Current Psychiatry - 15(4)
Publications
Topics
Legacy Keywords
suicide, suicidal, suicidal ideation, suicidal thoughts, depressed, depression, depressive disorders, depressive disorder, insomnia, sleep disorder, sleep disorders, fall asleep
Sections
Author and Disclosure Information

Dr. Vaughn is Professor and Case Distinguished Chair,  Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta, Georgia.

Author and Disclosure Information

Dr. Vaughn is Professor and Case Distinguished Chair,  Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta, Georgia.

Issue
Current Psychiatry - 15(4)
Issue
Current Psychiatry - 15(4)
Publications
Publications
Topics
Article Type
Display Headline
Treat insomnia in depressed, even suicidal, people
Display Headline
Treat insomnia in depressed, even suicidal, people
Legacy Keywords
suicide, suicidal, suicidal ideation, suicidal thoughts, depressed, depression, depressive disorders, depressive disorder, insomnia, sleep disorder, sleep disorders, fall asleep
Legacy Keywords
suicide, suicidal, suicidal ideation, suicidal thoughts, depressed, depression, depressive disorders, depressive disorder, insomnia, sleep disorder, sleep disorders, fall asleep
Sections
Disallow All Ads
Alternative CME
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

Antipsychotic polypharmacy back to monotherapy

Article Type
Changed
Fri, 09/28/2018 - 09:56
Display Headline
Antipsychotic polypharmacy back to monotherapy

References


1. Godleski LS, Kerler R, Barber JW, et al. Multiple versus single antipsychotic drug treatment in chronic psychosis. J Nerv Ment Dis. 1989;177(11):686-689.
2. Suzuki T, Uchida H, Tanaka KF, et al. Revising polypharmacy to a single antipsychotic regimen for patients with chronic schizophrenia. Int J Neuropsychopharmacol. 2004;7(2):133-142.
3. Essock SM, Schooler NR, Stroup TS, et al;
Schizophrenia Trials Network. Effectiveness of switching from antipsychotic polypharmacy to monotherapy. Am J Psychiatry. 2011;168(7):702-708.
4. Hori H, Yoshimura R, Katsuki A, et al. Switching to antipsychotic monotherapy can improve attention and processing speed, and social activity in chronic schizophrenia patients. J Psychiatr Res. 2013;47(12):1843-1848.
5. Constantine RJ. Andel R. McPherson M, et al. The risks and benefits of switching patients with schizophrenia or schizoaffective disorder from two to one antipsychotic medication: a randomized controlled trial. Schizophr Res. 2015;166(1-3):194-200.

Author and Disclosure Information

Dr. Graham is Clinical Pharmacy Specialist–Mental Health, Veterans Affairs, San Diego Healthcare System, San Diego, California.

Issue
Current Psychiatry - 15(4)
Publications
Topics
Legacy Keywords
antipsychotic, antipsychotics, polypharmacy, monotherapy, psychotic disorder, psychotic disorders, schizophrenia, psychosis, psychotic
Sections
Author and Disclosure Information

Dr. Graham is Clinical Pharmacy Specialist–Mental Health, Veterans Affairs, San Diego Healthcare System, San Diego, California.

Author and Disclosure Information

Dr. Graham is Clinical Pharmacy Specialist–Mental Health, Veterans Affairs, San Diego Healthcare System, San Diego, California.

References


1. Godleski LS, Kerler R, Barber JW, et al. Multiple versus single antipsychotic drug treatment in chronic psychosis. J Nerv Ment Dis. 1989;177(11):686-689.
2. Suzuki T, Uchida H, Tanaka KF, et al. Revising polypharmacy to a single antipsychotic regimen for patients with chronic schizophrenia. Int J Neuropsychopharmacol. 2004;7(2):133-142.
3. Essock SM, Schooler NR, Stroup TS, et al;
Schizophrenia Trials Network. Effectiveness of switching from antipsychotic polypharmacy to monotherapy. Am J Psychiatry. 2011;168(7):702-708.
4. Hori H, Yoshimura R, Katsuki A, et al. Switching to antipsychotic monotherapy can improve attention and processing speed, and social activity in chronic schizophrenia patients. J Psychiatr Res. 2013;47(12):1843-1848.
5. Constantine RJ. Andel R. McPherson M, et al. The risks and benefits of switching patients with schizophrenia or schizoaffective disorder from two to one antipsychotic medication: a randomized controlled trial. Schizophr Res. 2015;166(1-3):194-200.

References


1. Godleski LS, Kerler R, Barber JW, et al. Multiple versus single antipsychotic drug treatment in chronic psychosis. J Nerv Ment Dis. 1989;177(11):686-689.
2. Suzuki T, Uchida H, Tanaka KF, et al. Revising polypharmacy to a single antipsychotic regimen for patients with chronic schizophrenia. Int J Neuropsychopharmacol. 2004;7(2):133-142.
3. Essock SM, Schooler NR, Stroup TS, et al;
Schizophrenia Trials Network. Effectiveness of switching from antipsychotic polypharmacy to monotherapy. Am J Psychiatry. 2011;168(7):702-708.
4. Hori H, Yoshimura R, Katsuki A, et al. Switching to antipsychotic monotherapy can improve attention and processing speed, and social activity in chronic schizophrenia patients. J Psychiatr Res. 2013;47(12):1843-1848.
5. Constantine RJ. Andel R. McPherson M, et al. The risks and benefits of switching patients with schizophrenia or schizoaffective disorder from two to one antipsychotic medication: a randomized controlled trial. Schizophr Res. 2015;166(1-3):194-200.

Issue
Current Psychiatry - 15(4)
Issue
Current Psychiatry - 15(4)
Publications
Publications
Topics
Article Type
Display Headline
Antipsychotic polypharmacy back to monotherapy
Display Headline
Antipsychotic polypharmacy back to monotherapy
Legacy Keywords
antipsychotic, antipsychotics, polypharmacy, monotherapy, psychotic disorder, psychotic disorders, schizophrenia, psychosis, psychotic
Legacy Keywords
antipsychotic, antipsychotics, polypharmacy, monotherapy, psychotic disorder, psychotic disorders, schizophrenia, psychosis, psychotic
Sections
Disallow All Ads
Alternative CME
Use ProPublica

Manic after taking a vacation

Article Type
Changed
Tue, 12/11/2018 - 15:29
Display Headline
Manic after taking a vacation

CASE From soft-spoken to manic
Mr. K, age 36, an Asian male with no psychiatric history, arrives at the outpatient psychiatry clinic accompanied by his wife, after being referred from the emergency room the night before. He reports racing thoughts, euphoric mood, increased speech, hypergraphia, elevated self-esteem, decreased need for sleep, distractibility, and increased goal-directed activity. Notably, Mr. K states that he likes how he is feeling.

Mr. K’s wife says that his condition is a clear change from his baseline demeanor: soft-spoken and mild-mannered.

Mr. K reports that his symptoms started approximately 10 days earlier, after he returned from a cruise with his wife. During the cruise, he used a scopolamine patch to prevent motion sickness. Mr. K and his wife say that they believe that the scopolamine patch caused his symptoms.

Can scopolamine cause mania?
   a) No
   b) Yes; this is well-documented in the literature
   c) It is theoretically possible because of scopolamine’s antidepressant and central anticholinergic effects


TREATMENT
Lithium, close follow up
Mr. K has no history of psychiatric illness or substance use and no recent use of psycho­active substances—other than scopolamine—that could trigger a manic episode. His family history is significant for a younger brother who had a single manic episode at age 12 and a suicide attempt as a young adult.

Mr. K works full-time on rotating shifts—including some overnight shifts—as a manufacturing supervisor at a biotechnology company. He has been unable to work since returning from the cruise because of his psychiatric symptoms.

Mr. K is started on sustained-release (SR) lithium, 900 mg/d. In addition, the psychiatrist advises Mr. K to continue taking clonazepam, 0.5 to 1 mg as needed, which the emergency medicine physician prescribed, for insomnia. Mr. K is referred to a psychiatric intensive outpatient program (IOP), 3 days a week for 2 weeks, and is advised to stay home from work until symptoms stabilize.

Mr. K follows up closely with the psychiatrist in the clinic, every 1 to 2 weeks for the first month, as well as by several telephone and e-mail contacts. Lithium SR is titrated to 1,200 mg/d, to a therapeutic serum level of 1.1 mEq/L. Clonazepam is switched to quetiapine, 25 to 50 mg as needed, to address ongoing insomnia and to reduce the risk of dependency on clonazepam.

Mr. K’s mania gradually abates. He finishes the IOP and returns to work 3 weeks after his initial presentation. At an office visit, Mr. K’s wife gives the psychiatrist 2 scientific articles documenting the antidepressant effect of scopolamine.1,2 Mr. K and his wife both continue to believe that Mr. K’s manic episode was triggered by the scopolamine patch he used while on the cruise. They think this is important because Mr. K believes he would not have developed mania otherwise, and he does not want to take a mood stabilizer for the rest of his life.


The author’s observations

There are several proposed mechanisms for scopolamine’s antidepressant effect (Table 1).3-9 Scopolamine blocks central muscarinic cholinergic receptors, which reduces production of glutamate receptors and leads to reduced glutamate transmission and neurotoxicity.3,4 Scopolamine—similar to ketamine—could enhance synaptogenesis and synaptic signaling.5,6 Also, by blocking muscarinic autoreceptors, scopolamine results in an acute upregulation of acetylcholine release, which, in turn, influences the nicotinic, dopamine, serotonin, and neuropeptide Y systems. This action could contribute to anti-inflammatory effects, all of which can benefit mood.7-9 These antidepressant mechanisms also could explain why, theoretically, scopolamine could precipitate mania in a person predisposed to mental illness.

Proposed by Janowsky et al10 in 1972, the cholinergic−adrenergic balance hypothesis of affective disorders suggests that depression represents an excess of central cholinergic tone over adrenergic tone, and that mania represents the opposite imbalance. Several lines of evidence in the literature support this theory. For example, depressed patients have been found to have hypersensitive central cholinergic receptors.11,12 Also, central cholinesterase inhibition has been shown to affect pituitary hormone and epinephrine levels via central muscarinic receptors.13 In addition, scopolamine has been shown to attenuate these effects via the central anti-muscarinic mechanism.14

Rapid antidepressant therapy. Scopolamine is being studied as a rapid antidepressant treatment, although it usually is administered via IV infusion, rather than patch form, in trials.15-17 IV ketamine is another therapy being investigated for rapid treatment of depression, which might have downstream mechanisms of action related to scopolamine.5,18 Electroconvulsive therapy is a well-known for its quick antidepressant effect, which could involve synaptogenesis or effects on the neuroendocrine system.19 Sleep deprivation also can produce a rapid antidepressant effect20 (Table 21,2,5,6,15,16,18-20).



OUTCOME
Prone to motion sickness
Approximately 3.5 months after his initial presentation, Mr. K continues to do well with treatment. He is euthymic and functioning well at work. He and his wife are preparing for the birth of their first child.

 

 

Mr. K is prone to motion sickness, and asks if he can take over-the-counter dimenhydrinate tablets for long car rides. He reports that dimenhydrinate has worked well for him in the past without triggering manic episodes, and he did not anticipate needing to take it very often.

What would you tell Mr. K about dimenhydrinate for motion sickness during car rides?
   a) Mr. K should not take dimenhydrinate to prevent motion sickness because he experienced a manic episode triggered by a scopolamine patch
   b) Mr. K can use dimenhydrinate as much as he wants to prevent motion sickness because it poses no risk of mania
   c) Mr. K can use dimenhydrinate with caution and sparingly on a trial basis, as long as he is taking his mood stabilizer


FOLLOW UP Cautious use
The psychiatrist advised Mr. K to take dimenhydrinate cautiously when needed for long car rides. The psychiatrist feels this is safe because Mr. K is taking a mood stabilizer (lithium). Also, although dimenhydrinate has anticholinergic properties, occasional use is thought to pose less risk of triggering mania than the constant anticholinergic exposure over several days with a scopolamine patch. (The scopolamine patch contains 1.5 mg of the drug delivered over 3 days [ie, 0.5 mg/d]. In trials of IV scopolamine for depression, the dosage was 0.4 mcg/kg/d administered over 3 consecutive days.15-17 For an adult weighing 70 kg, this would be equivalent to 0.24 mg/d. Therefore, using a scopolamine patch over 3 days would appear to deliver a robust antidepressant-level dosage, even taking into account possible lower bioavailability with transdermal administration compared with IV infusion.)

The psychiatrist concludes that sporadic use of dimenhydrinate tablets for motion sickness during occasional long car rides poses less of a risk for Mr. K of triggering mania than repeat use of a scopolamine patch.


The author’s observations

Mr. K’s case is notable for several reasons:

  • Novelty. This might be the first report of scopolamine-induced mania in the literature. In clinical trials by Furey and Drevets,15 Drevets and Furey,16 and Ellis et al,17 no study participants who received scopolamine infusion developed mania or hypomania. Although it is possible that Mr. K’s manic episode could have occurred spontaneously and was coincidental to his scopolamine use, there are valid reasons why scopolamine could trigger mania in a vulnerable person.
  • Biochemical insight. The case underscores the role of the muscarinic cholinergic system in regulating mood.10
  • Rational medical care. Sensible clinical decision-making was needed when Mr. K asked about using dimenhydrinate for motion sickness during car rides. Although there might not be definitively correct answers for questions that arose during Mr. K’s care (in the absence of research literature), theoretical understanding of the antidepressant effects of anticholinergic medications helped inform the psychiatrist’s responses to Mr. K and his wife.


Bottom Line
Scopolamine, often applied via a transdermal patch to treat motion sickness, has some antidepressant effect, which could lead to mania in a person predisposed to mental illness. The drug’s action on central muscarinic cholinergic receptors could be responsible for its effect on mood. Mood stabilizers and stopping the offending agent are mainstay treatments for scopolamine-induced mania.


Related Resources

  • Furey ML, Zarate CA Jr. Pulsed intravenous administration of scopolamine produces rapid antidepressant effects and modest side effects. J Clin Psychiatry. 2013;74(8):850-851.

  • Khajavi D, Farokhnia M, Modabbernia A, et al. Oral scopolamine augmentation in moderate to severe major depressive disorder: a randomized, double-blind, placebo-controlled study. J Clin Psychiatry. 2012;73(11):1428-1433.

Drug Brand Names
Clonazepam • Klonopin
Dimenhydrinate • Dramamine
Lithium • Eskalith, Lithobid
Quetiapine • Seroquel

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


1. Drevets WC, Zarate CA Jr, Furey ML. Antidepressant effects of the muscarinic cholinergic receptor antagonist scopolamine: a review. Biol Psychiatry. 2013;73(12):1156-1163.
2. Jaffe RJ, Novakovic V, Peselow ED. Scopolamine as an antidepressant: a systematic review. Clin Neuropharmacol. 2013;36(1):24-26.
3. Rami A, Ausmeir F, Winckler J, et al. Differential effects of scopolamine on neuronal survival in ischemia and glutamate neurotoxicity: relationships to the excessive vulnerability of the dorsoseptal hippocampus. J Chem Neuroanat. 1997;13(3):201-208.
4. Benveniste M, Wilhelm J, Dingledine RJ, et al. Subunit-dependent modulation of kainate receptors by muscarinic acetylcholine receptors. Brain Res. 2010;1352:61-69.
5. Duman RS, Li N, Liu RJ, et al. Signaling pathways underlying the rapid antidepressant actions of ketamine. Neuropharmacology. 2012;62(1):35-41.
6. Voleti B, Navarria A, Liu R, et al. Scopolamine rapidly increases mammalian target of rapamycin complex 1 signaling, synaptogenesis, and antidepressant behavioral responses. Biol Psychiatry. 2013;74(10):742-749.
7. Overstreet DH, Friedman E, Mathé AA, et al. The Flinders Sensitive Line rat: a selectively bred putative animal model of depression. Neurosci Biobehav Rev. 2005;29(4-5):739-759.
8. Tizabi Y, Getachew B, Rezvani AH, et al. Antidepressant-like effects of nicotine and reduced nicotinic receptor binding in the Fawn-Hooded rat, an animal model of co-morbid depression and alcoholism. Prog Neuropsychopharmacol Biol Psychiatry. 2009;33(3):398-402.
9. Wang DW, Zhou RB, Yao YM. Role of cholinergic anti-inflammatory pathway in regulating host response and its interventional strategy for inflammatory diseases. Chin J Traumatol. 2009;12(6):355-364.
10. Janowsky DS, el-Yousef MK, Davis JM, et al. A cholinergic-adrenergic hypothesis of mania and depression. Lancet. 1972;2(7778):632-635.
11. Risch SC, Kalin NH, Janowsky DS. Cholinergic challenges in affective illness: behavioral and neuroendocrine correlates. J Clin Psychopharmacol. 1981;1(4):186-192.
12. Risch SC, Janowsky DS, Gillin JC. Muscarinic supersensitivity of anterior pituitary ACTH and β-endorphin release in major depressive illness. Peptides. 1983;4(5):789-792.
13. Risch SC, Janowsky DS, Mott MA, et al. Central and peripheral cholinesterase inhibition: effects on anterior pituitary and sympathomimetic function. Psychoneuroendocrinology. 1986;11(2):221-230.
14. Janowsky DS, Risch SC, Kennedy B, et al. Central muscarinic effects of physostigmine on mood, cardiovascular function, pituitary and adrenal neuroendocrine release. Psychopharmacology (Berl). 1986;89(2):150-154.
15. Furey ML, Drevets WC. Antidepressant efficacy of the antimuscarinic drug scopolamine: a randomized, placebo-controlled trial. Arch Gen Psychiatry. 2006;63(10):1121-1129.
16. Drevets WC, Furey ML. Replication of scopolamine’s antidepressant efficacy in major depressive disorder: a randomized, placebo-controlled clinical trial. Biol Psychiatry. 2010;67(5):432-438.
17. Ellis JS, Zarate CA Jr, Luckenbaugh DA, et al. Antidepressant treatment history as a predictor of response to scopolamine: clinical implications. J Affect Disord. 2014;162:39-42.
18. Newport DJ, Carpenter LL, McDonald WM, et al. Ketamine and other NMDA antagonists: early clinical trials and possible mechanisms in depression. Am J Psychiatry. 2015;172(10):950-966.
19. Bolwig TG. How does electroconvulsive therapy work? Theories on its mechanism. Can J Psychiatry. 2011;56(1):13-18.
20. Wu JC, Kelsoe JR, Schachat C, et al. Rapid and sustained antidepressant response with sleep deprivation and chronotherapy in bipolar disorder. Biol Psychiatry. 2009;66(3):298-301.

Article PDF
Author and Disclosure Information

Emjay Tan, MD
psychiatrist at Kaiser Permanente
San Bruno, California

Issue
Current Psychiatry - 15(4)
Publications
Topics
Page Number
45-49
Legacy Keywords
psychosis, psychotic episode, motion-sickness patch, scopolamine, dimenhydrinate, psychotic disorder, psychotic disorders, mania, manic, euphoric, racing thoughts, lithium
Sections
Author and Disclosure Information

Emjay Tan, MD
psychiatrist at Kaiser Permanente
San Bruno, California

Author and Disclosure Information

Emjay Tan, MD
psychiatrist at Kaiser Permanente
San Bruno, California

Article PDF
Article PDF

CASE From soft-spoken to manic
Mr. K, age 36, an Asian male with no psychiatric history, arrives at the outpatient psychiatry clinic accompanied by his wife, after being referred from the emergency room the night before. He reports racing thoughts, euphoric mood, increased speech, hypergraphia, elevated self-esteem, decreased need for sleep, distractibility, and increased goal-directed activity. Notably, Mr. K states that he likes how he is feeling.

Mr. K’s wife says that his condition is a clear change from his baseline demeanor: soft-spoken and mild-mannered.

Mr. K reports that his symptoms started approximately 10 days earlier, after he returned from a cruise with his wife. During the cruise, he used a scopolamine patch to prevent motion sickness. Mr. K and his wife say that they believe that the scopolamine patch caused his symptoms.

Can scopolamine cause mania?
   a) No
   b) Yes; this is well-documented in the literature
   c) It is theoretically possible because of scopolamine’s antidepressant and central anticholinergic effects


TREATMENT
Lithium, close follow up
Mr. K has no history of psychiatric illness or substance use and no recent use of psycho­active substances—other than scopolamine—that could trigger a manic episode. His family history is significant for a younger brother who had a single manic episode at age 12 and a suicide attempt as a young adult.

Mr. K works full-time on rotating shifts—including some overnight shifts—as a manufacturing supervisor at a biotechnology company. He has been unable to work since returning from the cruise because of his psychiatric symptoms.

Mr. K is started on sustained-release (SR) lithium, 900 mg/d. In addition, the psychiatrist advises Mr. K to continue taking clonazepam, 0.5 to 1 mg as needed, which the emergency medicine physician prescribed, for insomnia. Mr. K is referred to a psychiatric intensive outpatient program (IOP), 3 days a week for 2 weeks, and is advised to stay home from work until symptoms stabilize.

Mr. K follows up closely with the psychiatrist in the clinic, every 1 to 2 weeks for the first month, as well as by several telephone and e-mail contacts. Lithium SR is titrated to 1,200 mg/d, to a therapeutic serum level of 1.1 mEq/L. Clonazepam is switched to quetiapine, 25 to 50 mg as needed, to address ongoing insomnia and to reduce the risk of dependency on clonazepam.

Mr. K’s mania gradually abates. He finishes the IOP and returns to work 3 weeks after his initial presentation. At an office visit, Mr. K’s wife gives the psychiatrist 2 scientific articles documenting the antidepressant effect of scopolamine.1,2 Mr. K and his wife both continue to believe that Mr. K’s manic episode was triggered by the scopolamine patch he used while on the cruise. They think this is important because Mr. K believes he would not have developed mania otherwise, and he does not want to take a mood stabilizer for the rest of his life.


The author’s observations

There are several proposed mechanisms for scopolamine’s antidepressant effect (Table 1).3-9 Scopolamine blocks central muscarinic cholinergic receptors, which reduces production of glutamate receptors and leads to reduced glutamate transmission and neurotoxicity.3,4 Scopolamine—similar to ketamine—could enhance synaptogenesis and synaptic signaling.5,6 Also, by blocking muscarinic autoreceptors, scopolamine results in an acute upregulation of acetylcholine release, which, in turn, influences the nicotinic, dopamine, serotonin, and neuropeptide Y systems. This action could contribute to anti-inflammatory effects, all of which can benefit mood.7-9 These antidepressant mechanisms also could explain why, theoretically, scopolamine could precipitate mania in a person predisposed to mental illness.

Proposed by Janowsky et al10 in 1972, the cholinergic−adrenergic balance hypothesis of affective disorders suggests that depression represents an excess of central cholinergic tone over adrenergic tone, and that mania represents the opposite imbalance. Several lines of evidence in the literature support this theory. For example, depressed patients have been found to have hypersensitive central cholinergic receptors.11,12 Also, central cholinesterase inhibition has been shown to affect pituitary hormone and epinephrine levels via central muscarinic receptors.13 In addition, scopolamine has been shown to attenuate these effects via the central anti-muscarinic mechanism.14

Rapid antidepressant therapy. Scopolamine is being studied as a rapid antidepressant treatment, although it usually is administered via IV infusion, rather than patch form, in trials.15-17 IV ketamine is another therapy being investigated for rapid treatment of depression, which might have downstream mechanisms of action related to scopolamine.5,18 Electroconvulsive therapy is a well-known for its quick antidepressant effect, which could involve synaptogenesis or effects on the neuroendocrine system.19 Sleep deprivation also can produce a rapid antidepressant effect20 (Table 21,2,5,6,15,16,18-20).



OUTCOME
Prone to motion sickness
Approximately 3.5 months after his initial presentation, Mr. K continues to do well with treatment. He is euthymic and functioning well at work. He and his wife are preparing for the birth of their first child.

 

 

Mr. K is prone to motion sickness, and asks if he can take over-the-counter dimenhydrinate tablets for long car rides. He reports that dimenhydrinate has worked well for him in the past without triggering manic episodes, and he did not anticipate needing to take it very often.

What would you tell Mr. K about dimenhydrinate for motion sickness during car rides?
   a) Mr. K should not take dimenhydrinate to prevent motion sickness because he experienced a manic episode triggered by a scopolamine patch
   b) Mr. K can use dimenhydrinate as much as he wants to prevent motion sickness because it poses no risk of mania
   c) Mr. K can use dimenhydrinate with caution and sparingly on a trial basis, as long as he is taking his mood stabilizer


FOLLOW UP Cautious use
The psychiatrist advised Mr. K to take dimenhydrinate cautiously when needed for long car rides. The psychiatrist feels this is safe because Mr. K is taking a mood stabilizer (lithium). Also, although dimenhydrinate has anticholinergic properties, occasional use is thought to pose less risk of triggering mania than the constant anticholinergic exposure over several days with a scopolamine patch. (The scopolamine patch contains 1.5 mg of the drug delivered over 3 days [ie, 0.5 mg/d]. In trials of IV scopolamine for depression, the dosage was 0.4 mcg/kg/d administered over 3 consecutive days.15-17 For an adult weighing 70 kg, this would be equivalent to 0.24 mg/d. Therefore, using a scopolamine patch over 3 days would appear to deliver a robust antidepressant-level dosage, even taking into account possible lower bioavailability with transdermal administration compared with IV infusion.)

The psychiatrist concludes that sporadic use of dimenhydrinate tablets for motion sickness during occasional long car rides poses less of a risk for Mr. K of triggering mania than repeat use of a scopolamine patch.


The author’s observations

Mr. K’s case is notable for several reasons:

  • Novelty. This might be the first report of scopolamine-induced mania in the literature. In clinical trials by Furey and Drevets,15 Drevets and Furey,16 and Ellis et al,17 no study participants who received scopolamine infusion developed mania or hypomania. Although it is possible that Mr. K’s manic episode could have occurred spontaneously and was coincidental to his scopolamine use, there are valid reasons why scopolamine could trigger mania in a vulnerable person.
  • Biochemical insight. The case underscores the role of the muscarinic cholinergic system in regulating mood.10
  • Rational medical care. Sensible clinical decision-making was needed when Mr. K asked about using dimenhydrinate for motion sickness during car rides. Although there might not be definitively correct answers for questions that arose during Mr. K’s care (in the absence of research literature), theoretical understanding of the antidepressant effects of anticholinergic medications helped inform the psychiatrist’s responses to Mr. K and his wife.


Bottom Line
Scopolamine, often applied via a transdermal patch to treat motion sickness, has some antidepressant effect, which could lead to mania in a person predisposed to mental illness. The drug’s action on central muscarinic cholinergic receptors could be responsible for its effect on mood. Mood stabilizers and stopping the offending agent are mainstay treatments for scopolamine-induced mania.


Related Resources

  • Furey ML, Zarate CA Jr. Pulsed intravenous administration of scopolamine produces rapid antidepressant effects and modest side effects. J Clin Psychiatry. 2013;74(8):850-851.

  • Khajavi D, Farokhnia M, Modabbernia A, et al. Oral scopolamine augmentation in moderate to severe major depressive disorder: a randomized, double-blind, placebo-controlled study. J Clin Psychiatry. 2012;73(11):1428-1433.

Drug Brand Names
Clonazepam • Klonopin
Dimenhydrinate • Dramamine
Lithium • Eskalith, Lithobid
Quetiapine • Seroquel

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

CASE From soft-spoken to manic
Mr. K, age 36, an Asian male with no psychiatric history, arrives at the outpatient psychiatry clinic accompanied by his wife, after being referred from the emergency room the night before. He reports racing thoughts, euphoric mood, increased speech, hypergraphia, elevated self-esteem, decreased need for sleep, distractibility, and increased goal-directed activity. Notably, Mr. K states that he likes how he is feeling.

Mr. K’s wife says that his condition is a clear change from his baseline demeanor: soft-spoken and mild-mannered.

Mr. K reports that his symptoms started approximately 10 days earlier, after he returned from a cruise with his wife. During the cruise, he used a scopolamine patch to prevent motion sickness. Mr. K and his wife say that they believe that the scopolamine patch caused his symptoms.

Can scopolamine cause mania?
   a) No
   b) Yes; this is well-documented in the literature
   c) It is theoretically possible because of scopolamine’s antidepressant and central anticholinergic effects


TREATMENT
Lithium, close follow up
Mr. K has no history of psychiatric illness or substance use and no recent use of psycho­active substances—other than scopolamine—that could trigger a manic episode. His family history is significant for a younger brother who had a single manic episode at age 12 and a suicide attempt as a young adult.

Mr. K works full-time on rotating shifts—including some overnight shifts—as a manufacturing supervisor at a biotechnology company. He has been unable to work since returning from the cruise because of his psychiatric symptoms.

Mr. K is started on sustained-release (SR) lithium, 900 mg/d. In addition, the psychiatrist advises Mr. K to continue taking clonazepam, 0.5 to 1 mg as needed, which the emergency medicine physician prescribed, for insomnia. Mr. K is referred to a psychiatric intensive outpatient program (IOP), 3 days a week for 2 weeks, and is advised to stay home from work until symptoms stabilize.

Mr. K follows up closely with the psychiatrist in the clinic, every 1 to 2 weeks for the first month, as well as by several telephone and e-mail contacts. Lithium SR is titrated to 1,200 mg/d, to a therapeutic serum level of 1.1 mEq/L. Clonazepam is switched to quetiapine, 25 to 50 mg as needed, to address ongoing insomnia and to reduce the risk of dependency on clonazepam.

Mr. K’s mania gradually abates. He finishes the IOP and returns to work 3 weeks after his initial presentation. At an office visit, Mr. K’s wife gives the psychiatrist 2 scientific articles documenting the antidepressant effect of scopolamine.1,2 Mr. K and his wife both continue to believe that Mr. K’s manic episode was triggered by the scopolamine patch he used while on the cruise. They think this is important because Mr. K believes he would not have developed mania otherwise, and he does not want to take a mood stabilizer for the rest of his life.


The author’s observations

There are several proposed mechanisms for scopolamine’s antidepressant effect (Table 1).3-9 Scopolamine blocks central muscarinic cholinergic receptors, which reduces production of glutamate receptors and leads to reduced glutamate transmission and neurotoxicity.3,4 Scopolamine—similar to ketamine—could enhance synaptogenesis and synaptic signaling.5,6 Also, by blocking muscarinic autoreceptors, scopolamine results in an acute upregulation of acetylcholine release, which, in turn, influences the nicotinic, dopamine, serotonin, and neuropeptide Y systems. This action could contribute to anti-inflammatory effects, all of which can benefit mood.7-9 These antidepressant mechanisms also could explain why, theoretically, scopolamine could precipitate mania in a person predisposed to mental illness.

Proposed by Janowsky et al10 in 1972, the cholinergic−adrenergic balance hypothesis of affective disorders suggests that depression represents an excess of central cholinergic tone over adrenergic tone, and that mania represents the opposite imbalance. Several lines of evidence in the literature support this theory. For example, depressed patients have been found to have hypersensitive central cholinergic receptors.11,12 Also, central cholinesterase inhibition has been shown to affect pituitary hormone and epinephrine levels via central muscarinic receptors.13 In addition, scopolamine has been shown to attenuate these effects via the central anti-muscarinic mechanism.14

Rapid antidepressant therapy. Scopolamine is being studied as a rapid antidepressant treatment, although it usually is administered via IV infusion, rather than patch form, in trials.15-17 IV ketamine is another therapy being investigated for rapid treatment of depression, which might have downstream mechanisms of action related to scopolamine.5,18 Electroconvulsive therapy is a well-known for its quick antidepressant effect, which could involve synaptogenesis or effects on the neuroendocrine system.19 Sleep deprivation also can produce a rapid antidepressant effect20 (Table 21,2,5,6,15,16,18-20).



OUTCOME
Prone to motion sickness
Approximately 3.5 months after his initial presentation, Mr. K continues to do well with treatment. He is euthymic and functioning well at work. He and his wife are preparing for the birth of their first child.

 

 

Mr. K is prone to motion sickness, and asks if he can take over-the-counter dimenhydrinate tablets for long car rides. He reports that dimenhydrinate has worked well for him in the past without triggering manic episodes, and he did not anticipate needing to take it very often.

What would you tell Mr. K about dimenhydrinate for motion sickness during car rides?
   a) Mr. K should not take dimenhydrinate to prevent motion sickness because he experienced a manic episode triggered by a scopolamine patch
   b) Mr. K can use dimenhydrinate as much as he wants to prevent motion sickness because it poses no risk of mania
   c) Mr. K can use dimenhydrinate with caution and sparingly on a trial basis, as long as he is taking his mood stabilizer


FOLLOW UP Cautious use
The psychiatrist advised Mr. K to take dimenhydrinate cautiously when needed for long car rides. The psychiatrist feels this is safe because Mr. K is taking a mood stabilizer (lithium). Also, although dimenhydrinate has anticholinergic properties, occasional use is thought to pose less risk of triggering mania than the constant anticholinergic exposure over several days with a scopolamine patch. (The scopolamine patch contains 1.5 mg of the drug delivered over 3 days [ie, 0.5 mg/d]. In trials of IV scopolamine for depression, the dosage was 0.4 mcg/kg/d administered over 3 consecutive days.15-17 For an adult weighing 70 kg, this would be equivalent to 0.24 mg/d. Therefore, using a scopolamine patch over 3 days would appear to deliver a robust antidepressant-level dosage, even taking into account possible lower bioavailability with transdermal administration compared with IV infusion.)

The psychiatrist concludes that sporadic use of dimenhydrinate tablets for motion sickness during occasional long car rides poses less of a risk for Mr. K of triggering mania than repeat use of a scopolamine patch.


The author’s observations

Mr. K’s case is notable for several reasons:

  • Novelty. This might be the first report of scopolamine-induced mania in the literature. In clinical trials by Furey and Drevets,15 Drevets and Furey,16 and Ellis et al,17 no study participants who received scopolamine infusion developed mania or hypomania. Although it is possible that Mr. K’s manic episode could have occurred spontaneously and was coincidental to his scopolamine use, there are valid reasons why scopolamine could trigger mania in a vulnerable person.
  • Biochemical insight. The case underscores the role of the muscarinic cholinergic system in regulating mood.10
  • Rational medical care. Sensible clinical decision-making was needed when Mr. K asked about using dimenhydrinate for motion sickness during car rides. Although there might not be definitively correct answers for questions that arose during Mr. K’s care (in the absence of research literature), theoretical understanding of the antidepressant effects of anticholinergic medications helped inform the psychiatrist’s responses to Mr. K and his wife.


Bottom Line
Scopolamine, often applied via a transdermal patch to treat motion sickness, has some antidepressant effect, which could lead to mania in a person predisposed to mental illness. The drug’s action on central muscarinic cholinergic receptors could be responsible for its effect on mood. Mood stabilizers and stopping the offending agent are mainstay treatments for scopolamine-induced mania.


Related Resources

  • Furey ML, Zarate CA Jr. Pulsed intravenous administration of scopolamine produces rapid antidepressant effects and modest side effects. J Clin Psychiatry. 2013;74(8):850-851.

  • Khajavi D, Farokhnia M, Modabbernia A, et al. Oral scopolamine augmentation in moderate to severe major depressive disorder: a randomized, double-blind, placebo-controlled study. J Clin Psychiatry. 2012;73(11):1428-1433.

Drug Brand Names
Clonazepam • Klonopin
Dimenhydrinate • Dramamine
Lithium • Eskalith, Lithobid
Quetiapine • Seroquel

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


1. Drevets WC, Zarate CA Jr, Furey ML. Antidepressant effects of the muscarinic cholinergic receptor antagonist scopolamine: a review. Biol Psychiatry. 2013;73(12):1156-1163.
2. Jaffe RJ, Novakovic V, Peselow ED. Scopolamine as an antidepressant: a systematic review. Clin Neuropharmacol. 2013;36(1):24-26.
3. Rami A, Ausmeir F, Winckler J, et al. Differential effects of scopolamine on neuronal survival in ischemia and glutamate neurotoxicity: relationships to the excessive vulnerability of the dorsoseptal hippocampus. J Chem Neuroanat. 1997;13(3):201-208.
4. Benveniste M, Wilhelm J, Dingledine RJ, et al. Subunit-dependent modulation of kainate receptors by muscarinic acetylcholine receptors. Brain Res. 2010;1352:61-69.
5. Duman RS, Li N, Liu RJ, et al. Signaling pathways underlying the rapid antidepressant actions of ketamine. Neuropharmacology. 2012;62(1):35-41.
6. Voleti B, Navarria A, Liu R, et al. Scopolamine rapidly increases mammalian target of rapamycin complex 1 signaling, synaptogenesis, and antidepressant behavioral responses. Biol Psychiatry. 2013;74(10):742-749.
7. Overstreet DH, Friedman E, Mathé AA, et al. The Flinders Sensitive Line rat: a selectively bred putative animal model of depression. Neurosci Biobehav Rev. 2005;29(4-5):739-759.
8. Tizabi Y, Getachew B, Rezvani AH, et al. Antidepressant-like effects of nicotine and reduced nicotinic receptor binding in the Fawn-Hooded rat, an animal model of co-morbid depression and alcoholism. Prog Neuropsychopharmacol Biol Psychiatry. 2009;33(3):398-402.
9. Wang DW, Zhou RB, Yao YM. Role of cholinergic anti-inflammatory pathway in regulating host response and its interventional strategy for inflammatory diseases. Chin J Traumatol. 2009;12(6):355-364.
10. Janowsky DS, el-Yousef MK, Davis JM, et al. A cholinergic-adrenergic hypothesis of mania and depression. Lancet. 1972;2(7778):632-635.
11. Risch SC, Kalin NH, Janowsky DS. Cholinergic challenges in affective illness: behavioral and neuroendocrine correlates. J Clin Psychopharmacol. 1981;1(4):186-192.
12. Risch SC, Janowsky DS, Gillin JC. Muscarinic supersensitivity of anterior pituitary ACTH and β-endorphin release in major depressive illness. Peptides. 1983;4(5):789-792.
13. Risch SC, Janowsky DS, Mott MA, et al. Central and peripheral cholinesterase inhibition: effects on anterior pituitary and sympathomimetic function. Psychoneuroendocrinology. 1986;11(2):221-230.
14. Janowsky DS, Risch SC, Kennedy B, et al. Central muscarinic effects of physostigmine on mood, cardiovascular function, pituitary and adrenal neuroendocrine release. Psychopharmacology (Berl). 1986;89(2):150-154.
15. Furey ML, Drevets WC. Antidepressant efficacy of the antimuscarinic drug scopolamine: a randomized, placebo-controlled trial. Arch Gen Psychiatry. 2006;63(10):1121-1129.
16. Drevets WC, Furey ML. Replication of scopolamine’s antidepressant efficacy in major depressive disorder: a randomized, placebo-controlled clinical trial. Biol Psychiatry. 2010;67(5):432-438.
17. Ellis JS, Zarate CA Jr, Luckenbaugh DA, et al. Antidepressant treatment history as a predictor of response to scopolamine: clinical implications. J Affect Disord. 2014;162:39-42.
18. Newport DJ, Carpenter LL, McDonald WM, et al. Ketamine and other NMDA antagonists: early clinical trials and possible mechanisms in depression. Am J Psychiatry. 2015;172(10):950-966.
19. Bolwig TG. How does electroconvulsive therapy work? Theories on its mechanism. Can J Psychiatry. 2011;56(1):13-18.
20. Wu JC, Kelsoe JR, Schachat C, et al. Rapid and sustained antidepressant response with sleep deprivation and chronotherapy in bipolar disorder. Biol Psychiatry. 2009;66(3):298-301.

References


1. Drevets WC, Zarate CA Jr, Furey ML. Antidepressant effects of the muscarinic cholinergic receptor antagonist scopolamine: a review. Biol Psychiatry. 2013;73(12):1156-1163.
2. Jaffe RJ, Novakovic V, Peselow ED. Scopolamine as an antidepressant: a systematic review. Clin Neuropharmacol. 2013;36(1):24-26.
3. Rami A, Ausmeir F, Winckler J, et al. Differential effects of scopolamine on neuronal survival in ischemia and glutamate neurotoxicity: relationships to the excessive vulnerability of the dorsoseptal hippocampus. J Chem Neuroanat. 1997;13(3):201-208.
4. Benveniste M, Wilhelm J, Dingledine RJ, et al. Subunit-dependent modulation of kainate receptors by muscarinic acetylcholine receptors. Brain Res. 2010;1352:61-69.
5. Duman RS, Li N, Liu RJ, et al. Signaling pathways underlying the rapid antidepressant actions of ketamine. Neuropharmacology. 2012;62(1):35-41.
6. Voleti B, Navarria A, Liu R, et al. Scopolamine rapidly increases mammalian target of rapamycin complex 1 signaling, synaptogenesis, and antidepressant behavioral responses. Biol Psychiatry. 2013;74(10):742-749.
7. Overstreet DH, Friedman E, Mathé AA, et al. The Flinders Sensitive Line rat: a selectively bred putative animal model of depression. Neurosci Biobehav Rev. 2005;29(4-5):739-759.
8. Tizabi Y, Getachew B, Rezvani AH, et al. Antidepressant-like effects of nicotine and reduced nicotinic receptor binding in the Fawn-Hooded rat, an animal model of co-morbid depression and alcoholism. Prog Neuropsychopharmacol Biol Psychiatry. 2009;33(3):398-402.
9. Wang DW, Zhou RB, Yao YM. Role of cholinergic anti-inflammatory pathway in regulating host response and its interventional strategy for inflammatory diseases. Chin J Traumatol. 2009;12(6):355-364.
10. Janowsky DS, el-Yousef MK, Davis JM, et al. A cholinergic-adrenergic hypothesis of mania and depression. Lancet. 1972;2(7778):632-635.
11. Risch SC, Kalin NH, Janowsky DS. Cholinergic challenges in affective illness: behavioral and neuroendocrine correlates. J Clin Psychopharmacol. 1981;1(4):186-192.
12. Risch SC, Janowsky DS, Gillin JC. Muscarinic supersensitivity of anterior pituitary ACTH and β-endorphin release in major depressive illness. Peptides. 1983;4(5):789-792.
13. Risch SC, Janowsky DS, Mott MA, et al. Central and peripheral cholinesterase inhibition: effects on anterior pituitary and sympathomimetic function. Psychoneuroendocrinology. 1986;11(2):221-230.
14. Janowsky DS, Risch SC, Kennedy B, et al. Central muscarinic effects of physostigmine on mood, cardiovascular function, pituitary and adrenal neuroendocrine release. Psychopharmacology (Berl). 1986;89(2):150-154.
15. Furey ML, Drevets WC. Antidepressant efficacy of the antimuscarinic drug scopolamine: a randomized, placebo-controlled trial. Arch Gen Psychiatry. 2006;63(10):1121-1129.
16. Drevets WC, Furey ML. Replication of scopolamine’s antidepressant efficacy in major depressive disorder: a randomized, placebo-controlled clinical trial. Biol Psychiatry. 2010;67(5):432-438.
17. Ellis JS, Zarate CA Jr, Luckenbaugh DA, et al. Antidepressant treatment history as a predictor of response to scopolamine: clinical implications. J Affect Disord. 2014;162:39-42.
18. Newport DJ, Carpenter LL, McDonald WM, et al. Ketamine and other NMDA antagonists: early clinical trials and possible mechanisms in depression. Am J Psychiatry. 2015;172(10):950-966.
19. Bolwig TG. How does electroconvulsive therapy work? Theories on its mechanism. Can J Psychiatry. 2011;56(1):13-18.
20. Wu JC, Kelsoe JR, Schachat C, et al. Rapid and sustained antidepressant response with sleep deprivation and chronotherapy in bipolar disorder. Biol Psychiatry. 2009;66(3):298-301.

Issue
Current Psychiatry - 15(4)
Issue
Current Psychiatry - 15(4)
Page Number
45-49
Page Number
45-49
Publications
Publications
Topics
Article Type
Display Headline
Manic after taking a vacation
Display Headline
Manic after taking a vacation
Legacy Keywords
psychosis, psychotic episode, motion-sickness patch, scopolamine, dimenhydrinate, psychotic disorder, psychotic disorders, mania, manic, euphoric, racing thoughts, lithium
Legacy Keywords
psychosis, psychotic episode, motion-sickness patch, scopolamine, dimenhydrinate, psychotic disorder, psychotic disorders, mania, manic, euphoric, racing thoughts, lithium
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Be an activist to prevent edentulism among the mentally ill

Article Type
Changed
Tue, 12/11/2018 - 15:17
Display Headline
Be an activist to prevent edentulism among the mentally ill

Poor dental hygiene is a serious and prevalent problem among people with mental illness or cognitive impairment: Dental caries and periodontal disease are 3.4 times more common among the mentally ill than among the general population.1 Little has been published on the causes and prevention of these diseases among the mentally ill, however. Interprofessional education provides the opportunity to reinforce the connection between oral health and systemic health.

Untreated dental disease can result in edentulism (partial or complete tooth loss). Often, this condition leads to embarrassment, poor self-image, and social isolation—all of which can exacerbate the psychotic state and its symptoms. Working with your patient to improve oral health can, in turn, lead to better mental and physical health.


CASE REPORT
Edentulism in a man with schizophrenia
A 34-year-old man, given a diagnosis of schizophrenia at age 17, is admitted to the inpatient psychiatry unit for bizarre behavior. The next day, 4 maxillary and incisor teeth fall out suddenly while he is brushing his teeth. The patient is brought to emergency dental services.

Factors contributing to his tooth loss include:

  • schizophrenia
  • neglected oral hygiene
  • adverse effects of antipsychotic medication
  • lack of advice on the importance of oral hygiene
  • failure to recognize signs of a dental problem.


What else can lead to edentulism?

Breakdown of the periodontal attachment2 also can be caused by disinterest in oral hygiene practices; craving of, and preference for, carbohydrates because of reduced central serotonin activity3,4; and xerostomia.

Xerostomia, or dry mouth, caused by psychotropic agents and an altered immune response, facilitates growth of pathogenic bacteria and can lead to several dental diseases (Table). These conditions are exacerbated by consumption of chewing gum, sweets, and sugary drinks in response to constantly feeling thirsty from xerostomia. Advise patients to take frequent sips of fluid or let ice cubes melt in their mouth.

Bruxism. Patients taking a selective serotonin reuptake inhibitor or an atypical antipsychotic can develop a movement disorder (eg, extrapyramidal symptoms or tardive dyskinesia) that includes clenching, grinding of the teeth (bruxism), or both, which can worsen their periodontal condition.

Lack of skills, physical dexterity, and motivation to maintain good oral hygiene are common among people with mental illness. Most patients visit a dentist only when they experience a serious oral problem or an emergency (ie, trauma). Many dentists treat psychiatric patients by extracting the tooth that is causing the pain, instead of pursuing complex tooth preservation or restoration techniques because of (1) the extent of the disease, (2) lack of knowledge related to psychiatric illnesses, and (3) frequent and timely follow-ups.5

Providing education about oral health to patients, implementing preventive steps, and educating other medical specialities about the link between oral health and systemic health can help to reduce the burden of dental problems among mentally ill patients.


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

References


1. Persson K, Axtelius B, Söderfeldt B, et al. Oral health-related quality of life and dental status in an outpatient psychiatric population: a multivariate approach. Int J Ment Health Nurs. 2010;19(1):62-70.
2. Lalloo R, Kisely S, Amarasinghe H, et al. Oral health of patients on psychotropic medications: a study of outpatients in Queensland. Australas Psychiatry. 2013;21(4):338-342.
3. O’Neil A, Berk M, Venugopal K, et al. The association between poor dental health and depression: findings from a large-scale, population-based study (the NHANES study). Gen Hosp Psychiatry. 2014;36(3):266-270.
4. Kisely S, Quek LH, Paris J, et al. Advanced dental disease in people with severe mental illness: systematic review and meta-analysis. Br J Psychiatry. 2011;199(3):187-193.
5. Arnaiz A, Zumárraga M, Díez-Altuna I, et al. Oral health and the symptoms of schizophrenia. Psychiatry Res. 2011;188(1):24-28.

Article PDF
Author and Disclosure Information

Mahreen Raza, MD
Fellow

Ketan Hirapara, MBBS
Research Volunteer

Najeeb Hussain, MD
Assistant Professor

Department of Psychiatry
Rutgers New Jersey Medical School
Newark, New Jersey

Issue
Current Psychiatry - 15(4)
Publications
Topics
Page Number
56-57
Legacy Keywords
tooth loss, losing teeth, edentulism, oral hygiene, schizophrenia, psychosis, psychotic disorder, psychotic disorders, lost teeth, oral health, dental, dentist, dental hygiene, caries, dry mouth, xerostomnia, bruxism, bacteria, dental disease
Sections
Author and Disclosure Information

Mahreen Raza, MD
Fellow

Ketan Hirapara, MBBS
Research Volunteer

Najeeb Hussain, MD
Assistant Professor

Department of Psychiatry
Rutgers New Jersey Medical School
Newark, New Jersey

Author and Disclosure Information

Mahreen Raza, MD
Fellow

Ketan Hirapara, MBBS
Research Volunteer

Najeeb Hussain, MD
Assistant Professor

Department of Psychiatry
Rutgers New Jersey Medical School
Newark, New Jersey

Article PDF
Article PDF

Poor dental hygiene is a serious and prevalent problem among people with mental illness or cognitive impairment: Dental caries and periodontal disease are 3.4 times more common among the mentally ill than among the general population.1 Little has been published on the causes and prevention of these diseases among the mentally ill, however. Interprofessional education provides the opportunity to reinforce the connection between oral health and systemic health.

Untreated dental disease can result in edentulism (partial or complete tooth loss). Often, this condition leads to embarrassment, poor self-image, and social isolation—all of which can exacerbate the psychotic state and its symptoms. Working with your patient to improve oral health can, in turn, lead to better mental and physical health.


CASE REPORT
Edentulism in a man with schizophrenia
A 34-year-old man, given a diagnosis of schizophrenia at age 17, is admitted to the inpatient psychiatry unit for bizarre behavior. The next day, 4 maxillary and incisor teeth fall out suddenly while he is brushing his teeth. The patient is brought to emergency dental services.

Factors contributing to his tooth loss include:

  • schizophrenia
  • neglected oral hygiene
  • adverse effects of antipsychotic medication
  • lack of advice on the importance of oral hygiene
  • failure to recognize signs of a dental problem.


What else can lead to edentulism?

Breakdown of the periodontal attachment2 also can be caused by disinterest in oral hygiene practices; craving of, and preference for, carbohydrates because of reduced central serotonin activity3,4; and xerostomia.

Xerostomia, or dry mouth, caused by psychotropic agents and an altered immune response, facilitates growth of pathogenic bacteria and can lead to several dental diseases (Table). These conditions are exacerbated by consumption of chewing gum, sweets, and sugary drinks in response to constantly feeling thirsty from xerostomia. Advise patients to take frequent sips of fluid or let ice cubes melt in their mouth.

Bruxism. Patients taking a selective serotonin reuptake inhibitor or an atypical antipsychotic can develop a movement disorder (eg, extrapyramidal symptoms or tardive dyskinesia) that includes clenching, grinding of the teeth (bruxism), or both, which can worsen their periodontal condition.

Lack of skills, physical dexterity, and motivation to maintain good oral hygiene are common among people with mental illness. Most patients visit a dentist only when they experience a serious oral problem or an emergency (ie, trauma). Many dentists treat psychiatric patients by extracting the tooth that is causing the pain, instead of pursuing complex tooth preservation or restoration techniques because of (1) the extent of the disease, (2) lack of knowledge related to psychiatric illnesses, and (3) frequent and timely follow-ups.5

Providing education about oral health to patients, implementing preventive steps, and educating other medical specialities about the link between oral health and systemic health can help to reduce the burden of dental problems among mentally ill patients.


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

Poor dental hygiene is a serious and prevalent problem among people with mental illness or cognitive impairment: Dental caries and periodontal disease are 3.4 times more common among the mentally ill than among the general population.1 Little has been published on the causes and prevention of these diseases among the mentally ill, however. Interprofessional education provides the opportunity to reinforce the connection between oral health and systemic health.

Untreated dental disease can result in edentulism (partial or complete tooth loss). Often, this condition leads to embarrassment, poor self-image, and social isolation—all of which can exacerbate the psychotic state and its symptoms. Working with your patient to improve oral health can, in turn, lead to better mental and physical health.


CASE REPORT
Edentulism in a man with schizophrenia
A 34-year-old man, given a diagnosis of schizophrenia at age 17, is admitted to the inpatient psychiatry unit for bizarre behavior. The next day, 4 maxillary and incisor teeth fall out suddenly while he is brushing his teeth. The patient is brought to emergency dental services.

Factors contributing to his tooth loss include:

  • schizophrenia
  • neglected oral hygiene
  • adverse effects of antipsychotic medication
  • lack of advice on the importance of oral hygiene
  • failure to recognize signs of a dental problem.


What else can lead to edentulism?

Breakdown of the periodontal attachment2 also can be caused by disinterest in oral hygiene practices; craving of, and preference for, carbohydrates because of reduced central serotonin activity3,4; and xerostomia.

Xerostomia, or dry mouth, caused by psychotropic agents and an altered immune response, facilitates growth of pathogenic bacteria and can lead to several dental diseases (Table). These conditions are exacerbated by consumption of chewing gum, sweets, and sugary drinks in response to constantly feeling thirsty from xerostomia. Advise patients to take frequent sips of fluid or let ice cubes melt in their mouth.

Bruxism. Patients taking a selective serotonin reuptake inhibitor or an atypical antipsychotic can develop a movement disorder (eg, extrapyramidal symptoms or tardive dyskinesia) that includes clenching, grinding of the teeth (bruxism), or both, which can worsen their periodontal condition.

Lack of skills, physical dexterity, and motivation to maintain good oral hygiene are common among people with mental illness. Most patients visit a dentist only when they experience a serious oral problem or an emergency (ie, trauma). Many dentists treat psychiatric patients by extracting the tooth that is causing the pain, instead of pursuing complex tooth preservation or restoration techniques because of (1) the extent of the disease, (2) lack of knowledge related to psychiatric illnesses, and (3) frequent and timely follow-ups.5

Providing education about oral health to patients, implementing preventive steps, and educating other medical specialities about the link between oral health and systemic health can help to reduce the burden of dental problems among mentally ill patients.


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

References


1. Persson K, Axtelius B, Söderfeldt B, et al. Oral health-related quality of life and dental status in an outpatient psychiatric population: a multivariate approach. Int J Ment Health Nurs. 2010;19(1):62-70.
2. Lalloo R, Kisely S, Amarasinghe H, et al. Oral health of patients on psychotropic medications: a study of outpatients in Queensland. Australas Psychiatry. 2013;21(4):338-342.
3. O’Neil A, Berk M, Venugopal K, et al. The association between poor dental health and depression: findings from a large-scale, population-based study (the NHANES study). Gen Hosp Psychiatry. 2014;36(3):266-270.
4. Kisely S, Quek LH, Paris J, et al. Advanced dental disease in people with severe mental illness: systematic review and meta-analysis. Br J Psychiatry. 2011;199(3):187-193.
5. Arnaiz A, Zumárraga M, Díez-Altuna I, et al. Oral health and the symptoms of schizophrenia. Psychiatry Res. 2011;188(1):24-28.

References


1. Persson K, Axtelius B, Söderfeldt B, et al. Oral health-related quality of life and dental status in an outpatient psychiatric population: a multivariate approach. Int J Ment Health Nurs. 2010;19(1):62-70.
2. Lalloo R, Kisely S, Amarasinghe H, et al. Oral health of patients on psychotropic medications: a study of outpatients in Queensland. Australas Psychiatry. 2013;21(4):338-342.
3. O’Neil A, Berk M, Venugopal K, et al. The association between poor dental health and depression: findings from a large-scale, population-based study (the NHANES study). Gen Hosp Psychiatry. 2014;36(3):266-270.
4. Kisely S, Quek LH, Paris J, et al. Advanced dental disease in people with severe mental illness: systematic review and meta-analysis. Br J Psychiatry. 2011;199(3):187-193.
5. Arnaiz A, Zumárraga M, Díez-Altuna I, et al. Oral health and the symptoms of schizophrenia. Psychiatry Res. 2011;188(1):24-28.

Issue
Current Psychiatry - 15(4)
Issue
Current Psychiatry - 15(4)
Page Number
56-57
Page Number
56-57
Publications
Publications
Topics
Article Type
Display Headline
Be an activist to prevent edentulism among the mentally ill
Display Headline
Be an activist to prevent edentulism among the mentally ill
Legacy Keywords
tooth loss, losing teeth, edentulism, oral hygiene, schizophrenia, psychosis, psychotic disorder, psychotic disorders, lost teeth, oral health, dental, dentist, dental hygiene, caries, dry mouth, xerostomnia, bruxism, bacteria, dental disease
Legacy Keywords
tooth loss, losing teeth, edentulism, oral hygiene, schizophrenia, psychosis, psychotic disorder, psychotic disorders, lost teeth, oral health, dental, dentist, dental hygiene, caries, dry mouth, xerostomnia, bruxism, bacteria, dental disease
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Technology offers tools for ensuring adherence to medical therapy

Article Type
Changed
Thu, 03/28/2019 - 15:10
Display Headline
Technology offers tools for ensuring adherence to medical therapy

Nonadherence to medical therapy is a widespread and complex problem that is a significant variable in the treatment of psychiatric illness and in patients’ prognosis. More than 50% of people who have a chronic illness struggle to comply with their medication regimen—for many reasons.1

Many variables predict poor adherence, so it cannot be expected that a single solution will solve the problem entirely.2 Novel adherence technologies are available, as we discuss in this article, and more are in development.


What is nonadherence to medical therapy?

Nonadherence can be defined primarily as not taking prescribed medication in the recommended dosage or frequency, or not taking prescribed medication at all.3 Nonadherence can result in an increased risk of relapse, hospitalization, poor therapeutic response, and delayed remission and recovery.

Secondarily, non-attendance or irregular attendance at appointments with providers is a form of nonadherence that can have a negative impact on treatment outcomes.4


Why is medical adherence important in psychiatry?

Medication nonadherence has major consequences for psychiatric patients5 and for the greater health care system; it is estimated that, in the United States, the cost of nonadherence is as high as $300 billion a year.6 In psychiatry, the rate of nonadherence to medical therapy has been reported to be 11% to 80% of patients with schizophrenia; 12% to 64% with bipolar disorders; and 30% to 60% with depression.7-9 These surprising statistics make it imperative to design treatment strategies that include an effective patient-centric medication adherence plan, based on diagnosis, patient need, education, and support.


Why are patients nonadherent?

Many variables lead to patient nonadherence (Figure 1). The most common reason is that patients simply forget to take their medication.10 Among psychiatric patients, other reasons are:

  • lack of insight
  • negative emotional reaction to taking medication11
  • feeling better and no longer believing that the medication is needed12,13
  • distress associated with side effects14,15
  • high cost of medication15
  • patient’s perception that medication won’t be effective16,17
  • concern about substance abuse18
  • fear of dependency19
  • complicated dosing regimen20
  • general lack of motivation.21

Emotional barriers to medication nonadherence are an underestimated area that can benefit greatly from the expertise and understanding of psychiatrists. These barriers include a sense of losing control, self-stigmatization, denial, poor insight, and beliefs about illness and medications.

Additional patient variables that contribute to nonadherence include:

  • suboptimal health literacy
  • stigma and shame about the need for psychiatric treatment
  • lack of patient involvement in treatment decision-making.


Who is responsible for adherence?

Adherence to medical therapy is not the patient’s responsibility, exclusively. Rather, it is a collection of complex components that generally includes physicians and the health care system. Because barriers to medication adherence are complex and varied, solutions to improve adherence must be multifaceted.

Providers. Patients’ care often is managed by multiple physicians, which can lead to communication lapses about complicated drug regimens and potential adverse effects. To assist patients in adhering to their medication regimen, physicians should recognize, and acknowledge to the patient, that many psychiatric patients have difficulty taking their medications and provide advice and information in how to address this problem.

Families. Likewise, it is important to educate patients and their family about the need for medication—helping the patient see that it is his (her) choice and, indeed, his direct responsibility to take his medication and improve his health. The risk–benefit balance of treatment should be explained to the patient and his family, as well as the nature of the psychiatric diagnosis and how effective patient–physician collaboration can help him function and adhere to his medication regimen in a consistent, reliable manner.

The larger system. Health care systems can contribute to medication adherence by reducing time constraints on visits to providers, to allow time to discuss all aspects of medication adherence. Limited visits in the clinic means physicians are not able to (1) spend adequate time discussing the medication regimen to ensure full patient comprehension and (2) conduct an assessment of medication-taking behaviors. Team-based approaches could improve efficiency, patient understanding, adherence, and early detection of adherence issues.22,23

Strategies such as additional clinic visits and reminder calls to discuss adherence carry a cost, but their long-term advantage is that, if patients understand how to better adhere to their medication regimens, their actions will have a positive impact on their health care costs and outcomes and on the wider health economy—as a result of reduced hospital admissions and reduced need to care for patients whose condition deteriorates because of nonadherence. It is imperative that we build strong relationships with other providers to show that we are committed to building supportive, effective adherence support programs that focus on the individual patient’s needs.

 

 


What is the available technology?

There is no standard way to measure non­adherence. The most common, and simplest, measure—asking the patient—is unreliable and severely overestimates adherence.

Direct measures of adherence include observing the patient taking his medications and testing for the concentration of those medications in blood or urine. Indirect adherence assessment methods, such as pill counts, a medication diary, self-report, clinician ratings, pharmacy chart review, and electronic devices that monitor the opening of a lid or tablet strip, have all been used; yet reviews of those methods have shown less than favorable results.6

Pre-packaged pill packs have helped some patients with a simple method for medication management.

Electronic monitoring, using a medication vial cap device (Figure 2) that electronically records the date and time of bottle opening, has become common in general medicine and among patients with schizophrenia.6,13,24-26 Diaz et al24 reported that electronic monitoring detected a greater nonadherence rate (57%) than what prescribers reported (7%) or patients self-reported (5%)—demonstrating that prescribers and patients grossly overestimate adherence. In another study that looked at electronic monitoring, researchers reported that adherence was much higher in depressed youth (87%)27 than what had been seen in adults (67%) in a similar study.13

The downside to pill packs and electronic monitoring? There is no guarantee the patient has actually taken the medication despite the data reported by the system.

Event marker-signaling devices. Novel technologies have been developed to measure adherence:

Proteus Digital Health feedback system (www.proteus.com) requires that patients ingest a tablet containing a tiny, dietary mineral-based “ingestible event marker.” Upon contact with gastric fluid electrolytes, the event marker emits a unique signal that is transmitted through bodily tissue to a small receiver in a patch worn on the torso. The receiver then transmits a signal to a cellular phone, indicating the time and date when the medication was ingested (Figure 3).

A 4-week pilot study28 found that the ingestible event marker is feasible and acceptable to patients: 27 of 28 participants (96%) completed the study, with a mean adherence rate of 74%. Although the system identifies ingestible sensors with high accuracy and is easily tolerated by patients, the pilot study was brief; a longer duration of adherence while wearing the patch needs to be studied.

Breath analysis, facial recognition. Even directly observing ingestion of a medication can be problematic: Some patients don’t swallow the medication and spit it out later. One way around that subterfuge is to consider using other advanced medication adherence solutions that are breath-based or use facial recognition technology and confirm ingestion.

Xhale SMART (www.xhale.com/smart) is a handheld device that generates a reminder to the patient to take his medication; afterward, he (she) must blow into the device so that ingestion of the medication is detected (Figure 4). The medication has breath-detectable adherence markers already incorporated. The adherence marker then is released into the stomach and small intestine, where the adherence marker metabolite is transported through the bloodstream into the lungs and exhaled. The patient must breathe into a breath analysis device, which measures medication ingestion compared with a baseline breath print.

Several articles in the literature have reported the accuracy of this device in detecting the ingested metabolite in every participant, without adverse effects.29,30 Clinical data on the use of the breath-based detector is not available to the public at this time.

AiCure (www.aicure.com) is a facial recognition-based technology platform that can work through any smartphone. The device is powered by artificial intelligence software and motion-sensing technology that can detect, in real time, whether the patient is taking the medication as prescribed. Patients who take an incorrect dose, or who do not use the software, are automatically flagged for immediate follow-up. This technology enables real-time intervention by a provider with the nonadherent patient.

An important note: These innovative technological advances are tools that can help clinicians manage an important aspect of treatment, but they do not show the entire picture: The physician−patient relationship and the therapeutic alliance are key to optimal treatment adherence.


Engage and empower the patient

Novel adherence technologies are, as we’ve described, available, and more are being developed. Incorporating these technologies into clinical care requires continued input and support from clinicians and patients. Digital and mobile health applications are multi-beneficial: They can empower patients to self-manage medication regimens and appointments while they also receive social and psychological information and support as needed. Understanding one’s own illness can, ultimately, improve outcomes and significantly reduce health care costs.

Patient empowerment is key. The physician is an important influencer in this regard.

 

 

The role of the physician must not be undervalued in maintaining adherence to therapy; she (he) plays a vital role in continued patient engagement and behavioral training. Integrating physician-led oversight, patient education, and commitment, and novel digital mobile adherence technologies will help deliver better outcomes.

The push to engage. A “one size fits all” approach to maintaining adherence won’t be effective. We need to better understand the individual patient’s underlying cause(s) for nonadherence, then to tailor a solution to influence and change that behavior. One way to do this is by interacting and engaging more directly (and in a digital manner) with patients to monitor adherence.

A recent example of the move toward direct patient engagement is the agreement entered by Otsuka Pharmaceuticals and Proteus Digital Health to develop novel digital health products. The FDA has accepted for review the combination product of Otsuka’s brand of aripiprazole and Proteus’s ingestible sensor. If the product is approved by the FDA, physicians will be able to prescribe aripiprazole with the ingestible sensor embedded in the tablet and then measure medication adherence and other patient physiologic metrics (eg, activity, rest) through the wearable sensor patch and medical software application designed specifically for patient and physician use.

This technology could have huge potential in mental health care, where patients struggle with both adhering to their medication regimen and communicating with the health care team. Physicians could measure adherence when treating adults with schizophrenia, bipolar disorders, and major depressive disorder; flag those who are not adhering as having higher risk of disease progression and poorer outcome; and allow decisions to be made more quickly based on treatment need.

Developing and enhancing these collaborative and patient-centric approaches will increase self-monitoring and patient responsibility, and encourage behavior change.

‘All-in’ strategy. By continuing to use the latest technologies and connecting them to the range of stakeholders—physicians, nurses, pharmacists, payers—we will develop an all-inclusive adherence intervention strategy. All patients will be integrated, and all of them, and their family, will be provided with positive psychoeducational care and motivational counseling (Figure 5). In addition, such a support-based patient experience must be aligned with the work of clinical care providers. Compliance therapy and behavioral training, together with active patient engagement, can help improve insight, acceptance of treatment, and, over the long term, adherence.31,32


Bottom Line

Nonadherence to medical therapy is a large and detrimental phenomenon across medicine, but especially among people suffering from a brain disorder. Strategies that include a plan to improve adherence to medical therapy through behavioral and educational techniques and innovative technology might contribute to improved adherence. All stakeholders must be involved in this effort, not just patients.


Related Resources

  • Pitschel-Walz G, Bäuml J, Bender W, et al. Psychoeducation and compliance in the treatment of schizophrenia: results of the Munich Psychosis Information Project Study. 2006;67(3):443-452.
  • Sajatovic M, Levin JB, Sams J, et al. Symptom severity, self-reported adherence, and electronic pill monitoring in poorly adherent patients with bipolar disorder. Bipolar Disord. 2015;17(6):653-661.

Drug Brand Name
Aripiprazole • Abilify

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


1. World Health Organization. Adherence to long-term therapies: evidence for action. Geneva, Switzerland: World Health Organization; 2003.
2. Bosworth HB, Granger BB, Mendys P, et al. Medication adherence: a call for action. Am Heart J. 2011;162(3):412-424.
3. Crowe M, Wilson L, Inder M. Patients’ reports of the factors influencing medication adherence in bipolar disorder – an integrative review of the literature. Int J Nurs Stud. 2011;48(7):894-903.
4. Mert D, Turgut NH, Kelleci M, et al. Perspectives on reasons of medication nonadherence in psychiatric patients. Patient Prefer Adherence. 2015;9:87-93.
5. Chapman SC, Horne R. Medication nonadherence and psychiatry. Curr Opin Psychiatry. 2013;26(5):446-452.
6. Osterberg L, Blaschke T. Adherence to medication. N Engl J Med. 2005;353(5):487-497.
7. Thompson L, McCabe R. The effect of clinician-patient alliance and communication on treatment adherence in mental health care: a systematic review. BMC Psychiatry. 2012;12:87.
8. Yilmaz S, Buzlu S. Antipsikotik kullanan hastalarda ilaç yan etkileri ve ilaç uyumu. Florence Nightingale Hem˘girelik Dergisi. 2012;20(2):93-103.
9. Kelleci M, Ata EE. Psikiyatri Klini˘ginde yatan hastaların ilaç uyumları ve sosyal destekle iliskisi. [Drug compliance of patients hospitalized in the psychiatry clinic and the relationship with social support]. Psikiyatri Hemsireli˘gi Dergisi. 2011;2(suppl 3):105-110.
10. Bulloch AG, Patten SB. Non-adherence with psychotropic medications in the general population. Soc Psychiatry Psychiatr Epidemiol. 2010;45(1):47-56.
11. Rosenbaum L. Beyond belief—how people feel about taking medications for heart disease. N Engl J Med. 2015;372(2):183-187.
12. Cramer J, Rosenheck R, Kirk G, et al. Medication compliance feedback and monitoring in a clinical trial: predictions and outcomes. Value Health. 2003;6(5):566-573.
13. Nakonezny PA, Byerly MJ, Rush AJ. Electronic monitoring of antipsychotic medication adherence in outpatients with schizophrenia or schizoaffective disorder: an empirical evaluation of its reliability and predictive validity. Psychiatry Res. 2008;157(1-3):259-263.
14. Fortney JC, Pyne JM, Edlund MJ, et al. Reasons for antidepressant nonadherence among veterans treated in primary care clinics. J Clin Psychiatry. 2011;72(6):827-834.
15. Kennedy J, Tuleu I, Mackay K. Unfilled prescriptions of medicare beneficiaries: prevalence, reasons, and types of medicines prescribed. J Manag Care Pharm. 2008;14(6):553-560.
16. Hoencamp E, Stevens A, Haffmans J. Patients’ attitudes toward antidepressants. Psychiatr Serv. 2002;53(9):1180-1181.
17. Keller MB, Hirschfeld RM, Demyttenaere K, et al. Optimizing outcomes in depression: focus on antidepressant compliance. Int Clin Psychopharmacol. 2002;17(6):265-271.
18. Akerblad AC, Bengtsson F, Holgersson M, et al. Identification of primary care patients at risk of nonadherence to antidepressant treatment. Patient Prefer Adherence. 2008;2:376-386.
19. Brown C, Battista DR, Bruehlman R, et al. Beliefs about antidepressant medications in primary care patients: relationship to self-reported adherence. Med Care. 2005;43(12):1203-1207.
20. Demyttenaere K, Adelin A, Patrick M, et al. Six-month compliance with antidepressant medication in the treatment of major depressive disorder. Int Clin Psychopharmacol. 2008;23(1):36-42.
21. Massand PS. Tolerability and adherence issues in antidepressant therapy. Clin Ther. 2003;25(8):2289-2304.
22. Medicare Prescription Drug, Improvement, and Modernization Act of 2003. Pub L No. 108-173, 117 Stat 2066.
23. Brown MT, Bussell JK. Medication adherence: WHO cares? Mayo Clin Proc. 2011;86(4):304-314.
24. Diaz E, Neuse E, Sullivan MC, et al. Adherence to conventional and atypical antipsychotics after hospital discharge. J Clin Psychiatry. 2004;65(3):354-360.
25. Byerly M, Fisher R, Whatley K, et al. A comparison of electronic monitoring vs. clinician rating of antipsychotic adherence in outpatients with schizophrenia. Psychiatry Res. 2005;133(2-3):129-133.
26. Byerly MJ, Nakonezny PA, Lescouflair E. Antipsychotic medication adherence in schizophrenia. Psychiatr Clin North Am. 2007;30(3):437-452.
27. Nakonezny PA, Hughes CW, Mayes TL, et al. A comparison of various methods of measuring antidepressant medication adherence among children and adolescents with major depressive disorder in a 12-week open trial of fluoxetine. J Child Adolesc Psychopharmacol. 2010;20(5):431-439.
28. Kane JM, Perlis RH, DiCarlo LA, et al. First experience with a wireless system incorporating physiologic assessments and direct confirmation of digital tablet ingestions in ambulatory patients with schizophrenia or bipolar disorder. J Clin Psychiatry. 2013;74(6):e533-e540. doi: 10.4088/JCP.12m08222.
29. Morey TE, Booth MM, Prather RA, et al. Measurement of ethanol in gaseous breath using a miniature gas chromatograph. J Anal Toxicol. 2011;35(3):134-142.
30. Morey TE, Booth M, Wasdo S, et al. Oral adherence monitoring using a breath test to supplement highly active antiretroviral therapy. AIDS Behav. 2013;17(1):298-306.
31. Torem MS. Participatory pharmacotherapy: 10 strategies for enhancing adherence. Current Psychiatry. 2013;12(7):21-25.
32. Zygmunt A, Olfson M, Boyer CA, et al. Interventions to improve medication adherence in schizophrenia. Am J Psychiatry. 2002;159(10):1653-1664.

Article PDF
Author and Disclosure Information

Amir Kalali, MD
Head
Neuroscience Center of Excellence
Quintiles Inc.
San Diego, California
Member, Current Psychiatry Editorial Board

Sarah Richerson, PhD
Deputy Head
Neuroscience Center of Excellence

John Reites
Head
Digital Health Acceleration

Quintiles Inc.
San Diego, California

Issue
Current Psychiatry - 15(4)
Publications
Topics
Page Number
24-26,32-34,36
Legacy Keywords
technology, practice trends, medication adherence, nonadherence, pill packs, vial cap device, event marker-signaling devices, Proteus Digital Health feedback system, Xhale SMART Medication adherence monitoring system, electronic monitoring, event marker, facial recognition, breath analysis, AiCure, help patients adhere to medication
Sections
Author and Disclosure Information

Amir Kalali, MD
Head
Neuroscience Center of Excellence
Quintiles Inc.
San Diego, California
Member, Current Psychiatry Editorial Board

Sarah Richerson, PhD
Deputy Head
Neuroscience Center of Excellence

John Reites
Head
Digital Health Acceleration

Quintiles Inc.
San Diego, California

Author and Disclosure Information

Amir Kalali, MD
Head
Neuroscience Center of Excellence
Quintiles Inc.
San Diego, California
Member, Current Psychiatry Editorial Board

Sarah Richerson, PhD
Deputy Head
Neuroscience Center of Excellence

John Reites
Head
Digital Health Acceleration

Quintiles Inc.
San Diego, California

Article PDF
Article PDF

Nonadherence to medical therapy is a widespread and complex problem that is a significant variable in the treatment of psychiatric illness and in patients’ prognosis. More than 50% of people who have a chronic illness struggle to comply with their medication regimen—for many reasons.1

Many variables predict poor adherence, so it cannot be expected that a single solution will solve the problem entirely.2 Novel adherence technologies are available, as we discuss in this article, and more are in development.


What is nonadherence to medical therapy?

Nonadherence can be defined primarily as not taking prescribed medication in the recommended dosage or frequency, or not taking prescribed medication at all.3 Nonadherence can result in an increased risk of relapse, hospitalization, poor therapeutic response, and delayed remission and recovery.

Secondarily, non-attendance or irregular attendance at appointments with providers is a form of nonadherence that can have a negative impact on treatment outcomes.4


Why is medical adherence important in psychiatry?

Medication nonadherence has major consequences for psychiatric patients5 and for the greater health care system; it is estimated that, in the United States, the cost of nonadherence is as high as $300 billion a year.6 In psychiatry, the rate of nonadherence to medical therapy has been reported to be 11% to 80% of patients with schizophrenia; 12% to 64% with bipolar disorders; and 30% to 60% with depression.7-9 These surprising statistics make it imperative to design treatment strategies that include an effective patient-centric medication adherence plan, based on diagnosis, patient need, education, and support.


Why are patients nonadherent?

Many variables lead to patient nonadherence (Figure 1). The most common reason is that patients simply forget to take their medication.10 Among psychiatric patients, other reasons are:

  • lack of insight
  • negative emotional reaction to taking medication11
  • feeling better and no longer believing that the medication is needed12,13
  • distress associated with side effects14,15
  • high cost of medication15
  • patient’s perception that medication won’t be effective16,17
  • concern about substance abuse18
  • fear of dependency19
  • complicated dosing regimen20
  • general lack of motivation.21

Emotional barriers to medication nonadherence are an underestimated area that can benefit greatly from the expertise and understanding of psychiatrists. These barriers include a sense of losing control, self-stigmatization, denial, poor insight, and beliefs about illness and medications.

Additional patient variables that contribute to nonadherence include:

  • suboptimal health literacy
  • stigma and shame about the need for psychiatric treatment
  • lack of patient involvement in treatment decision-making.


Who is responsible for adherence?

Adherence to medical therapy is not the patient’s responsibility, exclusively. Rather, it is a collection of complex components that generally includes physicians and the health care system. Because barriers to medication adherence are complex and varied, solutions to improve adherence must be multifaceted.

Providers. Patients’ care often is managed by multiple physicians, which can lead to communication lapses about complicated drug regimens and potential adverse effects. To assist patients in adhering to their medication regimen, physicians should recognize, and acknowledge to the patient, that many psychiatric patients have difficulty taking their medications and provide advice and information in how to address this problem.

Families. Likewise, it is important to educate patients and their family about the need for medication—helping the patient see that it is his (her) choice and, indeed, his direct responsibility to take his medication and improve his health. The risk–benefit balance of treatment should be explained to the patient and his family, as well as the nature of the psychiatric diagnosis and how effective patient–physician collaboration can help him function and adhere to his medication regimen in a consistent, reliable manner.

The larger system. Health care systems can contribute to medication adherence by reducing time constraints on visits to providers, to allow time to discuss all aspects of medication adherence. Limited visits in the clinic means physicians are not able to (1) spend adequate time discussing the medication regimen to ensure full patient comprehension and (2) conduct an assessment of medication-taking behaviors. Team-based approaches could improve efficiency, patient understanding, adherence, and early detection of adherence issues.22,23

Strategies such as additional clinic visits and reminder calls to discuss adherence carry a cost, but their long-term advantage is that, if patients understand how to better adhere to their medication regimens, their actions will have a positive impact on their health care costs and outcomes and on the wider health economy—as a result of reduced hospital admissions and reduced need to care for patients whose condition deteriorates because of nonadherence. It is imperative that we build strong relationships with other providers to show that we are committed to building supportive, effective adherence support programs that focus on the individual patient’s needs.

 

 


What is the available technology?

There is no standard way to measure non­adherence. The most common, and simplest, measure—asking the patient—is unreliable and severely overestimates adherence.

Direct measures of adherence include observing the patient taking his medications and testing for the concentration of those medications in blood or urine. Indirect adherence assessment methods, such as pill counts, a medication diary, self-report, clinician ratings, pharmacy chart review, and electronic devices that monitor the opening of a lid or tablet strip, have all been used; yet reviews of those methods have shown less than favorable results.6

Pre-packaged pill packs have helped some patients with a simple method for medication management.

Electronic monitoring, using a medication vial cap device (Figure 2) that electronically records the date and time of bottle opening, has become common in general medicine and among patients with schizophrenia.6,13,24-26 Diaz et al24 reported that electronic monitoring detected a greater nonadherence rate (57%) than what prescribers reported (7%) or patients self-reported (5%)—demonstrating that prescribers and patients grossly overestimate adherence. In another study that looked at electronic monitoring, researchers reported that adherence was much higher in depressed youth (87%)27 than what had been seen in adults (67%) in a similar study.13

The downside to pill packs and electronic monitoring? There is no guarantee the patient has actually taken the medication despite the data reported by the system.

Event marker-signaling devices. Novel technologies have been developed to measure adherence:

Proteus Digital Health feedback system (www.proteus.com) requires that patients ingest a tablet containing a tiny, dietary mineral-based “ingestible event marker.” Upon contact with gastric fluid electrolytes, the event marker emits a unique signal that is transmitted through bodily tissue to a small receiver in a patch worn on the torso. The receiver then transmits a signal to a cellular phone, indicating the time and date when the medication was ingested (Figure 3).

A 4-week pilot study28 found that the ingestible event marker is feasible and acceptable to patients: 27 of 28 participants (96%) completed the study, with a mean adherence rate of 74%. Although the system identifies ingestible sensors with high accuracy and is easily tolerated by patients, the pilot study was brief; a longer duration of adherence while wearing the patch needs to be studied.

Breath analysis, facial recognition. Even directly observing ingestion of a medication can be problematic: Some patients don’t swallow the medication and spit it out later. One way around that subterfuge is to consider using other advanced medication adherence solutions that are breath-based or use facial recognition technology and confirm ingestion.

Xhale SMART (www.xhale.com/smart) is a handheld device that generates a reminder to the patient to take his medication; afterward, he (she) must blow into the device so that ingestion of the medication is detected (Figure 4). The medication has breath-detectable adherence markers already incorporated. The adherence marker then is released into the stomach and small intestine, where the adherence marker metabolite is transported through the bloodstream into the lungs and exhaled. The patient must breathe into a breath analysis device, which measures medication ingestion compared with a baseline breath print.

Several articles in the literature have reported the accuracy of this device in detecting the ingested metabolite in every participant, without adverse effects.29,30 Clinical data on the use of the breath-based detector is not available to the public at this time.

AiCure (www.aicure.com) is a facial recognition-based technology platform that can work through any smartphone. The device is powered by artificial intelligence software and motion-sensing technology that can detect, in real time, whether the patient is taking the medication as prescribed. Patients who take an incorrect dose, or who do not use the software, are automatically flagged for immediate follow-up. This technology enables real-time intervention by a provider with the nonadherent patient.

An important note: These innovative technological advances are tools that can help clinicians manage an important aspect of treatment, but they do not show the entire picture: The physician−patient relationship and the therapeutic alliance are key to optimal treatment adherence.


Engage and empower the patient

Novel adherence technologies are, as we’ve described, available, and more are being developed. Incorporating these technologies into clinical care requires continued input and support from clinicians and patients. Digital and mobile health applications are multi-beneficial: They can empower patients to self-manage medication regimens and appointments while they also receive social and psychological information and support as needed. Understanding one’s own illness can, ultimately, improve outcomes and significantly reduce health care costs.

Patient empowerment is key. The physician is an important influencer in this regard.

 

 

The role of the physician must not be undervalued in maintaining adherence to therapy; she (he) plays a vital role in continued patient engagement and behavioral training. Integrating physician-led oversight, patient education, and commitment, and novel digital mobile adherence technologies will help deliver better outcomes.

The push to engage. A “one size fits all” approach to maintaining adherence won’t be effective. We need to better understand the individual patient’s underlying cause(s) for nonadherence, then to tailor a solution to influence and change that behavior. One way to do this is by interacting and engaging more directly (and in a digital manner) with patients to monitor adherence.

A recent example of the move toward direct patient engagement is the agreement entered by Otsuka Pharmaceuticals and Proteus Digital Health to develop novel digital health products. The FDA has accepted for review the combination product of Otsuka’s brand of aripiprazole and Proteus’s ingestible sensor. If the product is approved by the FDA, physicians will be able to prescribe aripiprazole with the ingestible sensor embedded in the tablet and then measure medication adherence and other patient physiologic metrics (eg, activity, rest) through the wearable sensor patch and medical software application designed specifically for patient and physician use.

This technology could have huge potential in mental health care, where patients struggle with both adhering to their medication regimen and communicating with the health care team. Physicians could measure adherence when treating adults with schizophrenia, bipolar disorders, and major depressive disorder; flag those who are not adhering as having higher risk of disease progression and poorer outcome; and allow decisions to be made more quickly based on treatment need.

Developing and enhancing these collaborative and patient-centric approaches will increase self-monitoring and patient responsibility, and encourage behavior change.

‘All-in’ strategy. By continuing to use the latest technologies and connecting them to the range of stakeholders—physicians, nurses, pharmacists, payers—we will develop an all-inclusive adherence intervention strategy. All patients will be integrated, and all of them, and their family, will be provided with positive psychoeducational care and motivational counseling (Figure 5). In addition, such a support-based patient experience must be aligned with the work of clinical care providers. Compliance therapy and behavioral training, together with active patient engagement, can help improve insight, acceptance of treatment, and, over the long term, adherence.31,32


Bottom Line

Nonadherence to medical therapy is a large and detrimental phenomenon across medicine, but especially among people suffering from a brain disorder. Strategies that include a plan to improve adherence to medical therapy through behavioral and educational techniques and innovative technology might contribute to improved adherence. All stakeholders must be involved in this effort, not just patients.


Related Resources

  • Pitschel-Walz G, Bäuml J, Bender W, et al. Psychoeducation and compliance in the treatment of schizophrenia: results of the Munich Psychosis Information Project Study. 2006;67(3):443-452.
  • Sajatovic M, Levin JB, Sams J, et al. Symptom severity, self-reported adherence, and electronic pill monitoring in poorly adherent patients with bipolar disorder. Bipolar Disord. 2015;17(6):653-661.

Drug Brand Name
Aripiprazole • Abilify

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

Nonadherence to medical therapy is a widespread and complex problem that is a significant variable in the treatment of psychiatric illness and in patients’ prognosis. More than 50% of people who have a chronic illness struggle to comply with their medication regimen—for many reasons.1

Many variables predict poor adherence, so it cannot be expected that a single solution will solve the problem entirely.2 Novel adherence technologies are available, as we discuss in this article, and more are in development.


What is nonadherence to medical therapy?

Nonadherence can be defined primarily as not taking prescribed medication in the recommended dosage or frequency, or not taking prescribed medication at all.3 Nonadherence can result in an increased risk of relapse, hospitalization, poor therapeutic response, and delayed remission and recovery.

Secondarily, non-attendance or irregular attendance at appointments with providers is a form of nonadherence that can have a negative impact on treatment outcomes.4


Why is medical adherence important in psychiatry?

Medication nonadherence has major consequences for psychiatric patients5 and for the greater health care system; it is estimated that, in the United States, the cost of nonadherence is as high as $300 billion a year.6 In psychiatry, the rate of nonadherence to medical therapy has been reported to be 11% to 80% of patients with schizophrenia; 12% to 64% with bipolar disorders; and 30% to 60% with depression.7-9 These surprising statistics make it imperative to design treatment strategies that include an effective patient-centric medication adherence plan, based on diagnosis, patient need, education, and support.


Why are patients nonadherent?

Many variables lead to patient nonadherence (Figure 1). The most common reason is that patients simply forget to take their medication.10 Among psychiatric patients, other reasons are:

  • lack of insight
  • negative emotional reaction to taking medication11
  • feeling better and no longer believing that the medication is needed12,13
  • distress associated with side effects14,15
  • high cost of medication15
  • patient’s perception that medication won’t be effective16,17
  • concern about substance abuse18
  • fear of dependency19
  • complicated dosing regimen20
  • general lack of motivation.21

Emotional barriers to medication nonadherence are an underestimated area that can benefit greatly from the expertise and understanding of psychiatrists. These barriers include a sense of losing control, self-stigmatization, denial, poor insight, and beliefs about illness and medications.

Additional patient variables that contribute to nonadherence include:

  • suboptimal health literacy
  • stigma and shame about the need for psychiatric treatment
  • lack of patient involvement in treatment decision-making.


Who is responsible for adherence?

Adherence to medical therapy is not the patient’s responsibility, exclusively. Rather, it is a collection of complex components that generally includes physicians and the health care system. Because barriers to medication adherence are complex and varied, solutions to improve adherence must be multifaceted.

Providers. Patients’ care often is managed by multiple physicians, which can lead to communication lapses about complicated drug regimens and potential adverse effects. To assist patients in adhering to their medication regimen, physicians should recognize, and acknowledge to the patient, that many psychiatric patients have difficulty taking their medications and provide advice and information in how to address this problem.

Families. Likewise, it is important to educate patients and their family about the need for medication—helping the patient see that it is his (her) choice and, indeed, his direct responsibility to take his medication and improve his health. The risk–benefit balance of treatment should be explained to the patient and his family, as well as the nature of the psychiatric diagnosis and how effective patient–physician collaboration can help him function and adhere to his medication regimen in a consistent, reliable manner.

The larger system. Health care systems can contribute to medication adherence by reducing time constraints on visits to providers, to allow time to discuss all aspects of medication adherence. Limited visits in the clinic means physicians are not able to (1) spend adequate time discussing the medication regimen to ensure full patient comprehension and (2) conduct an assessment of medication-taking behaviors. Team-based approaches could improve efficiency, patient understanding, adherence, and early detection of adherence issues.22,23

Strategies such as additional clinic visits and reminder calls to discuss adherence carry a cost, but their long-term advantage is that, if patients understand how to better adhere to their medication regimens, their actions will have a positive impact on their health care costs and outcomes and on the wider health economy—as a result of reduced hospital admissions and reduced need to care for patients whose condition deteriorates because of nonadherence. It is imperative that we build strong relationships with other providers to show that we are committed to building supportive, effective adherence support programs that focus on the individual patient’s needs.

 

 


What is the available technology?

There is no standard way to measure non­adherence. The most common, and simplest, measure—asking the patient—is unreliable and severely overestimates adherence.

Direct measures of adherence include observing the patient taking his medications and testing for the concentration of those medications in blood or urine. Indirect adherence assessment methods, such as pill counts, a medication diary, self-report, clinician ratings, pharmacy chart review, and electronic devices that monitor the opening of a lid or tablet strip, have all been used; yet reviews of those methods have shown less than favorable results.6

Pre-packaged pill packs have helped some patients with a simple method for medication management.

Electronic monitoring, using a medication vial cap device (Figure 2) that electronically records the date and time of bottle opening, has become common in general medicine and among patients with schizophrenia.6,13,24-26 Diaz et al24 reported that electronic monitoring detected a greater nonadherence rate (57%) than what prescribers reported (7%) or patients self-reported (5%)—demonstrating that prescribers and patients grossly overestimate adherence. In another study that looked at electronic monitoring, researchers reported that adherence was much higher in depressed youth (87%)27 than what had been seen in adults (67%) in a similar study.13

The downside to pill packs and electronic monitoring? There is no guarantee the patient has actually taken the medication despite the data reported by the system.

Event marker-signaling devices. Novel technologies have been developed to measure adherence:

Proteus Digital Health feedback system (www.proteus.com) requires that patients ingest a tablet containing a tiny, dietary mineral-based “ingestible event marker.” Upon contact with gastric fluid electrolytes, the event marker emits a unique signal that is transmitted through bodily tissue to a small receiver in a patch worn on the torso. The receiver then transmits a signal to a cellular phone, indicating the time and date when the medication was ingested (Figure 3).

A 4-week pilot study28 found that the ingestible event marker is feasible and acceptable to patients: 27 of 28 participants (96%) completed the study, with a mean adherence rate of 74%. Although the system identifies ingestible sensors with high accuracy and is easily tolerated by patients, the pilot study was brief; a longer duration of adherence while wearing the patch needs to be studied.

Breath analysis, facial recognition. Even directly observing ingestion of a medication can be problematic: Some patients don’t swallow the medication and spit it out later. One way around that subterfuge is to consider using other advanced medication adherence solutions that are breath-based or use facial recognition technology and confirm ingestion.

Xhale SMART (www.xhale.com/smart) is a handheld device that generates a reminder to the patient to take his medication; afterward, he (she) must blow into the device so that ingestion of the medication is detected (Figure 4). The medication has breath-detectable adherence markers already incorporated. The adherence marker then is released into the stomach and small intestine, where the adherence marker metabolite is transported through the bloodstream into the lungs and exhaled. The patient must breathe into a breath analysis device, which measures medication ingestion compared with a baseline breath print.

Several articles in the literature have reported the accuracy of this device in detecting the ingested metabolite in every participant, without adverse effects.29,30 Clinical data on the use of the breath-based detector is not available to the public at this time.

AiCure (www.aicure.com) is a facial recognition-based technology platform that can work through any smartphone. The device is powered by artificial intelligence software and motion-sensing technology that can detect, in real time, whether the patient is taking the medication as prescribed. Patients who take an incorrect dose, or who do not use the software, are automatically flagged for immediate follow-up. This technology enables real-time intervention by a provider with the nonadherent patient.

An important note: These innovative technological advances are tools that can help clinicians manage an important aspect of treatment, but they do not show the entire picture: The physician−patient relationship and the therapeutic alliance are key to optimal treatment adherence.


Engage and empower the patient

Novel adherence technologies are, as we’ve described, available, and more are being developed. Incorporating these technologies into clinical care requires continued input and support from clinicians and patients. Digital and mobile health applications are multi-beneficial: They can empower patients to self-manage medication regimens and appointments while they also receive social and psychological information and support as needed. Understanding one’s own illness can, ultimately, improve outcomes and significantly reduce health care costs.

Patient empowerment is key. The physician is an important influencer in this regard.

 

 

The role of the physician must not be undervalued in maintaining adherence to therapy; she (he) plays a vital role in continued patient engagement and behavioral training. Integrating physician-led oversight, patient education, and commitment, and novel digital mobile adherence technologies will help deliver better outcomes.

The push to engage. A “one size fits all” approach to maintaining adherence won’t be effective. We need to better understand the individual patient’s underlying cause(s) for nonadherence, then to tailor a solution to influence and change that behavior. One way to do this is by interacting and engaging more directly (and in a digital manner) with patients to monitor adherence.

A recent example of the move toward direct patient engagement is the agreement entered by Otsuka Pharmaceuticals and Proteus Digital Health to develop novel digital health products. The FDA has accepted for review the combination product of Otsuka’s brand of aripiprazole and Proteus’s ingestible sensor. If the product is approved by the FDA, physicians will be able to prescribe aripiprazole with the ingestible sensor embedded in the tablet and then measure medication adherence and other patient physiologic metrics (eg, activity, rest) through the wearable sensor patch and medical software application designed specifically for patient and physician use.

This technology could have huge potential in mental health care, where patients struggle with both adhering to their medication regimen and communicating with the health care team. Physicians could measure adherence when treating adults with schizophrenia, bipolar disorders, and major depressive disorder; flag those who are not adhering as having higher risk of disease progression and poorer outcome; and allow decisions to be made more quickly based on treatment need.

Developing and enhancing these collaborative and patient-centric approaches will increase self-monitoring and patient responsibility, and encourage behavior change.

‘All-in’ strategy. By continuing to use the latest technologies and connecting them to the range of stakeholders—physicians, nurses, pharmacists, payers—we will develop an all-inclusive adherence intervention strategy. All patients will be integrated, and all of them, and their family, will be provided with positive psychoeducational care and motivational counseling (Figure 5). In addition, such a support-based patient experience must be aligned with the work of clinical care providers. Compliance therapy and behavioral training, together with active patient engagement, can help improve insight, acceptance of treatment, and, over the long term, adherence.31,32


Bottom Line

Nonadherence to medical therapy is a large and detrimental phenomenon across medicine, but especially among people suffering from a brain disorder. Strategies that include a plan to improve adherence to medical therapy through behavioral and educational techniques and innovative technology might contribute to improved adherence. All stakeholders must be involved in this effort, not just patients.


Related Resources

  • Pitschel-Walz G, Bäuml J, Bender W, et al. Psychoeducation and compliance in the treatment of schizophrenia: results of the Munich Psychosis Information Project Study. 2006;67(3):443-452.
  • Sajatovic M, Levin JB, Sams J, et al. Symptom severity, self-reported adherence, and electronic pill monitoring in poorly adherent patients with bipolar disorder. Bipolar Disord. 2015;17(6):653-661.

Drug Brand Name
Aripiprazole • Abilify

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


1. World Health Organization. Adherence to long-term therapies: evidence for action. Geneva, Switzerland: World Health Organization; 2003.
2. Bosworth HB, Granger BB, Mendys P, et al. Medication adherence: a call for action. Am Heart J. 2011;162(3):412-424.
3. Crowe M, Wilson L, Inder M. Patients’ reports of the factors influencing medication adherence in bipolar disorder – an integrative review of the literature. Int J Nurs Stud. 2011;48(7):894-903.
4. Mert D, Turgut NH, Kelleci M, et al. Perspectives on reasons of medication nonadherence in psychiatric patients. Patient Prefer Adherence. 2015;9:87-93.
5. Chapman SC, Horne R. Medication nonadherence and psychiatry. Curr Opin Psychiatry. 2013;26(5):446-452.
6. Osterberg L, Blaschke T. Adherence to medication. N Engl J Med. 2005;353(5):487-497.
7. Thompson L, McCabe R. The effect of clinician-patient alliance and communication on treatment adherence in mental health care: a systematic review. BMC Psychiatry. 2012;12:87.
8. Yilmaz S, Buzlu S. Antipsikotik kullanan hastalarda ilaç yan etkileri ve ilaç uyumu. Florence Nightingale Hem˘girelik Dergisi. 2012;20(2):93-103.
9. Kelleci M, Ata EE. Psikiyatri Klini˘ginde yatan hastaların ilaç uyumları ve sosyal destekle iliskisi. [Drug compliance of patients hospitalized in the psychiatry clinic and the relationship with social support]. Psikiyatri Hemsireli˘gi Dergisi. 2011;2(suppl 3):105-110.
10. Bulloch AG, Patten SB. Non-adherence with psychotropic medications in the general population. Soc Psychiatry Psychiatr Epidemiol. 2010;45(1):47-56.
11. Rosenbaum L. Beyond belief—how people feel about taking medications for heart disease. N Engl J Med. 2015;372(2):183-187.
12. Cramer J, Rosenheck R, Kirk G, et al. Medication compliance feedback and monitoring in a clinical trial: predictions and outcomes. Value Health. 2003;6(5):566-573.
13. Nakonezny PA, Byerly MJ, Rush AJ. Electronic monitoring of antipsychotic medication adherence in outpatients with schizophrenia or schizoaffective disorder: an empirical evaluation of its reliability and predictive validity. Psychiatry Res. 2008;157(1-3):259-263.
14. Fortney JC, Pyne JM, Edlund MJ, et al. Reasons for antidepressant nonadherence among veterans treated in primary care clinics. J Clin Psychiatry. 2011;72(6):827-834.
15. Kennedy J, Tuleu I, Mackay K. Unfilled prescriptions of medicare beneficiaries: prevalence, reasons, and types of medicines prescribed. J Manag Care Pharm. 2008;14(6):553-560.
16. Hoencamp E, Stevens A, Haffmans J. Patients’ attitudes toward antidepressants. Psychiatr Serv. 2002;53(9):1180-1181.
17. Keller MB, Hirschfeld RM, Demyttenaere K, et al. Optimizing outcomes in depression: focus on antidepressant compliance. Int Clin Psychopharmacol. 2002;17(6):265-271.
18. Akerblad AC, Bengtsson F, Holgersson M, et al. Identification of primary care patients at risk of nonadherence to antidepressant treatment. Patient Prefer Adherence. 2008;2:376-386.
19. Brown C, Battista DR, Bruehlman R, et al. Beliefs about antidepressant medications in primary care patients: relationship to self-reported adherence. Med Care. 2005;43(12):1203-1207.
20. Demyttenaere K, Adelin A, Patrick M, et al. Six-month compliance with antidepressant medication in the treatment of major depressive disorder. Int Clin Psychopharmacol. 2008;23(1):36-42.
21. Massand PS. Tolerability and adherence issues in antidepressant therapy. Clin Ther. 2003;25(8):2289-2304.
22. Medicare Prescription Drug, Improvement, and Modernization Act of 2003. Pub L No. 108-173, 117 Stat 2066.
23. Brown MT, Bussell JK. Medication adherence: WHO cares? Mayo Clin Proc. 2011;86(4):304-314.
24. Diaz E, Neuse E, Sullivan MC, et al. Adherence to conventional and atypical antipsychotics after hospital discharge. J Clin Psychiatry. 2004;65(3):354-360.
25. Byerly M, Fisher R, Whatley K, et al. A comparison of electronic monitoring vs. clinician rating of antipsychotic adherence in outpatients with schizophrenia. Psychiatry Res. 2005;133(2-3):129-133.
26. Byerly MJ, Nakonezny PA, Lescouflair E. Antipsychotic medication adherence in schizophrenia. Psychiatr Clin North Am. 2007;30(3):437-452.
27. Nakonezny PA, Hughes CW, Mayes TL, et al. A comparison of various methods of measuring antidepressant medication adherence among children and adolescents with major depressive disorder in a 12-week open trial of fluoxetine. J Child Adolesc Psychopharmacol. 2010;20(5):431-439.
28. Kane JM, Perlis RH, DiCarlo LA, et al. First experience with a wireless system incorporating physiologic assessments and direct confirmation of digital tablet ingestions in ambulatory patients with schizophrenia or bipolar disorder. J Clin Psychiatry. 2013;74(6):e533-e540. doi: 10.4088/JCP.12m08222.
29. Morey TE, Booth MM, Prather RA, et al. Measurement of ethanol in gaseous breath using a miniature gas chromatograph. J Anal Toxicol. 2011;35(3):134-142.
30. Morey TE, Booth M, Wasdo S, et al. Oral adherence monitoring using a breath test to supplement highly active antiretroviral therapy. AIDS Behav. 2013;17(1):298-306.
31. Torem MS. Participatory pharmacotherapy: 10 strategies for enhancing adherence. Current Psychiatry. 2013;12(7):21-25.
32. Zygmunt A, Olfson M, Boyer CA, et al. Interventions to improve medication adherence in schizophrenia. Am J Psychiatry. 2002;159(10):1653-1664.

References


1. World Health Organization. Adherence to long-term therapies: evidence for action. Geneva, Switzerland: World Health Organization; 2003.
2. Bosworth HB, Granger BB, Mendys P, et al. Medication adherence: a call for action. Am Heart J. 2011;162(3):412-424.
3. Crowe M, Wilson L, Inder M. Patients’ reports of the factors influencing medication adherence in bipolar disorder – an integrative review of the literature. Int J Nurs Stud. 2011;48(7):894-903.
4. Mert D, Turgut NH, Kelleci M, et al. Perspectives on reasons of medication nonadherence in psychiatric patients. Patient Prefer Adherence. 2015;9:87-93.
5. Chapman SC, Horne R. Medication nonadherence and psychiatry. Curr Opin Psychiatry. 2013;26(5):446-452.
6. Osterberg L, Blaschke T. Adherence to medication. N Engl J Med. 2005;353(5):487-497.
7. Thompson L, McCabe R. The effect of clinician-patient alliance and communication on treatment adherence in mental health care: a systematic review. BMC Psychiatry. 2012;12:87.
8. Yilmaz S, Buzlu S. Antipsikotik kullanan hastalarda ilaç yan etkileri ve ilaç uyumu. Florence Nightingale Hem˘girelik Dergisi. 2012;20(2):93-103.
9. Kelleci M, Ata EE. Psikiyatri Klini˘ginde yatan hastaların ilaç uyumları ve sosyal destekle iliskisi. [Drug compliance of patients hospitalized in the psychiatry clinic and the relationship with social support]. Psikiyatri Hemsireli˘gi Dergisi. 2011;2(suppl 3):105-110.
10. Bulloch AG, Patten SB. Non-adherence with psychotropic medications in the general population. Soc Psychiatry Psychiatr Epidemiol. 2010;45(1):47-56.
11. Rosenbaum L. Beyond belief—how people feel about taking medications for heart disease. N Engl J Med. 2015;372(2):183-187.
12. Cramer J, Rosenheck R, Kirk G, et al. Medication compliance feedback and monitoring in a clinical trial: predictions and outcomes. Value Health. 2003;6(5):566-573.
13. Nakonezny PA, Byerly MJ, Rush AJ. Electronic monitoring of antipsychotic medication adherence in outpatients with schizophrenia or schizoaffective disorder: an empirical evaluation of its reliability and predictive validity. Psychiatry Res. 2008;157(1-3):259-263.
14. Fortney JC, Pyne JM, Edlund MJ, et al. Reasons for antidepressant nonadherence among veterans treated in primary care clinics. J Clin Psychiatry. 2011;72(6):827-834.
15. Kennedy J, Tuleu I, Mackay K. Unfilled prescriptions of medicare beneficiaries: prevalence, reasons, and types of medicines prescribed. J Manag Care Pharm. 2008;14(6):553-560.
16. Hoencamp E, Stevens A, Haffmans J. Patients’ attitudes toward antidepressants. Psychiatr Serv. 2002;53(9):1180-1181.
17. Keller MB, Hirschfeld RM, Demyttenaere K, et al. Optimizing outcomes in depression: focus on antidepressant compliance. Int Clin Psychopharmacol. 2002;17(6):265-271.
18. Akerblad AC, Bengtsson F, Holgersson M, et al. Identification of primary care patients at risk of nonadherence to antidepressant treatment. Patient Prefer Adherence. 2008;2:376-386.
19. Brown C, Battista DR, Bruehlman R, et al. Beliefs about antidepressant medications in primary care patients: relationship to self-reported adherence. Med Care. 2005;43(12):1203-1207.
20. Demyttenaere K, Adelin A, Patrick M, et al. Six-month compliance with antidepressant medication in the treatment of major depressive disorder. Int Clin Psychopharmacol. 2008;23(1):36-42.
21. Massand PS. Tolerability and adherence issues in antidepressant therapy. Clin Ther. 2003;25(8):2289-2304.
22. Medicare Prescription Drug, Improvement, and Modernization Act of 2003. Pub L No. 108-173, 117 Stat 2066.
23. Brown MT, Bussell JK. Medication adherence: WHO cares? Mayo Clin Proc. 2011;86(4):304-314.
24. Diaz E, Neuse E, Sullivan MC, et al. Adherence to conventional and atypical antipsychotics after hospital discharge. J Clin Psychiatry. 2004;65(3):354-360.
25. Byerly M, Fisher R, Whatley K, et al. A comparison of electronic monitoring vs. clinician rating of antipsychotic adherence in outpatients with schizophrenia. Psychiatry Res. 2005;133(2-3):129-133.
26. Byerly MJ, Nakonezny PA, Lescouflair E. Antipsychotic medication adherence in schizophrenia. Psychiatr Clin North Am. 2007;30(3):437-452.
27. Nakonezny PA, Hughes CW, Mayes TL, et al. A comparison of various methods of measuring antidepressant medication adherence among children and adolescents with major depressive disorder in a 12-week open trial of fluoxetine. J Child Adolesc Psychopharmacol. 2010;20(5):431-439.
28. Kane JM, Perlis RH, DiCarlo LA, et al. First experience with a wireless system incorporating physiologic assessments and direct confirmation of digital tablet ingestions in ambulatory patients with schizophrenia or bipolar disorder. J Clin Psychiatry. 2013;74(6):e533-e540. doi: 10.4088/JCP.12m08222.
29. Morey TE, Booth MM, Prather RA, et al. Measurement of ethanol in gaseous breath using a miniature gas chromatograph. J Anal Toxicol. 2011;35(3):134-142.
30. Morey TE, Booth M, Wasdo S, et al. Oral adherence monitoring using a breath test to supplement highly active antiretroviral therapy. AIDS Behav. 2013;17(1):298-306.
31. Torem MS. Participatory pharmacotherapy: 10 strategies for enhancing adherence. Current Psychiatry. 2013;12(7):21-25.
32. Zygmunt A, Olfson M, Boyer CA, et al. Interventions to improve medication adherence in schizophrenia. Am J Psychiatry. 2002;159(10):1653-1664.

Issue
Current Psychiatry - 15(4)
Issue
Current Psychiatry - 15(4)
Page Number
24-26,32-34,36
Page Number
24-26,32-34,36
Publications
Publications
Topics
Article Type
Display Headline
Technology offers tools for ensuring adherence to medical therapy
Display Headline
Technology offers tools for ensuring adherence to medical therapy
Legacy Keywords
technology, practice trends, medication adherence, nonadherence, pill packs, vial cap device, event marker-signaling devices, Proteus Digital Health feedback system, Xhale SMART Medication adherence monitoring system, electronic monitoring, event marker, facial recognition, breath analysis, AiCure, help patients adhere to medication
Legacy Keywords
technology, practice trends, medication adherence, nonadherence, pill packs, vial cap device, event marker-signaling devices, Proteus Digital Health feedback system, Xhale SMART Medication adherence monitoring system, electronic monitoring, event marker, facial recognition, breath analysis, AiCure, help patients adhere to medication
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media